Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 97
1.
J Neurochem ; 2024 Jan 09.
Article En | MEDLINE | ID: mdl-38196269

Arrestins were discovered for their role in homologous desensitization of G-protein-coupled receptors (GPCRs). Later non-visual arrestins were shown to regulate several signaling pathways. Some of these pathways require arrestin binding to GPCRs, the regulation of others is receptor independent. Here, we demonstrate that arrestin-3 binds the E3 ubiquitin ligase parkin via multiple sites, preferentially interacting with its RING0 domain. Identification of the parkin domains involved suggests that arrestin-3 likely relieves parkin autoinhibition and/or stabilizes the enzymatically active "open" conformation of parkin. Arrestin-3 binding enhances ubiquitination by parkin of the mitochondrial protein mitofusin-1 and facilitates parkin-mediated mitophagy in HeLa cells. Furthermore, arrestin-3 and its mutant with enhanced parkin binding rescue mitofusin-1 ubiquitination and mitophagy in the presence of the Parkinson's disease-associated R275W parkin mutant, which is defective in both functions. Thus, modulation of parkin activity via arrestin-3 might be a novel strategy of anti-parkinsonian therapy.

2.
bioRxiv ; 2023 Oct 30.
Article En | MEDLINE | ID: mdl-37961199

In rodents with unilateral ablation of the substantia nigra neurons supplying dopamine to the striatum, chronic treatment with the dopamine precursor L-DOPA or dopamine agonists induces a progressive increase of behavioral responses, a process known as behavioral sensitization. The sensitization is blunted in arrestin-3 knockout mice. Using virus-mediated gene delivery to the dopamine-depleted striatum of arrestin-3 knockout mice, we found that the restoration of arrestin-3 fully rescued behavioral sensitization, whereas its mutant defective in JNK activation did not. A 25-residue arrestin-3-derived peptide that facilitates JNK3 activation in cells, expressed ubiquitously or selectively in the direct pathway striatal neurons, fully rescued sensitization, whereas an inactive homologous arrestin-2-derived peptide did not. Behavioral rescue was accompanied by the restoration of JNK3 activity and of JNK-dependent phosphorylation of the transcription factor c-Jun in the dopamine-depleted striatum. Thus, arrestin-3-dependent JNK3 activation in direct pathway neurons is a critical element of the molecular mechanism underlying sensitization.

3.
Curr Protoc ; 3(9): e839, 2023 Sep.
Article En | MEDLINE | ID: mdl-37668419

Only 1 out of 4 mammalian arrestin subtypes, arrestin-3, facilitates the activation of c-Jun N-terminal kinase (JNK) family kinases. Here, we describe two different sets of protocols used for elucidating the mechanisms involved. One is based on reconstitution of signaling modules from the following purified proteins: arrestin-3, MKK4, MKK7, JNK1, JNK2, and JNK3. The main advantage of this method is that it unambiguously establishes which effects are direct because only intended purified proteins are present in these assays. The key drawback is that the upstream-most kinases of these cascades, ASK1 or other MAP3Ks, are not available in purified form, limiting reconstitution to incomplete two-kinase modules. The other approach is used for analyzing the effects of arrestin-3 on JNK activation in intact cells. In this case, signaling modules include ASK1 and/or other MAP3Ks. However, as every cell expresses thousands of different proteins, their possible effects on the readout cannot be excluded. Nonetheless, the combination of in vitro reconstitution from purified proteins and cell-based assays makes it possible to elucidate the mechanisms of arrestin-3-dependent activation of JNK family kinases. © 2023 Wiley Periodicals LLC. Basic Protocol 1: Construction of arrestin-3-scaffolded MKK4/7-JNK1/2/3 signaling modules in vitro using purified proteins Alternate Protocol 1: Characterization of arrestin-3-mediated JNK1/2 activation by MKK4/7 by measurement of JNK1/2 phosphorylation using immunoblotting with anti-phospho-JNK antibody Support Protocol 1: Expression, purification, and activation of GST-MKK4 Support Protocol 2: Expression, purification, and activation of GST-MKK7-His6 Support Protocol 3: Expression, purification, and activation of tagless JNK1Α1 Support Protocol 4: Expression, purification, and activation of tagless JNK2Α2 Basic Protocol 2: Analysis of the role of arrestin-3 in ASK1/MKK4/MKK7-induced JNK activation in intact cells Alternate Protocol 2: Analysis of the role of arrestin-3 in MKK4-induced JNK activation in intact cells Basic Protocol 3: Characterization of the biphasic effect of arrestin-3 on ASK1/MKK7-stimulated JNK phosphorylation in cells.


