Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 345
Filter
1.
Brain Struct Funct ; 229(5): 1279-1298, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38703218

ABSTRACT

ß-synuclein, a member of the synuclein family, is frequently co-expressed with α-synuclein in the neural system, where it serves to inhibit abnormal aggregation of α-synuclein in neurodegenerative diseases. Beyond its role in pathological conditions, ß-synuclein plays various functions independently of α-synuclein. In our investigation, we discovered a broader expression of ß-synuclein in the mouse retina compared to α-synuclein. This widespread pattern implies its potential significance in the retina. Through detailed examination via light- and electron-microscopic immunocytochemistry, we identified ß-synuclein expression from the inner segment (IS) and outer segment (OS) of photoreceptor cells to the ganglion cell layer (GCL). Our findings unveiled unique features, including ß-synuclein immunoreactive IS and OS of cones, higher expression in cone pedicles than in rod spherules, absence in horizontal cells, limited expression in cone bipolar dendrites and somas, higher expression in cone bipolar terminals, presence in most amacrine cells, and expression in almost majority of somas in GCL with an absence in intrinsically photosensitive retinal ganglion cell (ipRGCs) processes. Notably, all cholinergic amacrine cells express high ß- but not α-synuclein, while dopaminergic amacrine cells express α-synuclein exclusively. These distinctive expression patterns offer valuable insights for further exploration into the functions of ß-synuclein and its potential role in synuclein pathology within the retina.


Subject(s)
Mice, Inbred C57BL , Retina , Retinal Ganglion Cells , alpha-Synuclein , beta-Synuclein , Animals , beta-Synuclein/metabolism , Retina/metabolism , alpha-Synuclein/metabolism , Retinal Ganglion Cells/metabolism , Amacrine Cells/metabolism , Mice , Male , Retinal Bipolar Cells/metabolism
2.
Exp Eye Res ; 242: 109872, 2024 May.
Article in English | MEDLINE | ID: mdl-38514024

ABSTRACT

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


Subject(s)
Cell Adhesion Molecules , Disease Models, Animal , Electroretinography , Eye Proteins , Retinoschisis , Animals , Retinoschisis/genetics , Retinoschisis/physiopathology , Mice , Eye Proteins/genetics , Eye Proteins/metabolism , Photoreceptor Cells, Vertebrate/pathology , Mice, Inbred C57BL , Mutation , Ependymoglial Cells/pathology , Ependymoglial Cells/metabolism , Male , Retinal Bipolar Cells/pathology , Retinal Bipolar Cells/metabolism
3.
J Biol Chem ; 300(4): 107119, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38428819

ABSTRACT

Synaptic transmission from retinal photoreceptors to downstream ON-type bipolar cells (BCs) depends on the postsynaptic metabotropic glutamate receptor mGluR6, located at the BC dendritic tips. Glutamate binding to mGluR6 initiates G-protein signaling that ultimately leads to BC depolarization in response to light. The mGluR6 receptor also engages in trans-synaptic interactions with presynaptic ELFN adhesion proteins. The roles of post-translational modifications in mGluR6 trafficking and function are unknown. Treatment with glycosidase enzymes PNGase F and Endo H demonstrated that both endogenous and heterologously expressed mGluR6 contain complex N-glycosylation acquired in the Golgi. Pull-down experiments with ELFN1 and ELFN2 extracellular domains revealed that these proteins interact exclusively with the complex glycosylated form of mGluR6. Mutation of the four predicted N-glycosylation sites, either singly or in combination, revealed that all four sites are glycosylated. Single mutations partially reduced, but did not abolish, surface expression in heterologous cells, while triple mutants had little or no surface expression, indicating that no single glycosylation site is necessary or sufficient for plasma membrane trafficking. Mutation at N445 severely impaired both ELFN1 and ELFN2 binding. All single mutants exhibited dendritic tip enrichment in rod BCs, as did the triple mutant with N445 as the sole N-glycosylation site, demonstrating that glycosylation at N445 is sufficient but not necessary for dendritic tip localization. The quadruple mutant was completely mislocalized. These results reveal a key role for complex N-glycosylation in regulating mGluR6 trafficking and ELFN binding, and by extension, function of the photoreceptor synapses.


