Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters











Publication year range
1.
Proc Natl Acad Sci U S A ; 118(45)2021 11 09.
Article in English | MEDLINE | ID: mdl-34732574

ABSTRACT

Cone photoreceptors mediate daylight vision in vertebrates. Changes in neurotransmitter release at cone synapses encode visual information and is subject to precise control by negative feedback from enigmatic horizontal cells. However, the mechanisms that orchestrate this modulation are poorly understood due to a virtually unknown landscape of molecular players. Here, we report a molecular player operating selectively at cone synapses that modulates effects of horizontal cells on synaptic release. Using an unbiased proteomic screen, we identified an adhesion GPCR Latrophilin3 (LPHN3) in horizontal cell dendrites that engages in transsynaptic control of cones. We detected and characterized a prominent splice isoform of LPHN3 that excludes a element with inhibitory influence on transsynaptic interactions. A gain-of-function mouse model specifically routing LPHN3 splicing to this isoform but not knockout of LPHN3 diminished CaV1.4 calcium channel activity profoundly disrupted synaptic release by cones and resulted in synaptic transmission deficits. These findings offer molecular insight into horizontal cell modulation on cone synaptic function and more broadly demonstrate the importance of alternative splicing in adhesion GPCRs for their physiological function.


Subject(s)
Alternative Splicing , Receptors, G-Protein-Coupled/metabolism , Receptors, Peptide/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Synapses/metabolism , Animals , Calcium Channels/metabolism , Mice , Mice, Knockout , Protein Isoforms/metabolism , Proteome , Receptors, G-Protein-Coupled/genetics , Receptors, Peptide/genetics
2.
Nat Commun ; 11(1): 3328, 2020 07 03.
Article in English | MEDLINE | ID: mdl-32620864

ABSTRACT

Genes encoding cell-surface proteins control nervous system development and are implicated in neurological disorders. These genes produce alternative mRNA isoforms which remain poorly characterized, impeding understanding of how disease-associated mutations cause pathology. Here we introduce a strategy to define complete portfolios of full-length isoforms encoded by individual genes. Applying this approach to neural cell-surface molecules, we identify thousands of unannotated isoforms expressed in retina and brain. By mass spectrometry we confirm expression of newly-discovered proteins on the cell surface in vivo. Remarkably, we discover that the major isoform of a retinal degeneration gene, CRB1, was previously overlooked. This CRB1 isoform is the only one expressed by photoreceptors, the affected cells in CRB1 disease. Using mouse mutants, we identify a function for this isoform at photoreceptor-glial junctions and demonstrate that loss of this isoform accelerates photoreceptor death. Therefore, our isoform identification strategy enables discovery of new gene functions relevant to disease.


Subject(s)
Genetic Variation , Membrane Proteins/genetics , Photoreceptor Cells, Vertebrate/metabolism , RNA Isoforms/genetics , Retina/metabolism , Retinal Degeneration/genetics , Amino Acid Sequence , Animals , Base Sequence , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental , Humans , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , RNA Isoforms/metabolism , Retina/cytology , Retina/growth & development , Retinal Degeneration/metabolism , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid
3.
eNeuro ; 7(1)2020.
Article in English | MEDLINE | ID: mdl-31959619

ABSTRACT

The first retinal synapse, photoreceptor→bipolar cell (BC), is both anatomically and functionally complex. Within the same synaptic region, a change in presynaptic glutamate release is sensed by both ON BCs (DBCs) via the metabotropic glutamate receptor 6 (mGluR6), and OFF BCs (HBCs) via ionotropic glutamate receptors to establish parallel signaling pathways that preferentially encode light increments (ON) or decrements (OFF), respectively. The synaptic structural organization of ON and OFF-type BCs at the photoreceptor terminal differs. DBCs make an invaginating synapse that contains a diverse but incompletely understood complex of interacting proteins (signalplex). HBCs make primarily flat contacts that contain an apparent different set of proteins that is equally uncharacterized. LRIT3 is a synaptic protein known to be essential for ON pathway visual function. In both male and female mice, we demonstrate that LRIT3 interacts with and is required for expression of nyctalopin, and thus TRPM1 at all DBC dendritic tips, but DBC signalplex components are not required for LRIT3 expression. Using whole-cell and multielectrode array (MEA) electrophysiology and glutamate imaging, we demonstrate that the loss of LRIT3 impacts both ON and OFF signaling pathway function. Without LRIT3, excitatory input to type 1 BCs is reduced, as are the visually evoked responses of many OFF retinal ganglion cells (RGCs). We conclude that the absence of LRIT3 expression disrupts excitatory input to OFF BCs and, thus disrupts the normal function of OFF RGCs.