JNK Mitogen-Activated Protein Kinases , Protein Processing, Post-Translational , Animals , Phosphorylation , beta-Arrestin 2 , Arrestins , MAP Kinase Kinase 4 , beta-Arrestin 1/genetics , Mammals
4.
Cells ; 12(12)2023 06 06.
Article En | MEDLINE | ID: mdl-37371033

Arrestins bind active phosphorylated G protein-coupled receptors (GPCRs). Among the four mammalian subtypes, only arrestin-3 facilitates the activation of JNK3 in cells. In available structures, Lys-295 in the lariat loop of arrestin-3 and its homologue Lys-294 in arrestin-2 directly interact with the activator-attached phosphates. We compared the roles of arrestin-3 conformational equilibrium and Lys-295 in GPCR binding and JNK3 activation. Several mutants with enhanced ability to bind GPCRs showed much lower activity towards JNK3, whereas a mutant that does not bind GPCRs was more active. The subcellular distribution of mutants did not correlate with GPCR recruitment or JNK3 activation. Charge neutralization and reversal mutations of Lys-295 differentially affected receptor binding on different backgrounds but had virtually no effect on JNK3 activation. Thus, GPCR binding and arrestin-3-assisted JNK3 activation have distinct structural requirements, suggesting that facilitation of JNK3 activation is the function of arrestin-3 that is not bound to a GPCR.


Arrestins , Receptors, G-Protein-Coupled , Animals , beta-Arrestin 2/metabolism , Phosphorylation/physiology , Arrestins/metabolism , Receptors, G-Protein-Coupled/metabolism , Protein Binding/physiology , Mammals/metabolism
5.
Curr Protoc ; 3(6): e821, 2023 Jun.
Article En | MEDLINE | ID: mdl-37367499

Arrestins were first discovered as proteins that selectively bind active phosphorylated GPCRs and suppress (arrest) their G protein-mediated signaling. Nonvisual arrestins are also recognized as signaling proteins regulating a variety of cellular pathways. Arrestins are highly flexible; they can assume many different conformations. In their receptor-bound conformation, arrestins have higher affinity for a subset of binding partners. This explains how receptor activation regulates certain branches of arrestin-dependent signaling via arrestin recruitment to GPCRs. However, free arrestins are also active molecular entities that regulate other signaling pathways and localize signaling proteins to particular subcellular compartments. Recent findings suggest that the two visuals, arrestin-1 and arrestin-4, which are expressed in photoreceptor cells, not only regulate signaling via binding to photopigments but also interact with several nonreceptor partners, critically affecting the health and survival of photoreceptor cells. Detailed in this overview are GPCR-dependent and independent modes of arrestin-mediated regulation of cellular signaling. © 2023 Wiley Periodicals LLC.


Arrestin , Signal Transduction , Arrestin/metabolism , Signal Transduction/physiology , Arrestins/chemistry , Arrestins/metabolism , GTP-Binding Proteins/metabolism
6.
Int J Mol Sci ; 24(10)2023 May 17.
Article En | MEDLINE | ID: mdl-37240250

Arrestin-1, or visual arrestin, exhibits an exquisite selectivity for light-activated phosphorylated rhodopsin (P-Rh*) over its other functional forms. That selectivity is believed to be mediated by two well-established structural elements in the arrestin-1 molecule, the activation sensor detecting the active conformation of rhodopsin and the phosphorylation sensor responsive to the rhodopsin phosphorylation, which only active phosphorylated rhodopsin can engage simultaneously. However, in the crystal structure of the arrestin-1-rhodopsin complex there are arrestin-1 residues located close to rhodopsin, which do not belong to either sensor. Here we tested by site-directed mutagenesis the functional role of these residues in wild type arrestin-1 using a direct binding assay to P-Rh* and light-activated unphosphorylated rhodopsin (Rh*). We found that many mutations either enhanced the binding only to Rh* or increased the binding to Rh* much more than to P-Rh*. The data suggest that the native residues in these positions act as binding suppressors, specifically inhibiting the arrestin-1 binding to Rh* and thereby increasing arrestin-1 selectivity for P-Rh*. This calls for the modification of a widely accepted model of the arrestin-receptor interactions.


Arrestin , Rhodopsin , Rhodopsin/genetics , Rhodopsin/metabolism , Arrestin/metabolism , Mutation , Phosphorylation , Protein Binding
7.
bioRxiv ; 2023 May 01.
Article En | MEDLINE | ID: mdl-37205393

Arrestins bind active phosphorylated G protein-coupled receptors (GPCRs). Among the four mammalian subtypes, only arrestin-3 facilitates the activation of JNK3 in cells. In available structures, Lys-295 in the lariat loop of arrestin-3 and its homologue Lys-294 in arrestin-2 directly interact with the activator-attached phosphates. We compared the role of arrestin-3 conformational equilibrium and of Lys-295 in GPCR binding and JNK3 activation. Several mutants with enhanced ability to bind GPCRs showed much lower activity towards JNK3, whereas a mutant that does not bind GPCRs was more active. Subcellular distribution of mutants did not correlate with GPCR recruitment or JNK3 activation. Charge neutralization and reversal mutations of Lys-295 differentially affected receptor binding on different backgrounds, but had virtually no effect on JNK3 activation. Thus, GPCR binding and arrestin-3-assisted JNK3 activation have distinct structural requirements, suggesting that facilitation of JNK3 activation is the function of arrestin-3 that is not bound to a GPCR.

8.
Pharmacol Rev ; 75(5): 854-884, 2023 09.
Article En | MEDLINE | ID: mdl-37028945

The two ß-arrestins, ß-arrestin-1 and -2 (systematic names: arrestin-2 and -3, respectively), are multifunctional intracellular proteins that regulate the activity of a very large number of cellular signaling pathways and physiologic functions. The two proteins were discovered for their ability to disrupt signaling via G protein-coupled receptors (GPCRs) via binding to the activated receptors. However, it is now well recognized that both ß-arrestins can also act as direct modulators of numerous cellular processes via either GPCR-dependent or -independent mechanisms. Recent structural, biophysical, and biochemical studies have provided novel insights into how ß-arrestins bind to activated GPCRs and downstream effector proteins. Studies with ß-arrestin mutant mice have identified numerous physiologic and pathophysiological processes regulated by ß-arrestin-1 and/or -2. Following a short summary of recent structural studies, this review primarily focuses on ß-arrestin-regulated physiologic functions, with particular focus on the central nervous system and the roles of ß-arrestins in carcinogenesis and key metabolic processes including the maintenance of glucose and energy homeostasis. This review also highlights potential therapeutic implications of these studies and discusses strategies that could prove useful for targeting specific ß-arrestin-regulated signaling pathways for therapeutic purposes. SIGNIFICANCE STATEMENT: The two ß-arrestins, structurally closely related intracellular proteins that are evolutionarily highly conserved, have emerged as multifunctional proteins able to regulate a vast array of cellular and physiological functions. The outcome of studies with ß-arrestin mutant mice and cultured cells, complemented by novel insights into ß-arrestin structure and function, should pave the way for the development of novel classes of therapeutically useful drugs capable of regulating specific ß-arrestin functions.


Arrestins , Signal Transduction , Mice , Animals , beta-Arrestins/metabolism , Arrestins/chemistry , Arrestins/metabolism , Receptors, G-Protein-Coupled/metabolism , beta-Arrestin 1/metabolism
9.
Sci Signal ; 16(778): eadg9504, 2023 03 28.
Article En | MEDLINE | ID: mdl-36976864

cAMP signaling in the nucleus leads to the expression of immediate early genes in neurons and learning and memory. In this issue of Science Signaling, Martinez et al. found that activation of the ß2-adrenergic receptor enhances nuclear cAMP signaling that supports learning and memory in mice by removing the phosphodiesterase PDE4D5 from the nucleus through arrestin3 bound to the internalized receptor.


Cyclic Nucleotide Phosphodiesterases, Type 4 , Signal Transduction , Mice , Animals , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Cell Nucleus/metabolism , beta-Arrestin 2/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism
10.
Curr Top Behav Neurosci ; 60: 29-45, 2023.
Article En | MEDLINE | ID: mdl-35505061

When the rat D3 dopamine receptor (D3R) was cloned and the distribution of its mRNA examined in 1990-1991, it attracted attention due to its peculiar distribution in the brain quite different from that of its closest relative, the D2 receptor. In the rat brain, the D3R mRNA is enriched in the limbic striatum as opposed to the D2 receptor, which is highly expressed in the motor striatal areas. Later studies in the primate and human brain confirmed relative enrichment of the D3R in the limbic striatum but also demonstrated higher abundance of the D3R in the primate as compared to the rodent brain. Additionally, in the rodent brain, the D3R in the dorsal striatum appears to be co-expressed with the D1 dopamine receptor-bearing striatal neurons giving rise to the direct output striatal pathway, although the picture is less clear with respect to the nucleus accumbens. In contrast, in the primate striatum, the D3R co-localizes with the D2 receptor throughout the basal ganglia as well as in extrastriatal brain areas. The relative abundance of the D3R in the limbic striatum, its output structures, secondary targets, and some of the other connected limbic territories may underpin its role in reward, drug dependence, and impulse control. Selective expression of D3R in the brain proliferative areas may point to its important role in the neural development as well as in neurodevelopmental abnormalities associated with schizophrenia and other developmental brain disorders.


Receptors, Dopamine D2 , Receptors, Dopamine D3 , Rats , Humans , Animals , Receptors, Dopamine D3/genetics , Receptors, Dopamine D3/metabolism , Receptors, Dopamine D2/genetics , Receptors, Dopamine D2/metabolism , Brain/metabolism , Dopamine , RNA, Messenger/metabolism
11.
Int J Mol Sci ; 23(22)2022 Nov 11.
Article En | MEDLINE | ID: mdl-36430370

Arrestins preferentially bind active phosphorylated G protein-coupled receptors (GPCRs). The middle loop, highly conserved in all arrestin subtypes, is localized in the central crest on the GPCR-binding side. Upon receptor binding, it directly interacts with bound GPCR and demonstrates the largest movement of any arrestin element in the structures of the complexes. Comprehensive mutagenesis of the middle loop of rhodopsin-specific arrestin-1 suggests that it primarily serves as a suppressor of binding to non-preferred forms of the receptor. Several mutations in the middle loop increase the binding to unphosphorylated light-activated rhodopsin severalfold, which makes them candidates for improving enhanced phosphorylation-independent arrestins. The data also suggest that enhanced forms of arrestin do not bind GPCRs exactly like the wild-type protein. Thus, the structures of the arrestin-receptor complexes, in all of which different enhanced arrestin mutants and reengineered receptors were used, must be interpreted with caution.


Arrestin , Rhodopsin , Arrestin/metabolism , Rhodopsin/metabolism , Arrestins/metabolism , Receptors, G-Protein-Coupled/metabolism , Protein Binding
12.
Int J Mol Sci ; 23(15)2022 Aug 04.
Article En | MEDLINE | ID: mdl-35955810

Arrestins were first discovered as suppressors of G protein-mediated signaling by G protein-coupled receptors. It was later demonstrated that arrestins also initiate several signaling branches, including mitogen-activated protein kinase cascades. Arrestin-3-dependent activation of the JNK family can be recapitulated with peptide fragments, which are monofunctional elements distilled from this multi-functional arrestin protein. Here, we use maltose-binding protein fusions of arrestin-3-derived peptides to identify arrestin elements that bind kinases of the ASK1-MKK4/7-JNK3 cascade and the shortest peptide facilitating JNK signaling. We identified a 16-residue arrestin-3-derived peptide expressed as a Venus fusion that leads to activation of JNK3α2 in cells. The strength of the binding to the kinases does not correlate with peptide activity. The ASK1-MKK4/7-JNK3 cascade has been implicated in neuronal apoptosis. While inhibitors of MAP kinases exist, short peptides are the first small molecule tools that can activate MAP kinases.


Arrestin , Mitogen-Activated Protein Kinase 10 , Arrestin/metabolism , Arrestins/metabolism , Mitogen-Activated Protein Kinase 10/metabolism , Peptides/metabolism , Peptides/pharmacology , Phosphorylation/physiology , Protein Binding/physiology , beta-Arrestin 2/metabolism , beta-Arrestins/metabolism
13.
Int J Mol Sci ; 23(13)2022 Jun 29.
Article En | MEDLINE | ID: mdl-35806256

Three out of four subtypes of arrestin proteins expressed in mammals self-associate, each forming oligomers of a distinct kind. Monomers and oligomers have different subcellular localization and distinct biological functions. Here we summarize existing evidence regarding arrestin oligomerization and discuss specific functions of monomeric and oligomeric forms, although too few of the latter are known. The data on arrestins highlight biological importance of oligomerization of signaling proteins. Distinct modes of oligomerization might be an important contributing factor to the functional differences among highly homologous members of the arrestin protein family.


Arrestin , Arrestins , Animals , Arrestin/genetics , Arrestin/metabolism , Arrestins/metabolism , Mammals/metabolism , beta-Arrestins/metabolism
14.
Trends Biochem Sci ; 47(7): 570-581, 2022 07.
Article En | MEDLINE | ID: mdl-35396120

Three classes of G-protein-coupled receptor (GPCR) partners - G proteins, GPCR kinases, and arrestins - preferentially bind active GPCRs. Our analysis suggests that the structures of GPCRs bound to these interaction partners available today do not reveal a clear conformational basis for signaling bias, which would have enabled the rational design of biased GRCR ligands. In view of this, three possibilities are conceivable: (i) there are no generalizable GPCR conformations conducive to binding a particular type of partner; (ii) subtle differences in the orientation of individual residues and/or their interactions not easily detectable in the receptor-transducer structures determine partner preference; or (iii) the dynamics of GPCR binding to different types of partners rather than the structures of the final complexes might underlie transducer bias.


Arrestins , Receptors, G-Protein-Coupled , Arrestins/chemistry , Arrestins/metabolism , Ligands , Protein Binding , Receptors, G-Protein-Coupled/metabolism , Signal Transduction
16.
Biomolecules ; 11(2)2021 02 04.
Article En | MEDLINE | ID: mdl-33557162

Arrestins are a small family of four proteins in most vertebrates that bind hundreds of different G protein-coupled receptors (GPCRs). Arrestin binding to a GPCR has at least three functions: precluding further receptor coupling to G proteins, facilitating receptor internalization, and initiating distinct arrestin-mediated signaling. The molecular mechanism of arrestin-GPCR interactions has been extensively studied and discussed from the "arrestin perspective", focusing on the roles of arrestin elements in receptor binding. Here, we discuss this phenomenon from the "receptor perspective", focusing on the receptor elements involved in arrestin binding and emphasizing existing gaps in our knowledge that need to be filled. It is vitally important to understand the role of receptor elements in arrestin activation and how the interaction of each of these elements with arrestin contributes to the latter's transition to the high-affinity binding state. A more precise knowledge of the molecular mechanisms of arrestin activation is needed to enable the construction of arrestin mutants with desired functional characteristics.


Arrestin/metabolism , Arrestins/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Binding Sites , Cattle , Crystallography, X-Ray , Fishes , Humans , Hydrophobic and Hydrophilic Interactions , Mice , Models, Molecular , Mutation , Phosphorylation , Protein Binding , Protein Conformation , Protein Domains , Protein Isoforms , Signal Transduction/physiology
17.
J Neurochem ; 156(4): 435-444, 2021 02.
Article En | MEDLINE | ID: mdl-32594524

Arrestins demonstrate strong preference for phosphorylated over unphosphorylated receptors, but how arrestins "sense" receptor phosphorylation is unclear. A conserved lysine in the lariat loop of arrestins directly binds the phosphate in crystal structures of activated arrestin-1, -2, and -3. The lariat loop supplies two negative charges to the central polar core, which must be disrupted for arrestin activation and high-affinity receptor binding. Therefore, we hypothesized that receptor-attached phosphates pull the lariat loop via this lysine, thus removing the negative charges and destabilizing the polar core. We tested the role of this lysine by introducing charge elimination (Lys->Ala) and reversal (Lys->Glu) mutations in arrestin-1, -2, and -3. These mutations in arrestin-1 only moderately reduced phospho-rhodopsin binding and had no detectable effect on arrestin-2 and -3 binding to cognate non-visual receptors in cells. The mutations of Lys300 in bovine and homologous Lys301 in mouse arrestin-1 on the background of pre-activated mutants had variable effects on the binding to light-activated phosphorylated rhodopsin, while affecting the binding to unphosphorylated rhodopsin to a greater extent. Thus, conserved lysine in the lariat loop participates in receptor binding, but does not play a critical role in phosphate-induced arrestin activation.


Arrestins/metabolism , Biosensing Techniques/methods , Lysine/metabolism , Phosphates/metabolism , Animals , Arrestins/chemistry , Binding Sites/physiology , Cattle , Lysine/chemistry , Mice , Phosphates/chemistry , Protein Binding/physiology , Protein Structure, Secondary
18.
J Neurochem ; 157(4): 1138-1152, 2021 05.
Article En | MEDLINE | ID: mdl-33159335

The finger loop in the central crest of the receptor-binding site of arrestins engages the cavity between the transmembrane helices of activated G-protein-coupled receptors. Therefore, it was hypothesized to serve as the sensor that detects the activation state of the receptor. We performed comprehensive mutagenesis of the finger loop in bovine visual arrestin-1, generated mutant radiolabeled proteins by cell-free translation, and determined the effects of mutations on the in vitro binding of arrestin-1 to purified phosphorylated light-activated rhodopsin. This interaction is driven by two factors, rhodopsin activation and rhodopsin-attached phosphates. Therefore, the binding of arrestin-1 to light-activated unphosphorylated rhodopsin is low. To evaluate the role of the finger loop specifically in the recognition of the active receptor conformation, we tested the effects of these mutations in the context of truncated arrestin-1 that demonstrates much higher binding to unphosphorylated activated and phosphorylated inactive rhodopsin. The majority of finger loop residues proved important for arrestin-1 binding to light-activated rhodopsin, with six mutations affecting the binding exclusively to this form. Thus, the finger loop is the key element of arrestin-1 activation sensor. The data also suggest that arrestin-1 and its enhanced mutant bind various functional forms of rhodopsin differently.


Arrestin/chemistry , Arrestin/metabolism , Protein Structure, Secondary/physiology , Animals , Binding Sites , Cattle , Protein Binding
20.
J Neurosci ; 40(42): 8055-8069, 2020 10 14.
Article En | MEDLINE | ID: mdl-32948676

Members of the arrestin superfamily have great propensity of self-association, but the physiological significance of this phenomenon is unclear. To determine the biological role of visual arrestin-1 oligomerization in rod photoreceptors, we expressed mutant arrestin-1 with severely impaired self-association in mouse rods and analyzed mice of both sexes. We show that the oligomerization-deficient mutant is capable of quenching rhodopsin signaling normally, as judged by electroretinography and single-cell recording. Like wild type, mutant arrestin-1 is largely excluded from the outer segments in the dark, proving that the normal intracellular localization is not due the size exclusion of arrestin-1 oligomers. In contrast to wild type, supraphysiological expression of the mutant causes shortening of the outer segments and photoreceptor death. Thus, oligomerization reduces the cytotoxicity of arrestin-1 monomer, ensuring long-term photoreceptor survival.SIGNIFICANCE STATEMENT Visual arrestin-1 forms dimers and tetramers. The biological role of its oligomerization is unclear. To test the role of arrestin-1 self-association, we expressed oligomerization-deficient mutant in arrestin-1 knock-out mice. The mutant quenches light-induced rhodopsin signaling like wild type, demonstrating that in vivo monomeric arrestin-1 is necessary and sufficient for this function. In rods, arrestin-1 moves from the inner segments and cell bodies in the dark to the outer segments in the light. Nonoligomerizing mutant undergoes the same translocation, demonstrating that the size of the oligomers is not the reason for arrestin-1 exclusion from the outer segments in the dark. High expression of oligomerization-deficient arrestin-1 resulted in rod death. Thus, oligomerization reduces the cytotoxicity of high levels of arrestin-1 monomer.


Arrestins/metabolism , Arrestins/physiology , Adaptation, Ocular , Animals , Arrestins/genetics , Cell Survival , Electroretinography , Female , Light Signal Transduction , Male , Mice , Mice, Knockout , Mice, Transgenic , Mutation/genetics , Retina/anatomy & histology , Retina/growth & development , Retinal Rod Photoreceptor Cells/metabolism , Rhodopsin/physiology
...