Subject(s)
Receptors, Metabotropic Glutamate , Animals , Humans , Mice , Glycosylation , HEK293 Cells , Protein Processing, Post-Translational , Protein Transport , Receptors, Metabotropic Glutamate/metabolism , Receptors, Metabotropic Glutamate/genetics , Retinal Bipolar Cells/metabolism , Synapses/metabolism , Synaptic Transmission/physiology
4.
J Biol Chem ; 300(4): 106794, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38403245

ABSTRACT

Retinal bipolar and amacrine cells receive visual information from photoreceptors and participate in the first steps of image processing in the retina. Several studies have suggested the operation of aerobic glycolysis and a lactate shuttle system in the retina due to the high production of this metabolite under aerobic conditions. However, whether bipolar cells form part of this metabolic circuit remains unclear. Here, we show that the monocarboxylate transporter 2 is expressed and functional in inner retinal neurons. Additionally, we used genetically encoded FRET nanosensors to demonstrate the ability of inner retinal neurons to consume extracellular lactate as an alternative to glucose. In rod bipolar cells, lactate consumption allowed cells to maintain the homeostasis of ions and electrical responses. We also found that lactate synthesis and transporter inhibition caused functional alterations and an increased rate of cell death. Overall, our data shed light on a notable but still poorly understood aspect of retinal metabolism.


Subject(s)
Lactic Acid , Monocarboxylic Acid Transporters , Retinal Bipolar Cells , Animals , Mice , Energy Metabolism , Glucose/metabolism , Lactic Acid/metabolism , Monocarboxylic Acid Transporters/metabolism , Monocarboxylic Acid Transporters/genetics , Retinal Bipolar Cells/metabolism
5.
PLoS One ; 18(11): e0290257, 2023.
Article in English | MEDLINE | ID: mdl-37910517

ABSTRACT

The retina is an intricately organized neural tissue built on cone and rod pathways for color and night vision. Genetic mutations that disrupt the proper function of the rod circuit contribute to blinding diseases including retinitis pigmentosa and congenital stationary night blindness (CSNB). Down Syndrome cell adhesion molecule like 1 (Dscaml1) is expressed by rods, rod bipolar cells (RBCs), and sub-populations of amacrine cells, and has been linked to a middle age onset of CSNB in humans. However, how Dscaml1 contributes to this visual deficit remains unexplored. Here, we probed Dscaml1's role in the maintenance of the rod-to-RBC synapse using a loss of function mouse model. We used immunohistochemistry to investigate the anatomical formation and maintenance of the rod-to-RBC synapse in the young, adult, and aging retina. We generated 3D reconstructions, using serial electron micrographs, of rod spherules and RBCs to measure the number of invaginating neurites, RBC dendritic tip number, and RBC mitochondrial morphology. We find that while rod-to-RBC synapses form and are maintained, similar to wildtype, that there is an increase in the number of invaginating neurites in rod spherules, a reduction in RBC dendritic tips, and reduced mitochondrial volume and complexity in the Dscaml1 mutant retina compared to controls. We also observed precocious sprouting of RBC dendrites into the outer nuclear layer (ONL) of the Dscaml1 mutant retina compared to controls. These results contribute to our knowledge of Dscaml1's role in rod circuit development and maintenance and give additional insight into possible genetic therapy targets for blinding diseases and disorders like CSNB.


Subject(s)
Retina , Synapses , Humans , Mice , Animals , Synapses/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Retinal Bipolar Cells/metabolism , Aging/genetics
6.
Mol Cell Neurosci ; 126: 103875, 2023 09.
Article in English | MEDLINE | ID: mdl-37352898

ABSTRACT

Metabotropic glutamate receptor 6 (mGluR6) predominantly localizes to the postsynaptic sites of retinal ON-bipolar cells, at which it recognizes glutamate released from photoreceptors. The C-terminal domain (CTD) of mGluR6 contains a cluster of basic amino acids resembling motifs for endoplasmic reticulum (ER) retention. We herein investigated whether these basic residues are involved in regulating the subcellular localization of mGluR6 in 293T cells expressing mGluR6 CTD mutants using immunocytochemistry, immunoprecipitation, and flow cytometry. We showed that full-length mGluR6 localized to the ER and cell surface, whereas mGluR6 mutants with 15- and 20-amino acid deletions from the C terminus localized to the ER, but were deficient at the cell surface. We also demonstrated that the cell surface deficiency of mGluR6 mutants was rescued by introducing an alanine substitution at basic residues within the CTD. The surface-deficient mGluR6 mutant still did not localize to the cell surface and was retained in the ER when co-expressed with surface-expressible constructs, including full-length mGluR6, even though surface-deficient and surface-expressible constructs formed heteromeric complexes. The co-expression of the surface-deficient mGluR6 mutant reduced the surface levels of surface-expressible constructs. These results indicate that basic residues in the mGluR6 CTD served as ER retention signals. We suggest that exposed ER retention motifs in the aberrant assembly containing truncated or misfolded mGluR6 prevent these protein complexes from being transported to the cell surface.


