Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
Biomolecules ; 14(6)2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38927058

ABSTRACT

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.


Subject(s)
Retina , Voltage-Dependent Anion Channels , Humans , Voltage-Dependent Anion Channels/metabolism , Retina/metabolism , Animals , Oxidative Stress , Retinal Diseases/metabolism , Retinal Diseases/pathology , Mitochondria/metabolism , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/pathology
3.
Transl Neurodegener ; 11(1): 58, 2022 12 28.
Article in English | MEDLINE | ID: mdl-36578022

ABSTRACT

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.


Subject(s)
Alzheimer Disease , Mice , Animals , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Mitochondrial Proteins , Amyloid beta-Peptides/metabolism , Brain/metabolism , Mitochondria/metabolism
4.
J Neurochem ; 158(5): 1131-1150, 2021 09.
Article in English | MEDLINE | ID: mdl-34265077

ABSTRACT

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.


Subject(s)
Chemokine CCL2/antagonists & inhibitors , Lycium , MAP Kinase Signaling System/drug effects , Palmitates/therapeutic use , Retinal Degeneration/prevention & control , Retinitis Pigmentosa/prevention & control , STAT3 Transcription Factor/antagonists & inhibitors , Xanthophylls/therapeutic use , Animals , Chemokine CCL2/metabolism , Female , MAP Kinase Signaling System/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Palmitates/isolation & purification , Palmitates/pharmacology , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/pathology , STAT3 Transcription Factor/metabolism , Xanthophylls/isolation & purification , Xanthophylls/pharmacology
5.
Sci Rep ; 11(1): 9376, 2021 04 30.
Article in English | MEDLINE | ID: mdl-33931669

ABSTRACT

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.


Subject(s)
Aminoacyltransferases/physiology , Fibroblasts/metabolism , Light , Nervous System/metabolism , RGS Proteins/metabolism , Retinal Bipolar Cells/metabolism , Animals , Female , Fibroblasts/pathology , Fibroblasts/radiation effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nervous System/pathology , Nervous System/radiation effects , RGS Proteins/genetics , Retinal Bipolar Cells/pathology , Retinal Bipolar Cells/radiation effects , Signal Transduction
6.
Neuropharmacology ; 139: 137-149, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29940208

ABSTRACT

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.


Subject(s)
6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Neuroprotective Agents/pharmacology , Receptors, Ionotropic Glutamate/antagonists & inhibitors , Retinal Degeneration/prevention & control , Action Potentials/drug effects , Animals , Disease Models, Animal , Female , Male , Mice, Inbred C57BL , Mice, Transgenic , Photoreceptor Cells, Vertebrate/drug effects , Photoreceptor Cells, Vertebrate/pathology , Retinal Bipolar Cells/drug effects , Retinal Bipolar Cells/physiology , Retinal Degeneration/pathology , Retinal Degeneration/physiopathology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Retinal Ganglion Cells/physiology , Tissue Culture Techniques , Vision, Ocular/drug effects
7.
Invest Ophthalmol Vis Sci ; 59(1): 597-611, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29372259

ABSTRACT

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.


Subject(s)
Disease Models, Animal , Drugs, Chinese Herbal/therapeutic use , Photoreceptor Cells, Vertebrate/drug effects , Retinitis Pigmentosa/prevention & control , Animals , Animals, Newborn , Behavior, Animal/physiology , Electroretinography , Injections, Intraperitoneal , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Photoreceptor Cells, Vertebrate/physiology , Retinitis Pigmentosa/physiopathology , Vision, Ocular/physiology
8.
Invest Ophthalmol Vis Sci ; 57(6): 2788-96, 2016 05 01.
Article in English | MEDLINE | ID: mdl-27214687

ABSTRACT

PURPOSE: Frizzled3 (Fzd3), a member of the core planar cell polarity (PCP) family in mammals, contributes to visual development by guiding axonal projections of some retinal ganglion cells. However, its other functions in the maturation of the visual system, especially the retina, remain elusive. The present study explores the role of Fzd3 in retinal development by focusing on rod bipolar cells (RBCs). METHODS: Frizzled3 was conditionally removed from the retina of Isl1-Cre;Fzd3f/- mice using the Cre-loxP system. Electroretinograms (ERGs) were performed to measure the light response of retinas. Frizzled3 expression was monitored by ß-galactosidase (ß-gal) staining and anti-ß-gal immunostaining. Immunofluorescence was used to examine cellular distribution and morphology during development, and electron microscopy was applied to visualize the dendritic invaginations of RBCs. RESULTS: Electroretinograms showed decreased b-wave amplitudes, and lower b- to a-wave ratios in Isl1-Cre;Fzd3f/- than in control (Isl1-Cre;Fzd3f/+) mice. In RBCs, where Fzd3 was expressed and inactivated, the planar organization, shape, and orientation of somas were disrupted. From P10, dendrites of these RBCs displayed reduced arborization with mistargeting. Furthermore, their dendritic invaginations into rod terminals were suppressed, and the density of rod ribbons in the OPL was reduced. CONCLUSIONS: Frizzled3 is required to shape the pattern of RBC somas and dendrites, and the structural and functional connectivity between rods and RBCs. Our results highlight novel functions for Fzd3 in regulating retinal development.