Subject(s)
Membrane Proteins , Retina , Retinal Bipolar Cells , Signal Transduction , Animals , Female , Male , Mice , Mice, Inbred C57BL , Synapses
4.
Adv Exp Med Biol ; 1185: 251-255, 2019.
Article in English | MEDLINE | ID: mdl-31884620

ABSTRACT

Mutations in the gene Crumbs homolog 1 (CRB1) are responsible for several retinopathies that are diverse in severity and phenotype. Thus, there is considerable incentive to determine how disruption of this gene causes disease. Progress on this front will aid in developing molecular diagnostics that can predict disease severity with the ultimate goal of developing therapies for CRB1 retinopathies via gene replacement. The purpose of this review is to summarize what is known regarding CRB1 and highlights information outstanding. Doing so will provide a framework toward a thorough understanding of CRB1 at the molecular and protein level with the ultimate goal of deciphering how it contributes to the disease.


Subject(s)
Eye Proteins/genetics , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Retinal Diseases/genetics , Animals , Disease Models, Animal , Humans , Mice , Mice, Knockout , Mutation , Paraneoplastic Syndromes, Ocular
5.
Cell Rep ; 27(11): 3107-3116.e3, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31189098

ABSTRACT

Throughout the CNS, interactions between pre- and postsynaptic adhesion molecules establish normal synaptic structure and function. Leucine-rich repeat (LRR) domain-containing proteins are a large family that has a diversity of ligands, and their absence can cause disease. At the first retinal synapse, the absence of LRIT3 expression leads to the disassembly of the postsynaptic glutamate signaling complex (signalplex) expressed on depolarizing bipolar cell (DBC) dendrites. The prevalent view is that assembly of the signalplex results from direct postsynaptic protein:protein interactions. In contrast, we demonstrate that LRIT3 is expressed presynaptically, in rod photoreceptors (rods), and when we restore LRIT3 expression in Lrit3-/- rods, we restore expression of the postsynaptic glutamate signalplex and rod-driven vision. Our results demonstrate that, in the retina, the LRR-containing protein LRIT3 acts as a transsynaptic organizer of the postsynaptic complex required for normal synaptic function.


Subject(s)
Glutamic Acid/metabolism , Membrane Proteins/metabolism , Synapses/metabolism , TRPM Cation Channels/metabolism , Animals , Dendrites/metabolism , Dendrites/physiology , Female , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Retinal Bipolar Cells/metabolism , Retinal Bipolar Cells/physiology , Retinal Rod Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/physiology , Synapses/physiology , Synaptic Potentials
6.
Elife ; 72018 04 03.
Article in English | MEDLINE | ID: mdl-29611808

ABSTRACT

A common strategy by which developing neurons locate their synaptic partners is through projections to circuit-specific neuropil sublayers. Once established, sublayers serve as a substrate for selective synapse formation, but how sublayers arise during neurodevelopment remains unknown. Here, we identify the earliest events that initiate formation of the direction-selective circuit in the inner plexiform layer of mouse retina. We demonstrate that radially migrating newborn starburst amacrine cells establish homotypic contacts on arrival at the inner retina. These contacts, mediated by the cell-surface protein MEGF10, trigger neuropil innervation resulting in generation of two sublayers comprising starburst-cell dendrites. This dendritic scaffold then recruits projections from circuit partners. Abolishing MEGF10-mediated contacts profoundly delays and ultimately disrupts sublayer formation, leading to broader direction tuning and weaker direction-selectivity in retinal ganglion cells. Our findings reveal a mechanism by which differentiating neurons transition from migratory to mature morphology, and highlight this mechanism's importance in forming circuit-specific sublayers.