Subject(s)
Receptors, Metabotropic Glutamate , Receptors, Metabotropic Glutamate/genetics , Receptors, Metabotropic Glutamate/metabolism , Retinal Bipolar Cells/metabolism , Glutamic Acid/metabolism , Endoplasmic Reticulum/metabolism
7.
Invest Ophthalmol Vis Sci ; 63(11): 8, 2022 10 03.
Article in English | MEDLINE | ID: mdl-36227606

ABSTRACT

Purpose: Loss of retinoschisin (RS1) function underlies X-linked retinoschisis (XLRS) pathology. In the retina, both photoreceptor inner segments and bipolar cells express RS1. However, the loss of RS1 function causes schisis primarily in the inner retina. To understand these cell type-specific phenotypes, we decoupled RS1 effects in bipolar cells from that in photoreceptors. Methods: Bipolar cell transgene RS1 expression was achieved using two inner retina-specific promoters: (1) a minimal promoter engineered from glutamate receptor, metabotropic glutamate receptor 6 gene (mini-mGluR6/ Grm6) and (2) MiniPromoter (Ple155). Adeno-associated virus vectors encoding RS1 gene under either the mini-mGluR6 or Ple-155 promoter were delivered to the XLRS mouse retina through intravitreal or subretinal injection on postnatal day 14. Retinal structure and function were assessed 5 weeks later: immunohistochemistry for morphological characterization, optical coherence tomography and electroretinography (ERG) for structural and functional evaluation. Results: Immunohistochemical analysis of RS1expression showed that expression with the MiniPromoter (Ple155) was heavily enriched in bipolar cells. Despite variations in vector penetrance and gene transfer efficiency across the injected retinas, those retinal areas with robust bipolar cell RS1 expression showed tightly packed bipolar cells with fewer cavities and marked improvement in inner retinal structure and synaptic function as judged by optical coherence tomography and electroretinography, respectively. Conclusions: These results demonstrate that RS1 gene expression primarily in bipolar cells of the XLRS mouse retina, independent of photoreceptor expression, can ameliorate retinoschisis structural pathology and provide further evidence of RS1 role in cell adhesion.


Subject(s)
Cysts , Retinoschisis , Animals , Mice , Cysts/metabolism , Cysts/pathology , Electroretinography , Eye Proteins/genetics , Eye Proteins/metabolism , Retina/metabolism , Retina/pathology , Retinal Bipolar Cells/metabolism , Retinoschisis/genetics , Retinoschisis/metabolism
8.
Nat Commun ; 13(1): 5574, 2022 09 26.
Article in English | MEDLINE | ID: mdl-36163124

ABSTRACT

Motion sensing is a critical aspect of vision. We studied the representation of motion in mouse retinal bipolar cells and found that some bipolar cells are radially direction selective, preferring the origin of small object motion trajectories. Using a glutamate sensor, we directly observed bipolar cells synaptic output and found that there are radial direction selective and non-selective bipolar cell types, the majority being selective, and that radial direction selectivity relies on properties of the center-surround receptive field. We used these bipolar cell receptive fields along with connectomics to design biophysical models of downstream cells. The models and additional experiments demonstrated that bipolar cells pass radial direction selective excitation to starburst amacrine cells, which contributes to their directional tuning. As bipolar cells provide excitation to most amacrine and ganglion cells, their radial direction selectivity may contribute to motion processing throughout the visual system.