Subject(s)
Frizzled Receptors/genetics , Gene Expression Regulation, Developmental , Retinal Bipolar Cells/physiology , Retinal Ganglion Cells/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Animals , Animals, Newborn , Cell Count , Disease Models, Animal , Electroretinography , Female , Frizzled Receptors/metabolism , Male , Mice , Mice, Mutant Strains , Microscopy, Electron , Retinal Ganglion Cells/ultrastructure , Retinal Rod Photoreceptor Cells/ultrastructure
9.
Eur J Neurosci ; 43(11): 1509-22, 2016 06.
Article in English | MEDLINE | ID: mdl-27037829

ABSTRACT

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.


Subject(s)
Extracellular Matrix Proteins/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Receptors, Metabotropic Glutamate/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Synapses/ultrastructure , Animals , Dendrites/metabolism , Female , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , GTPase-Activating Proteins/metabolism , Male , Mice , Mice, Knockout , Retinal Bipolar Cells/metabolism , Retinal Bipolar Cells/ultrastructure , Retinal Rod Photoreceptor Cells/ultrastructure , Signal Transduction , TRPM Cation Channels/metabolism
10.
Sci Rep ; 6: 20940, 2016 Feb 17.
Article in English | MEDLINE | ID: mdl-26883481

ABSTRACT

Transmission from photoreceptors to ON bipolar cells in mammalian retina is mediated by a sign-inverting cascade. Upon binding glutamate, the metabotropic glutamate receptor mGluR6 activates the heterotrimeric G-protein Gαoß3γ13, and this leads to closure of the TRPM1 channel (melastatin). TRPM1 is thought to be constitutively open, but the mechanism that leads to its closure is unclear. We investigated this question in mouse rod bipolar cells by dialyzing reagents that modify the activity of either Gαo or Gßγ and then observing their effects on the basal holding current. After opening the TRPM1 channels with light, a constitutively active mutant of Gαo closed the channel, but wild-type Gαo did not. After closing the channels by dark adaptation, phosducin or inactive Gαo (both sequester Gßγ) opened the channel while the active mutant of Gαo did not. Co-immunoprecipitation showed that TRPM1 interacts with Gß3 and with the active and inactive forms of Gαo. Furthermore, bioluminescent energy transfer assays indicated that while Gαo interacts with both the N- and the C- termini of TRPM1, Gßγ interacts only with the N-terminus. Our physiological and biochemical results suggest that both Gαo and Gßγ bind TRPM1 channels and cooperate to close them.


Subject(s)
Heterotrimeric GTP-Binding Proteins/metabolism , Ion Channel Gating , Retinal Bipolar Cells/metabolism , TRPM Cation Channels/metabolism , Animals , Cell Line , Dark Adaptation , Darkness , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Heterotrimeric GTP-Binding Proteins/genetics , Humans , Light , Mice , Models, Animal , Mutation , Myristic Acid/pharmacology , Protein Binding , Rats , Retinal Bipolar Cells/radiation effects
11.
J Physiol ; 593(7): 1531-50, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25416620

ABSTRACT

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.


Subject(s)
Heterotrimeric GTP-Binding Proteins/physiology , Receptors, Metabotropic Glutamate/physiology , Retinal Bipolar Cells/physiology , Animals , Electroretinography , Female , Heterotrimeric GTP-Binding Proteins/genetics , Male , Mice, Inbred C57BL , Mice, Knockout
12.
Front Mol Neurosci ; 7: 60, 2014.
Article in English | MEDLINE | ID: mdl-25018695