Subject(s)
Amacrine Cells/physiology , Neuropil/physiology , Retina/embryology , Retinal Ganglion Cells/physiology , Animals , Membrane Proteins/metabolism , Mice
7.
Vis Neurosci ; 33: E009, 2016 01.
Article in English | MEDLINE | ID: mdl-27471951

ABSTRACT

Cacna1s encodes the α1S subunit (Cav1.1) of voltage-dependent calcium channels, and is required for normal skeletal and cardiac muscle function, where it couples with the ryanodine receptor to regulate muscle contraction. Recently CACNA1S was reported to be expressed on the tips of retinal depolarizing bipolar cells (DBCs) and colocalized with metabotropic glutamate receptor 6 (mGluR6), which is critical to DBC signal transduction. Further, in mGluR6 knockout mice, expression at this location is down regulated. We examined RNAseq data from mouse retina and found expression of a novel isoform of Cacna1s. To determine if CACNA1S was a functional component of the DBC signal transduction cascade, we performed immunohistochemistry to visualize its expression in several mouse lines that lack DBC function. Immunohistochemical staining with antibodies to CACNA1S show punctate labeling at the tips of DBCs in wild type (WT) retinas that are absent in Gpr179 nob5 mutant retinas and decreased in Grm6 -/- mouse retinas. CACNA1S and transient receptor potential cation channel, subfamily M, member 1 (TRPM1) staining also colocalized in WT retinas. Western blot analyses for CACNA1S of either retinal lysates or proteins after immunoprecipitation with the CACNA1S antibody failed to show the presence of bands expected for CACNA1S. Mass spectrometric analysis of CACNA1S immunoprecipitated proteins also failed to detect any peptides matching CACNA1S. Immunohistochemistry and western blotting after expression of GPR179 in HEK293T cells indicate that the CACNA1S antibody used here and in the retinal studies published to date, cross-reacts with GPR179. These data suggest caution should be exercised in conferring a role for CACNA1S in DBC signal transduction based solely on immunohistochemical staining.


Subject(s)
Antigen-Antibody Reactions , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/immunology , Gene Expression Regulation/physiology , Receptors, G-Protein-Coupled/immunology , Retina/metabolism , Retinal Bipolar Cells/metabolism , Amino Acid Sequence , Animals , Blotting, Western , Cross Reactions , Female , HEK293 Cells , Humans , Immunohistochemistry , Immunologic Deficiency Syndromes , Male , Mass Spectrometry , Mice , Mice, Knockout , Molecular Sequence Data , Primary Immunodeficiency Diseases , Protein Isoforms , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Signal Transduction
8.
Elife ; 4: e08149, 2015 Dec 02.
Article in English | MEDLINE | ID: mdl-26633812

ABSTRACT

In the inner plexiform layer (IPL) of the mouse retina, ~70 neuronal subtypes organize their neurites into an intricate laminar structure that underlies visual processing. To find recognition proteins involved in lamination, we utilized microarray data from 13 subtypes to identify differentially-expressed extracellular proteins and performed a high-throughput biochemical screen. We identified ~50 previously-unknown receptor-ligand pairs, including new interactions among members of the FLRT and Unc5 families. These proteins show laminar-restricted IPL localization and induce attraction and/or repulsion of retinal neurites in culture, placing them in an ideal position to mediate laminar targeting. Consistent with a repulsive role in arbor lamination, we observed complementary expression patterns for one interaction pair, FLRT2-Unc5C, in vivo. Starburst amacrine cells and their synaptic partners, ON-OFF direction-selective ganglion cells, express FLRT2 and are repelled by Unc5C. These data suggest a single molecular mechanism may have been co-opted by synaptic partners to ensure joint laminar restriction.


Subject(s)
Cell Communication , Membrane Glycoproteins/metabolism , Neurons/physiology , Receptors, Cell Surface/metabolism , Retina/anatomy & histology , Retina/physiology , Animals , Biochemistry/methods , Gene Expression Profiling , Mice , Microarray Analysis , Netrin Receptors , Protein Binding , Protein Interaction Mapping , Proteome/analysis
9.
Neuron ; 86(4): 855-857, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25996129

ABSTRACT

Different types of retinal ganglion cells (RGCs) project to distinct brain targets. In this issue of Neuron, Osterhout et al. (2015) and Sun et al. (2015) identify how direction-selective RGC axons match with their targets and the consequences for visual function when targeting is impaired.