Subject(s)
Amacrine Cells , Retinal Bipolar Cells , Amacrine Cells/metabolism , Animals , Glutamic Acid/metabolism , Mice , Retina/metabolism , Retinal Bipolar Cells/metabolism
9.
eNeuro ; 9(3)2022.
Article in English | MEDLINE | ID: mdl-35523583

ABSTRACT

Excitatory amino acid transporters (EAATs) control visual signal transmission in the retina by rapidly removing glutamate released from photoreceptors and bipolar cells (BCs). Although it has been reported that EAAT2 and EAAT5 are expressed at presynaptic terminals of photoreceptors and some BCs in mammals, the distinct functions of these two glutamate transporters in retinal synaptic transmission, especially at a single synapse, remain elusive. In this study, we found that EAAT2 was expressed in all BC types while coexisting with EAAT5 in rod bipolar (RB) cells and several types of cone BCs from mice of either sex. Our immunohistochemical study, together with a recently published literature (Gehlen et al., 2021), showed that EAAT2 and EAAT5 were both located in RB axon terminals near release sites. Optogenetic, electrophysiological and pharmacological analyses, however, demonstrated that EAAT2 and EAAT5 regulated neurotransmission at RB→AII amacrine cell synapses in significantly different ways: EAAT5 dramatically affected both the peak amplitude and kinetics of postsynaptic responses in AIIs, whereas EAAT2 had either relatively small or opposite effects. By contrast, blockade of EAAT1/GLAST, which was exclusively expressed in Müller cells, showed no obvious effect on AII responses, indicating that glutamate uptake by Müller cells did not influence synaptic transmission from RB terminals. Furthermore, we found that temporal resolution at RB→AII synapses was reduced substantially by blockade of EAAT5 but not EAAT2. Taken together, our work reveals the distinct functions of EAAT2 and EAAT5 in signal transmission at RB ribbon synapses.


Subject(s)
Amino Acid Transport System X-AG , Excitatory Amino Acid Transporter 2/metabolism , Excitatory Amino Acid Transporter 5/metabolism , Retinal Bipolar Cells , Amino Acid Transport System X-AG/metabolism , Animals , Glutamic Acid/metabolism , Mammals/metabolism , Mice , Presynaptic Terminals/metabolism , Retina/metabolism , Retinal Bipolar Cells/metabolism , Synaptic Transmission/physiology
10.
J Comp Neurol ; 530(10): 1700-1728, 2022 07.
Article in English | MEDLINE | ID: mdl-35152437

ABSTRACT

Bipolar cells convey signals from photoreceptors in the outer retina to amacrine and ganglion cells in the inner retina. In mammals, there are typically 10-15 types of cone bipolar cells and one type of rod bipolar cell. Different types of cone bipolar cells are thought to code and transmit different features of a complex visual stimulus, thereby generating parallel channels that uniquely filter and transform the photoreceptor outputs. Differential synaptic connectivity and expression of ligand- and voltage-gated ion channels are thought to be important mechanisms for processing and filtering visual signals. Whereas the biophysical basis for such mechanisms has been investigated more extensively in rat retina, there is a lack of quantitative morphological data necessary for advancing the structure-function correlation in this species, as recent connectomics investigations have focused on mouse retina. Here, we performed whole-cell recordings from cone and rod bipolar cells in rat retinal slices, filled the cells with fluorescent dyes, and acquired image stacks by multiphoton excitation microscopy. Following deconvolution, we performed digital reconstruction and morphometric analysis of 25 cone and 14 rod bipolar cells. Compared to previous descriptions, the extent and complexity of branching of the axon terminal was surprisingly high. By precisely quantifying the level of stratification of the axon terminals in the inner plexiform layer, we have generated a reference system for reliable classification of individual cells in future studies focused on correlating physiological and morphological properties. The implemented workflow can be extended to the development of morphologically realistic compartmental models for these neurons.


Subject(s)
Dendrites , Retina , Animals , Axons , Dendrites/metabolism , Mammals , Mice , Rats , Retina/physiology , Retinal Bipolar Cells/metabolism , Retinal Cone Photoreceptor Cells/physiology
11.
Cells ; 11(1)2022 01 04.
Article in English | MEDLINE | ID: mdl-35011723

ABSTRACT

Retinal degeneration is a common feature in peroxisomal disorders leading to blindness. Peroxisomes are present in the different cell types of the retina; however, their precise contribution to retinal integrity is still unclear. We previously showed that mice lacking the central peroxisomal ß-oxidation enzyme, multifunctional protein 2 (MFP2), develop an early onset retinal decay including photoreceptor cell death. To decipher the function of peroxisomal ß-oxidation in photoreceptors, we generated cell type selective Mfp2 knockout mice, using the Crx promotor targeting photoreceptors and bipolar cells. Surprisingly, Crx-Mfp2-/- mice maintained photoreceptor length and number until the age of 1 year. A negative electroretinogram was indicative of preserved photoreceptor phototransduction, but impaired downstream bipolar cell signaling from the age of 6 months. The photoreceptor ribbon synapse was affected, containing free-floating ribbons and vesicles with altered size and density. The bipolar cell interneurons sprouted into the ONL and died. Whereas docosahexaenoic acid levels were normal in the neural retina, levels of lipids containing very long chain polyunsaturated fatty acids were highly increased. Crx-Pex5-/- mice, in which all peroxisomal functions are inactivated in photoreceptors and bipolar cells, developed the same phenotype as Crx-Mfp2-/- mice. In conclusion, the early photoreceptor death in global Mfp2-/- mice is not driven cell autonomously. However, peroxisomal ß-oxidation is essential for the integrity of photoreceptor ribbon synapses and of bipolar cells.


Subject(s)
Peroxisomes/metabolism , Photoreceptor Cells/metabolism , Retinal Bipolar Cells/metabolism , Animals , Humans , Mice , Mice, Knockout
12.
Cell Rep ; 38(1): 110191, 2022 01 04.
Article in English | MEDLINE | ID: mdl-34986354

ABSTRACT

How do neuronal subtypes emerge during development? Recent molecular studies have profiled existing neuronal diversity, but neuronal subtype genesis remains elusive. The 15 types of mouse retinal bipolar interneurons are characterized by distinct functions, morphologies, and transcriptional profiles. Here, we develop a comprehensive spatiotemporal map of bipolar subtype genesis in the murine retina. Combining multiplexed detection of 16 RNA markers with timed delivery of 5-ethynyl uridine (EdU) and bromodeoxyuridine (BrdU), we analyze more than 30,000 single cells in full retinal sections to classify all bipolar subtypes and their birthdates. We find that bipolar subtype birthdates are ordered and follow a centrifugal developmental axis. Spatial analysis reveals a striking wave pattern of bipolar subtype birthdates, and lineage analyses suggest clonal restriction on homotypic subtype production. These results inspire a hierarchical developmental model, with ordered subtype genesis within lineages. Our results provide insight into neuronal subtype development and establish a framework for studying subtype diversification.


Subject(s)
Cell Lineage/physiology , Neurogenesis/physiology , Retinal Bipolar Cells/cytology , Spatio-Temporal Analysis , Animals , Female , Gene Expression Regulation, Developmental/genetics , Male , Mice , Mice, Inbred C57BL , RNA/genetics , Retina/cytology , Retina/metabolism , Retinal Bipolar Cells/metabolism
13.
Dev Biol ; 481: 30-42, 2022 01.
Article in English | MEDLINE | ID: mdl-34534525

ABSTRACT

The bipolar interneurons of the mammalian retina have evolved as a diverse set of cells with distinct subtype characteristics, which reflect specialized contributions to visual circuitry. Fifteen subtypes of bipolar interneurons have been identified in the mouse retina, each with characteristic gene expression, morphology, and light responses. This review provides an overview of the developmental events that underlie the generation of the diverse bipolar cell class, summarizing the current knowledge of genetic programs that establish and maintain bipolar subtype fates, as well as the events that shape the final distribution of bipolar subtypes. With much left to be discovered, bipolar interneurons present an ideal model system for studying the interplay between cell-autonomous and non-cell-autonomous mechanisms that influence neuronal subtype development within the central nervous system.


Subject(s)
Cell Differentiation , Central Nervous System/embryology , Gene Expression Regulation, Developmental , Neurogenesis , Retina/embryology , Retinal Bipolar Cells/metabolism , Animals , Mice
14.
Int J Mol Sci ; 22(23)2021 Dec 03.
Article in English | MEDLINE | ID: mdl-34884916

ABSTRACT

The viral gene delivery of optogenetic actuators to the surviving inner retina has been proposed as a strategy for restoring vision in advanced retinal degeneration. We investigated the safety of ectopic expression of human rod opsin (hRHO), and two channelrhodopsins (enhanced sensitivity CoChR-3M and red-shifted ReaChR) by viral gene delivery in ON bipolar cells of the mouse retina. Adult Grm6Cre mice were bred to be retinally degenerate or non-retinally degenerate (homozygous and heterozygous for the rd1Pde6b mutation, respectively) and intravitreally injected with recombinant adeno-associated virus AAV2/2(quad Y-F) serotype containing a double-floxed inverted transgene comprising one of the opsins of interest under a CMV promoter. None of the opsins investigated caused changes in retinal thickness; induced apoptosis in the retina or in transgene expressing cells; or reduced expression of PKCα (a specific bipolar cell marker). No increase in retinal inflammation at the level of gene expression (IBA1/AIF1) was found within the treated mice compared to controls. The expression of hRHO, CoChR or ReaChR under a strong constitutive promoter in retinal ON bipolar cells following intravitreal delivery via AAV2 does not cause either gross changes in retinal health, or have a measurable impact on the survival of targeted cells.


Subject(s)
Channelrhodopsins/genetics , Genetic Variation , Retinal Bipolar Cells/metabolism , Rod Opsins/genetics , Animals , Channelrhodopsins/metabolism , Dependovirus/genetics , Humans , Intravitreal Injections , Mice , Optogenetics , Rod Opsins/metabolism , Transduction, Genetic
15.
J Biol Chem ; 297(6): 101418, 2021 12.
Article in English | MEDLINE | ID: mdl-34793838

ABSTRACT

Signals from retinal photoreceptors are processed in two parallel channels-the ON channel responds to light increments, while the OFF channel responds to light decrements. The ON pathway is mediated by ON type bipolar cells (BCs), which receive glutamatergic synaptic input from photoreceptors via a G-protein-coupled receptor signaling cascade. The metabotropic glutamate receptor mGluR6 is located at the dendritic tips of all ON-BCs and is required for synaptic transmission. Thus, it is critically important for delivery of information from photoreceptors into the ON pathway. In addition to detecting glutamate, mGluR6 participates in interactions with other postsynaptic proteins, as well as trans-synaptic interactions with presynaptic ELFN proteins. Mechanisms of mGluR6 synaptic targeting and functional interaction with other synaptic proteins are unknown. Here, we show that multiple regions in the mGluR6 ligand-binding domain are necessary for both synaptic localization in BCs and ELFN1 binding in vitro. However, these regions were not required for plasma membrane localization in heterologous cells, indicating that secretory trafficking and synaptic localization are controlled by different mechanisms. In contrast, the mGluR6 C-terminus was dispensable for synaptic localization. In mGluR6 null mice, localization of the postsynaptic channel protein TRPM1 was compromised. Introducing WT mGluR6 rescued TRPM1 localization, while a C-terminal deletion mutant had significantly reduced rescue ability. We propose a model in which trans-synaptic ELFN1 binding is necessary for mGluR6 postsynaptic localization, whereas the C-terminus has a role in mediating TRPM1 trafficking. These findings reveal different sequence determinants of the multifunctional roles of mGluR6 in ON-BCs.


Subject(s)
Receptors, Metabotropic Glutamate/metabolism , Retinal Bipolar Cells/metabolism , Synapses/metabolism , Synaptic Transmission , TRPM Cation Channels/metabolism , Animals , Cell Line , Mice , Mutation , Photoreceptor Cells, Vertebrate/metabolism , Protein Domains , Protein Transport , Receptors, Metabotropic Glutamate/genetics , Synapses/genetics , TRPM Cation Channels/genetics
16.
Exp Eye Res ; 212: 108770, 2021 11.
Article in English | MEDLINE | ID: mdl-34562437

ABSTRACT

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


Subject(s)
Autoantibodies/blood , Paraneoplastic Syndromes, Ocular/immunology , Retinal Bipolar Cells/metabolism , TRPM Cation Channels/immunology , Aged , Blotting, Western , Electroretinography , Female , Humans , Male , Middle Aged , Paraneoplastic Syndromes, Ocular/metabolism , Paraneoplastic Syndromes, Ocular/pathology , Retinal Bipolar Cells/pathology , Retrospective Studies , Tumor Cells, Cultured
17.
Int J Mol Sci ; 22(15)2021 Jul 29.
Article in English | MEDLINE | ID: mdl-34360929

ABSTRACT

Complexins (Cplxs) 1 to 4 are components of the presynaptic compartment of chemical synapses where they regulate important steps in synaptic vesicle exocytosis. In the retina, all four Cplxs are present, and while we know a lot about Cplxs 3 and 4, little is known about Cplxs 1 and 2. Here, we performed in situ hybridization experiments and bioinformatics and exploited Cplx 1 and Cplx 2 single-knockout mice combined with immunocytochemistry and light microscopy to characterize in detail the cell type and synapse-specific distribution of Cplx 1 and Cplx 2. We found that Cplx 2 and not Cplx 1 is the main isoform expressed in normal and displaced amacrine cells and ganglion cells in mouse retinae and that amacrine cells seem to operate with a single Cplx isoform at their conventional chemical synapses. Surprising was the finding that retinal function, determined with electroretinographic recordings, was altered in Cplx 1 but not Cplx 2 single-knockout mice. In summary, the results provide an important basis for future studies on the function of Cplxs 1 and 2 in the processing of visual signals in the mammalian retina.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Amacrine Cells/metabolism , Nerve Tissue Proteins/metabolism , Photoreceptor Cells/metabolism , Retinal Bipolar Cells/metabolism , Retinal Ganglion Cells/metabolism , Retinal Horizontal Cells/metabolism , SNARE Proteins/metabolism , Synapses/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Animals , Cells, Cultured , Computational Biology/methods , Electroretinography/methods , Female , Immunohistochemistry/methods , In Situ Hybridization/methods , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics
18.
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
19.
Int J Mol Sci ; 22(8)2021 Apr 14.
Article in English | MEDLINE | ID: mdl-33919796

ABSTRACT

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


Subject(s)
Calcium/metabolism , Cone Dystrophy/genetics , Cone Dystrophy/physiopathology , Guanylate Cyclase-Activating Proteins/genetics , Mutation/genetics , Retinal Bipolar Cells/pathology , Retinal Rod Photoreceptor Cells/pathology , Synaptic Transmission , Atrophy , Cations , Cone Dystrophy/diagnostic imaging , Disease Progression , Electroretinography , Female , Fundus Oculi , Guanylate Cyclase/metabolism , Guanylate Cyclase-Activating Proteins/chemistry , Heterozygote , Humans , Hydrodynamics , Hydrophobic and Hydrophilic Interactions , Middle Aged , Molecular Dynamics Simulation , Phenotype , Protein Aggregates , Protein Stability , Protein Structure, Quaternary , Retinal Bipolar Cells/metabolism , Retinal Pigment Epithelium/pathology , Retinal Rod Photoreceptor Cells/metabolism , Tomography, Optical Coherence
20.
Cell Rep ; 35(3): 109022, 2021 04 20.
Article in English | MEDLINE | ID: mdl-33882303

ABSTRACT

Age-related macular degeneration and other macular diseases result in the loss of light-sensing cone photoreceptors, causing irreversible sight impairment. Photoreceptor replacement may restore vision by transplanting healthy cells, which must form new synaptic connections with the recipient retina. Despite recent advances, convincing evidence of functional connectivity arising from transplanted human cone photoreceptors in advanced retinal degeneration is lacking. Here, we show restoration of visual function after transplantation of purified human pluripotent stem cell-derived cones into a mouse model of advanced degeneration. Transplanted human cones elaborate nascent outer segments and make putative synapses with recipient murine bipolar cells (BCs), which themselves undergo significant remodeling. Electrophysiological and behavioral assessments demonstrate restoration of surprisingly complex light-evoked retinal ganglion cell responses and improved light-evoked behaviors in treated animals. Stringent controls exclude alternative explanations, including material transfer and neuroprotection. These data provide crucial validation for photoreceptor replacement therapy and for the potential to rescue cone-mediated vision.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Macular Degeneration/therapy , Organoids/transplantation , Recovery of Function/physiology , Retinal Cone Photoreceptor Cells/metabolism , Animals , Biomarkers/metabolism , Cell Differentiation , Dependovirus/genetics , Dependovirus/metabolism , Disease Models, Animal , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Macular Degeneration/genetics , Macular Degeneration/metabolism , Macular Degeneration/pathology , Male , Mice , Mice, Transgenic , Mycotoxins/genetics , Mycotoxins/metabolism , Organoids/cytology , Organoids/metabolism , Peripherins/genetics , Peripherins/metabolism , Photic Stimulation , Primary Cell Culture , Protein Kinase C-alpha/genetics , Protein Kinase C-alpha/metabolism , Receptors, Glutamate/genetics , Receptors, Glutamate/metabolism , Retinal Bipolar Cells/cytology , Retinal Bipolar Cells/metabolism , Retinal Cone Photoreceptor Cells/cytology , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism , Synapses/metabolism , Transplantation, Heterologous , Vision, Ocular/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...