ABSTRACT

The ubiquitous second messenger cGMP is synthesized by guanylyl cyclase and hydrolyzed by phosphodiesterase (PDE). cGMP mediates numerous signaling pathways in multiple tissues. In the retina, cGMP regulates signaling in nearly every cell class including photoreceptors, bipolar cells, amacrine cells, and ganglion cells. In order to understand the specific role of cGMP and its regulating enzymes in different cell types, it is first necessary to localize these components and dissect their influence on the circuits. Here we tested the contribution of PDE9A to retinal processing by recording the electroretinograms (ERG) of PDE9A (™/™) (KO) mice and by localizing the enzyme. We found that while the scotopic ERG of KO was the same as that of wild type (WT) in both amplitude and kinetics, the photopic ERG was greatly affected. The greatest effect was on the recovery of the b-wave; the falling phase and the b-wave duration were significantly longer in the KO mice for all photopic stimuli (UV, green, or saturating white flashes). The rising phase was slower in KO than in WT for UV and green stimuli. For certain stimuli, amplitudes of both the a- and b-waves were smaller than in WT. Using Lac-Z expression in KO retinas as a reporter for PDE9A expression pattern, we found that PDE9A is localized to GABA-positive and GABA-negative amacrine cells, and likely also to certain types of ganglion cells. Our results indicate that PDE9A, by controlling the level of cGMP, modulates inhibitory processes within the cone pathway. We speculate that these circuits involve NO/cGMP signaling pathways.

13.
Invest Ophthalmol Vis Sci ; 55(3): 1483-92, 2014 Mar 10.
Article in English | MEDLINE | ID: mdl-24519419

ABSTRACT

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.


Subject(s)
Calcium Channels/genetics , DNA/genetics , Dendrites/metabolism , Gene Expression Regulation , Receptors, Metabotropic Glutamate/genetics , Retinal Bipolar Cells/metabolism , Animals , Blotting, Western , Calcium Channels/biosynthesis , Calcium Channels, L-Type , Female , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , Rats , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/biosynthesis , Retinal Bipolar Cells/cytology , Signal Transduction
14.
Pigment Cell Melanoma Res ; 26(3): 348-56, 2013 May.
Article in English | MEDLINE | ID: mdl-23452348

ABSTRACT

Mutations in TRPM1, a calcium channel expressed in retinal bipolar cells and epidermal melanocytes, cause complete congenital stationary night blindness with no discernible skin phenotype. In the retina, TRPM1 activity is negatively coupled to metabotropic glutamate receptor 6 (mGluR6) signaling through Gαo and TRPM1 mutations result in the loss of responsiveness of TRPM1 to mGluR6 signaling. Here, we show that human melanocytes express mGluR6, and treatment of melanocytes with L-AP4, a type III mGluR-selective agonist, enhances Ca(2+) uptake. Knockdown of TRPM1 or mGluR6 by shRNA abolished L-AP4-induced Ca(2+) influx and TRPM1 currents, showing that TRPM1 activity in melanocytes is positively coupled to mGluR6 signaling. Gαo protein is absent in melanocytes. However, forced expression of Gαo restored negative coupling of TRPM1 to mGluR6 signaling, but treatment with pertussis toxin, an inhibitor of Gi /Go proteins, did not affect basal or mGluR6-induced Ca(2+) uptake. Additionally, chronic stimulation of mGluR6 altered melanocyte morphology and increased melanin content. These data suggest differences in coupling of TRPM1 function to mGluR6 signaling explain different cellular responses to glutamate in the retina and the skin.


Subject(s)
Melanins/metabolism , Melanocytes/metabolism , Receptors, Glutamate/metabolism , TRPM Cation Channels/metabolism , Calcium/metabolism , Cell Proliferation/drug effects , Epidermal Cells , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Gene Expression Regulation/drug effects , Glutamic Acid/pharmacology , Humans , Infant, Newborn , Male , Melanocytes/cytology , Melanocytes/drug effects , Patch-Clamp Techniques , Propionates/pharmacology , Receptors, Glutamate/genetics , Signal Transduction/drug effects
15.
J Neurosci ; 33(12): 5182-94, 2013 Mar 20.
Article in English | MEDLINE | ID: mdl-23516284

ABSTRACT

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.


Subject(s)
Heterotrimeric GTP-Binding Proteins/genetics , Retinal Cone Photoreceptor Cells/physiology , Transducin/genetics , Vision, Ocular/physiology , Animals , Color , Female , GABA Plasma Membrane Transport Proteins/genetics , Green Fluorescent Proteins/genetics , Heterotrimeric GTP-Binding Proteins/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Neurological , Photic Stimulation , Retinal Photoreceptor Cell Outer Segment/physiology , Transducin/physiology , Ultraviolet Rays
16.
J Biol Chem ; 288(10): 7420-9, 2013 Mar 08.
Article in English | MEDLINE | ID: mdl-23339194

ABSTRACT

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.


Subject(s)
Cell Membrane/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , Ion Channel Gating/physiology , Potassium Channels, Inwardly Rectifying/metabolism , Animals , Female , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein beta Subunits/genetics , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/genetics , GTP-Binding Protein gamma Subunits/metabolism , HEK293 Cells , Heterotrimeric GTP-Binding Proteins/genetics , Humans , Ion Channel Gating/genetics , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Oocytes/metabolism , Oocytes/physiology , Patch-Clamp Techniques , Potassium Channels, Inwardly Rectifying/genetics , Protein Binding , Retina/metabolism , Two-Hybrid System Techniques , Xenopus
17.
J Neurosci ; 32(33): 11343-55, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22895717

ABSTRACT

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.


Subject(s)
GTP-Binding Protein beta Subunits/metabolism , Gene Expression Regulation/genetics , Retinal Bipolar Cells/metabolism , Synapses/physiology , Animals , Choline O-Acetyltransferase/metabolism , Dendrites/ultrastructure , Electric Stimulation , Electroretinography , GTP-Binding Protein alpha Subunits/metabolism , GTP-Binding Protein beta Subunits/deficiency , GTPase-Activating Proteins/metabolism , Green Fluorescent Proteins/genetics , Heterotrimeric GTP-Binding Proteins/genetics , Heterotrimeric GTP-Binding Proteins/metabolism , Immunoprecipitation , In Vitro Techniques , Light , Membrane Potentials/drug effects , Membrane Potentials/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Transmission , Nerve Tissue Proteins/metabolism , Patch-Clamp Techniques , Photic Stimulation , Propionates/pharmacology , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/deficiency , Receptors, Metabotropic Glutamate/genetics , Retina/cytology , Retinal Bipolar Cells/drug effects , Retinal Bipolar Cells/ultrastructure , Retinal Cone Photoreceptor Cells/metabolism , Synapses/genetics , Synapses/metabolism , Synapses/ultrastructure , TRPM Cation Channels/metabolism , Visual Pathways/physiology
18.
Article in English | MEDLINE | ID: mdl-24003403

ABSTRACT

Glutamate, a key neurotransmitter in the vertebrate retina, acts via ionotropic and metabotropic receptors. Retina expresses mRNA for all metabotropic glutamate receptors and proteins for all but mGluR3. Every retinal cell class expresses one or more of these receptors. In general, these receptors are present presynaptically and serve to modulate synaptic transmission. While mGluRs on the photoreceptor terminal act as autoreceptors to titer glutamate levels, those on horizontal cell processes seem to shape the light response. Similarly, autoreceptors on bipolar axon terminals modulate glutamate release and the receptors on amacrine and ganglion cells modulate feedforward signals by modulating K+ or Ca2+ current to fine tune light responses. Since most of the mGluR sub-types are present in amacrine and ganglion cells that belong to many cell types, the pathways downstream of mGluRs are highly diverse with primarily modulatory effects. An exception to most mGluRs which have modulatory function is mGluR6 because it plays a key role in the feedforward transmission from photoreceptors to ON bipolar cells and is also required for the correct localization of the synaptic proteins in the dendritic tips. In humans, mutations in the gene encoding mGluR6 cause autosomal recessive night blindness. In addition, mGluRs appear to play a trophic role in development and after retinal damage, suggesting potential future therapeutic implications.

19.
J Neurophysiol ; 107(3): 948-57, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22131384

ABSTRACT

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.


Subject(s)
Receptors, Metabotropic Glutamate/physiology , Retinal Bipolar Cells/physiology , TRPM Cation Channels/physiology , Animals , Capsaicin/pharmacology , Dendrites/chemistry , Dendrites/physiology , Gene Deletion , Heterotrimeric GTP-Binding Proteins/analysis , Heterotrimeric GTP-Binding Proteins/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Metabotropic Glutamate/genetics , Retinal Bipolar Cells/drug effects , Sensory System Agents/pharmacology , TRPM Cation Channels/analysis
20.
J Histochem Cytochem ; 59(12): 1076-86, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22034516

ABSTRACT

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.


Subject(s)
Receptors, Metabotropic Glutamate/metabolism , Alternative Splicing , Animals , Brain/metabolism , Calcium/metabolism , Cornea/metabolism , Endothelial Cells/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Kidney/metabolism , Lymphoid Tissue/metabolism , Male , Mice , Mice, Transgenic , Organ Specificity , Promoter Regions, Genetic , Propionates/pharmacology , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/genetics , Retina/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Testis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...