Subject(s)
Axons/metabolism , Brain/metabolism , Contactins/metabolism , Eye Movements/physiology , Retina/metabolism , Retinal Ganglion Cells/metabolism , Visual Pathways/metabolism , Visual Pathways/physiology , Animals
10.
J Neurosci ; 34(18): 6334-43, 2014 Apr 30.
Article in English | MEDLINE | ID: mdl-24790204

ABSTRACT

Parallel visual pathways are initiated at the first retinal synapse by signaling between the rod and cone photoreceptors and two general classes of bipolar cells. For normal function, ON or depolarizing bipolar cells (DBCs) require the G-protein-coupled receptor, mGluR6, an intact G-protein-coupled cascade and the transient receptor potential melastatin 1 (TRPM1) cation channel. In addition, another seven transmembrane protein, GPR179, is required for DBC function and recruits the regulators of G-protein signaling (RGS) proteins, RGS7 and RGS11, to the dendritic tips of the DBCs. Here we use the Gpr179(nob5) mouse, which lacks GPR179 and has a no b-wave electroretinogram (ERG) phenotype, to demonstrate that despite the absence of both GPR179 and RGS7/RGS11, a small dark-adapted ERG b-wave remains and can be enhanced with long duration flashes. Consistent with the ERG, the mGluR6-mediated gating of TRPM1 can be evoked pharmacologically in Gpr179(nob5) and RGS7(-/-)/RGS11(-/-) rod BCs if strong stimulation conditions are used. In contrast, direct gating of TRPM1 by capsaicin in RGS7(-/-)/RGS11(-/-) and WT rod BCs is similar, but severely compromised in Gpr179(nob5) rod BCs. Noise and standing current analyses indicate that the remaining channels in Gpr179(nob5) and RGS7(-/-)/RGS11(-/-) rod BCs have a very low open probability. We propose that GPR179 along with RGS7 and RGS11 controls the ability of the mGluR6 cascade to gate TRPM1. In addition to its role in localizing RGS7 and RGS11 to the dendritic tips, GPR179 via a direct interaction with the TRPM1 channel alters its ability to be gated directly by capsaicin.


Subject(s)
Gene Expression Regulation/physiology , Receptors, G-Protein-Coupled/metabolism , Receptors, Metabotropic Glutamate/metabolism , Retinal Bipolar Cells/metabolism , Signal Transduction/physiology , Animals , Capsaicin/pharmacology , Cell Line, Transformed , Excitatory Amino Acid Antagonists/pharmacology , Gene Expression Regulation/genetics , Glycine/analogs & derivatives , Glycine/pharmacology , Glycine Agents/pharmacology , Humans , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proteoglycans/metabolism , Receptors, GABA-A/genetics , Retina/cytology , Retina/metabolism , Retinal Bipolar Cells/cytology , Retinal Bipolar Cells/drug effects , Signal Transduction/genetics , Strychnine/pharmacology , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism
11.
Mol Vis ; 19: 2615-25, 2013.
Article in English | MEDLINE | ID: mdl-24415894

ABSTRACT

PURPOSE: To identify the mutation responsible for an abnormal electroretinogram (ERG) in a transgenic mouse line (tg21) overexpressing erythropoietin (Epo). The tg21 line was generated on a mixed (C3H; C57BL/6) background and lacked the b-wave component of the ERG. This no-b-wave (nob) ERG is seen in other mouse models with depolarizing bipolar cell (DBC) dysfunction and in patients with the complete form of congenital stationary night blindness (cCSNB). We determined the basis for the nob ERG phenotype and screened C3H mice for the mutation to evaluate whether this finding is important for the vision research community. METHODS: ERGs were used to examine retinal function. The retinal structure of the transgenic mice was investigated using histology and immunohistochemistry. Inverse PCR was performed to identify the insertion site of the Epo transgene in the mouse genome. Affected mice were backcrossed to follow the inheritance pattern of the nob ERG phenotype. Quantitative real-time PCR (qRT PCR), Sanger sequencing, and immunohistochemistry were used to identify the mutation causing the defect. Additional C3H sublines were screened for the detected mutation. RESULTS: Retinal histology and blood vessel structure were not disturbed, and no loss of DBCs was observed in the tg21 nob mice. The mutation causing the nob ERG phenotype is inherited independently of the tg21 transgene. The qRT PCR experiments revealed that the nob ERG phenotype reflected a mutation in Gpr179, a gene involved in DBC signal transduction. PCR analysis confirmed the presence of the Gpr179(nob5) insertional mutation in intron 1 of Gpr179. Screening for mutations in other C3H-derived lines revealed that C3H.Pde6b(+) mice carry the Gpr179 (nob5) allele whereas C3H/HeH mice do not. CONCLUSIONS: We identified the presence of the Gpr179(nob5) mutation causing DBC dysfunction in a C3H-derived transgenic mouse line. The nob phenotype is not related to the presence of the transgene. The Gpr179(nob5) allele can be added to the list of background alleles that impact retinal function in commonly used mouse lines. By providing primers to distinguish between Gpr179 mutant and wild-type alleles, this study allows investigators to monitor for the presence of the Gpr179(nob5) mutation in other mouse lines derived from C3H.


Subject(s)
Alleles , Erythropoietin/genetics , Mutagenesis, Insertional , Night Blindness/genetics , Receptors, G-Protein-Coupled/genetics , Retinal Bipolar Cells/metabolism , Animals , Crosses, Genetic , Disease Models, Animal , Electroretinography , Erythropoietin/metabolism , Female , Founder Effect , Gene Expression , Humans , Male , Mice , Mice, Inbred C3H , Mice, Transgenic , Night Blindness/metabolism , Night Blindness/pathology , Receptors, G-Protein-Coupled/metabolism , Retinal Bipolar Cells/pathology , Signal Transduction , Transgenes
12.
J Neurophysiol ; 108(9): 2442-51, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22896717

ABSTRACT

Mutations in TRPM1 are found in humans with an autosomal recessive form of complete congenital stationary night blindness (cCSNB). The Trpm1(-/-) mouse has been an important animal model for this condition. Here we report a new mouse mutant, tvrm27, identified in a chemical mutagenesis screen. Genetic mapping of the no b-wave electroretinogram (ERG) phenotype of tvrm27 localized the mutation to a chromosomal region that included Trpm1. Complementation testing with Trpm1(-/-) mice confirmed a mutation in Trpm1. Sequencing identified a nucleotide change in exon 23, converting a highly conserved alanine within the pore domain to threonine (p.A1068T). Consistent with prior studies of Trpm1(-/-) mice, no anatomical changes were noted in the Trpm1(tvrm27/tvrm27) retina. The Trpm1(tvrm27/tvrm27) phenotype is distinguished from that of Trpm1(-/-) by the retention of TRPM1 expression on the dendritic tips of depolarizing bipolar cells (DBCs). While ERG b-wave amplitudes of Trpm1(+/-) heterozygotes are comparable to wild type, those of Trpm1(+/tvrm27) mice are reduced by 32%. A similar reduction in the response of Trpm1(+/tvrm27) DBCs to LY341495 or capsaicin is evident in whole cell recordings. These data indicate that the p.A1068T mutant TRPM1 acts as a dominant negative with respect to TRPM1 channel function. Furthermore, these data indicate that the number of functional TRPM1 channels at the DBC dendritic tips is a key factor in defining DBC response amplitude. The Trpm1(tvrm27/tvrm27) mutant will be useful for elucidating the role of TRPM1 in DBC signal transduction, for determining how Trpm1 mutations impact central visual processing, and for evaluating experimental therapies for cCSNB.


Subject(s)
Point Mutation , Retinal Bipolar Cells/physiology , TRPM Cation Channels/genetics , Action Potentials/drug effects , Action Potentials/physiology , Amino Acid Sequence , Amino Acids/pharmacology , Animals , Capsaicin/pharmacology , Chromosome Mapping , Chromosomes, Mammalian/genetics , Dendrites/physiology , Disease Models, Animal , Exons , Eye Diseases, Hereditary/genetics , Genetic Diseases, X-Linked/genetics , Heterozygote , Mice , Mice, Transgenic , Molecular Sequence Data , Mutation, Missense , Myopia/genetics , Night Blindness/genetics , Retina/pathology , Retina/physiology , Sequence Analysis, DNA , TRPM Cation Channels/metabolism , Threonine/genetics , Xanthenes/pharmacology
13.
J Cell Biol ; 197(6): 711-9, 2012 Jun 11.
Article in English | MEDLINE | ID: mdl-22689652

ABSTRACT

The extent and temporal characteristics of G protein-coupled receptor (GPCR) signaling are shaped by the regulator of G protein signaling (RGS) proteins, which promote G protein deactivation. With hundreds of GPCRs and dozens of RGS proteins, compartmentalization plays a key role in establishing signaling specificity. However, the molecular details and mechanisms of this process are poorly understood. In this paper, we report that the R7 group of RGS regulators is controlled by interaction with two previously uncharacterized orphan GPCRs: GPR158 and GPR179. We show that GPR158/179 recruited RGS complexes to the plasma membrane and augmented their ability to regulate GPCR signaling. The loss of GPR179 in a mouse model of night blindness prevented targeting of RGS to the postsynaptic compartment of bipolar neurons in the retina, illuminating the role of GPR179 in night vision. We propose that the interaction of RGS proteins with orphan GPCRs promotes signaling selectivity in G protein pathways.


Subject(s)
GTP-Binding Proteins/metabolism , RGS Proteins/metabolism , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Animals , Cell Communication , Cell Membrane/metabolism , Disease Models, Animal , HEK293 Cells , Humans , Immunoprecipitation , Mass Spectrometry , Mice , Microscopy, Confocal , Neurons/metabolism , Receptors, G-Protein-Coupled/metabolism , Transfection
14.
Am J Hum Genet ; 90(2): 331-9, 2012 Feb 10.
Article in English | MEDLINE | ID: mdl-22325362

ABSTRACT

Complete congenital stationary night blindness (cCSNB) is a clinically and genetically heterogeneous group of retinal disorders characterized by nonprogressive impairment of night vision, absence of the electroretinogram (ERG) b-wave, and variable degrees of involvement of other visual functions. We report here that mutations in GPR179, encoding an orphan G protein receptor, underlie a form of autosomal-recessive cCSNB. The Gpr179(nob5/nob5) mouse model was initially discovered by the absence of the ERG b-wave, a component that reflects depolarizing bipolar cell (DBC) function. We performed genetic mapping, followed by next-generation sequencing of the critical region and detected a large transposon-like DNA insertion in Gpr179. The involvement of GPR179 in DBC function was confirmed in zebrafish and humans. Functional knockdown of gpr179 in zebrafish led to a marked reduction in the amplitude of the ERG b-wave. Candidate gene analysis of GPR179 in DNA extracted from patients with cCSNB identified GPR179-inactivating mutations in two patients. We developed an antibody against mouse GPR179, which robustly labeled DBC dendritic terminals in wild-type mice. This labeling colocalized with the expression of GRM6 and was absent in Gpr179(nob5/nob5) mutant mice. Our results demonstrate that GPR179 plays a critical role in DBC signal transduction and expands our understanding of the mechanisms that mediate normal rod vision.


Subject(s)
Mutation , Myopia/genetics , Myopia/physiopathology , Night Blindness/genetics , Night Blindness/physiopathology , Receptors, G-Protein-Coupled/genetics , Retinal Bipolar Cells/metabolism , Retinal Bipolar Cells/physiology , Animals , Chromosome Mapping/methods , Dark Adaptation/genetics , Electroretinography/methods , Eye Diseases, Hereditary , Gene Knockdown Techniques/methods , Genetic Diseases, X-Linked , Heterozygote , Humans , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Myopia/metabolism , Night Blindness/metabolism , Pedigree , Receptors, Metabotropic Glutamate/genetics , Retinal Rod Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/physiology , Signal Transduction , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL