RCVRN Mouse

Recoverin Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

Gene Structure and Expression

RCVRN in mice encodes a protein with three EF-hand calcium-binding domains and a myristoylated N-terminal region. Key features include:

ParameterDescription
Genomic LocationChromosome 11 (NC_000077.7)
Protein Structure23 kDa, myristoylated N-terminus, four EF-hand motifs
Expression PatternPrimarily in rod and cone photoreceptors; translocates to synaptic terminals under light

Functional Domains:

  • Calcium Binding: Regulates rhodopsin kinase (GRK1) activity via calcium-dependent conformational changes .

  • Membrane Interactions: Myristoyl group facilitates interaction with GRK1 in photoreceptor outer segments .

Functional Role in Photoreceptors

RCVRN modulates light adaptation by controlling rhodopsin phosphorylation and response recovery.

Key Mechanisms:

  1. Light Adaptation: Inhibits GRK1 at high calcium levels (darkness), prolonging metarhodopsin II (Rh*) lifetime and rod sensitivity .

  2. Response Dynamics:

    • Wild-Type: Slower response recovery under continuous light, enabling detection of weak stimuli .

    • Knockout (Rcvrn⁻/⁻): Accelerated Rh* inactivation and reduced sensitivity to sustained light .

Knockout Models and Phenotypic Observations

Genetic ablation of RCVRN in mice reveals critical roles in photoreceptor physiology.

ParameterWild-TypeRcvrn⁻/⁻Source
Rh Lifetime*Prolonged under high Ca²⁺Reduced by ~50%
Response RecoverySlower under continuous lightFaster recovery
Sensitivity to LightMaintained under steady lightReduced sensitivity
Outer Segment StructureIntactNo reported structural defects

Clinical Relevance:

  • Cancer-Associated Retinopathy (CAR): Recoverin is a primary antigen in CAR, a paraneoplastic syndrome causing photoreceptor degeneration . Mouse models (e.g., R64 peptide) mimic CAR pathology, enabling therapeutic testing .

Gene-Environmental Interactions

RCVRN expression is influenced by environmental and chemical factors, though data primarily derive from rat studies.

ChemicalEffect on RCVRN ExpressionMechanismSource
Bisphenol A↓ mRNA expressionEpigenetic modifications
Copper Deficiency↑ mRNA expressionOxidative stress response
Aflatoxin B1↑ Gene methylationDNA damage

Note: Rat-specific data; cross-species extrapolation requires validation.

Aging and Retinal Health

Aging mouse models show RCVRN-related declines in photoreceptor function.

Key Findings:

  • Aging RPE: ↓ Expression of visual cycle genes (e.g., Rpe65, Rdh8) and ↑ inflammatory markers (e.g., C3, Cxcr1) .

  • Photoreceptor Senescence: Reduced recoverin expression correlates with impaired light adaptation in aged mice .

Research Applications

RCVRN mouse models are pivotal for studying retinal diseases and photoreceptor development.

ApplicationMethodologyOutcomeSource
Retinal Organoids3D culture of hiPSCsRCVRN+ photoreceptors labeled via eGFP; outer segment-like structures
CAR TherapeuticsCTLA4 pathway modulationReduced retinal dysfunction in peptide models
Gene EditingCRISPR/Cas9 knockoutsFunctional assays for phototransduction defects

Product Specs

Introduction
Recoverin, a member of the neuronal calcium sensor family, is a 23kDa protein found in retinal photoreceptor cells. This protein, involved in intracellular signal transduction, exhibits heterogeneous acylation and calcium-binding properties. Its structure consists of four EF-hands, two of which bind to calcium ions. Upon calcium binding, the acyl group is extruded from a hydrophobic cleft within the protein. This extrusion triggers the translocation of recoverin from its soluble form to the disc membrane. Functionally, recoverin is involved in regulating the phototransduction cascade's termination in the retina. It achieves this by inhibiting the phosphorylation of photo-activated rhodopsin. Recoverin's role in inhibiting rhodopsin kinase, an enzyme that regulates rhodopsin phosphorylation, is crucial. By modulating rhodopsin phosphorylation, recoverin influences the eye's ability to adapt to light exposure and recover from it. In clinical contexts, recoverin serves as a detectable serological protein marker. Its presence is observed in patients diagnosed with cancer-associated retinopathy, a paraneoplastic syndrome.
Description
Recombinant Recoverin Mouse, produced in E. coli, is a single, non-glycosylated polypeptide chain. This protein comprises 225 amino acids (1-202a.a.) with a molecular weight of 25.8 kDa. The recombinant protein is engineered with a 23 amino acid His-tag fused at its N-terminus. Purification is achieved through proprietary chromatographic techniques.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The protein is supplied at a concentration of 1mg/ml in a buffer consisting of Phosphate Buffered Saline (pH 7.4), 10% glycerol, and 1mM DTT.
Stability
For short-term storage (up to 4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to store the product frozen at -20°C. To ensure long-term stability, adding a carrier protein like HSA or BSA (0.1%) is advisable. It's important to avoid repeated freeze-thaw cycles.
Purity
The purity of the protein is determined to be greater than 90% based on SDS-PAGE analysis.
Synonyms

RCV1, Cancer-associated retinopathy protein, Protein CAR, RCVRN, Recoverin, S-modulin.

Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MGSMGNSKSG ALSKEILEEL QLNTKFTEEE LSAWYQSFLK ECPSGRITRQ EFESIYSKFF PDSDPKAYAQ HVFRSFDANS DGTLDFKEYV IALHMTTAGK PTQKLEWAFS LYDVDGNGTI SKNEVLEIVM AIFKMIKPED VKLLPDDENT PEKRAEKIWA FFGKKEDDKL TEEEFIEGTL ANKEILRLIQ FEPQKVKERI KEKKQ.

Q&A

What is RCVRN and why is it significant in mouse retinal research?

RCVRN (recoverin) is a well-established pan-photoreceptor marker protein in the mammalian retina that has become increasingly important in retinal research. In the human fetal retina, recoverin expression begins in photoreceptor precursors around 13 fetal weeks and subsequently becomes widely expressed in both cone and rod photoreceptors throughout development . In the adult retina, recoverin primarily localizes to the photoreceptor outer segment where it plays a crucial role in the phototransduction cascade .

The significance of RCVRN in mouse retinal research stems from its consistent expression across nearly all developmental stages and photoreceptor subtypes. This comprehensive expression pattern makes RCVRN an ideal candidate for developing reporter systems to monitor photoreceptor development, function, and degeneration. Mouse models with RCVRN modifications allow researchers to visualize, track, and isolate photoreceptors for detailed molecular and functional analyses.

How does RCVRN expression change during mouse retinal development?

RCVRN expression follows a specific temporal pattern during mouse retinal development that parallels human development. Expression begins in early photoreceptor precursors and continues throughout maturation of both rod and cone photoreceptors. Developmental tracking studies using RCVRN-reporter mouse models have demonstrated that RCVRN expression steadily increases as photoreceptors mature.

In mouse retinal organoid (RO) models derived from RCVRN-eGFP reporter lines, researchers can dynamically monitor photoreceptor development in live cultures. The RCVRN-eGFP specifically and steadily labels photoreceptor cells from early photoreceptor precursors through to mature rods and cones . This expression pattern makes RCVRN an excellent developmental marker for tracking photoreceptor lineage in both normal development and disease models.

What are the methodological approaches for generating RCVRN mouse models?

Generating RCVRN mouse models typically involves CRISPR/Cas9 genome editing techniques to insert reporter sequences at the endogenous RCVRN locus. A methodical approach includes:

  • Guide RNA design: Selection of a guide RNA targeting the RCVRN gene with high cleavage activity (>50% predicted efficiency). The sgRNA is cloned into a vector such as pSpCas9 (BB)-2A-Puro (PX459) .

  • Targeting plasmid construction: Creation of a targeting plasmid containing reporter sequences (such as P2A-eGFP) flanked by homology arms corresponding to the RCVRN locus .

  • Transfection: Electroporation of guide-carrying plasmids and targeting plasmids into mouse embryonic stem cells or zygotes using optimized parameters (e.g., 1,100 v; 10 ms; 3 pulses as described in some protocols) .

  • Selection: Treatment with puromycin (typically 0.4 μg/ml for 7 days) to select resistant clones .

  • Validation: Confirmation of correct targeting through PCR and Sanger sequencing to ensure in-frame integration of the reporter sequence .

  • Chimera generation: Injection of correctly targeted cells into blastocysts and implantation into pseudopregnant females.

  • Germline transmission: Breeding of chimeric offspring to establish stable RCVRN reporter mouse lines.

This methodical approach ensures precise genetic modification while maintaining the endogenous expression pattern of RCVRN.

What are the primary applications of RCVRN mouse models in retinal research?

RCVRN mouse models serve multiple crucial applications in retinal research:

  • Developmental studies: Tracking photoreceptor genesis, migration, and maturation during retinal development.

  • Cell isolation: FACS-based isolation of RCVRN-expressing photoreceptors for downstream applications including transcriptomic and proteomic analyses. For example, researchers have used RCVRN-eGFP reporter lines to separate eGFP-positive photoreceptors from retinal organoids at day 150 of differentiation .

  • Disease modeling: Investigating photoreceptor degeneration in models of retinal dystrophies by monitoring changes in RCVRN-expressing cells over time. Studies have revealed significant declines in gene expression of photoreceptor markers including RCVRN in Rho−/−OPN1-GFP mice, a model of rod-cone dystrophy .

  • Therapeutic development: Evaluating the efficacy of potential treatments by assessing their effects on RCVRN-expressing photoreceptor survival and function.

  • Transcriptome analysis: Establishing reference databases of gene expression profiles in RCVRN-positive photoreceptors, as demonstrated by RNA sequencing studies of FACS-sorted eGFP+ cells from RCVRN-eGFP reporter retinal organoids .

How can researchers optimize FACS protocols for isolating RCVRN-expressing photoreceptors?

Optimizing FACS protocols for isolating RCVRN-expressing photoreceptors requires careful attention to several methodological factors:

  • Tissue dissociation: Mechanically separate retinal organoids or retinal tissue into small pieces using a needle with 1 ml syringe, followed by enzymatic digestion using papain dissociation systems (e.g., Worthington Biochemical) according to manufacturer's instructions . The dissociation protocol must balance completeness of dissociation against cell viability.

  • Buffer optimization: Resuspend cells in sorting buffer containing PBS with 1 mM EDTA and 2% (vol/vol) FBS to maintain cell integrity during sorting . Optimization of buffer composition based on downstream applications may be necessary.

  • Temperature control: Perform cell sorting at 4°C using flow cytometers such as BD FACSAria III to minimize cellular stress and preserve RNA/protein integrity .

  • Collection parameters: Collect both RCVRN-positive (e.g., eGFP+) and RCVRN-negative (e.G., eGFP-) populations in specialized collecting buffer composed of 50% culture medium and 50% (vol/vol) FBS to maximize post-sort viability .

  • Gating strategies: Develop precise gating parameters based on fluorescence intensity, forward/side scatter properties, and viability staining to ensure pure populations.

  • Validation of sorted populations: Confirm purity of sorted fractions through qPCR for photoreceptor-specific markers and immunocytochemistry.

  • CD marker enrichment: Consider using additional surface markers such as CD133, which has been identified as a potential biomarker for positive selection of photoreceptors in RCVRN-eGFP models .

This optimized protocol yields high-quality photoreceptor isolates suitable for downstream molecular analyses including RNA-seq, proteomics, and functional studies.

What computational approaches can be used to analyze RCVRN-associated gene networks in the mouse retina?

Advanced computational approaches for analyzing RCVRN-associated gene networks in the mouse retina include:

  • Bayesian network analysis: This statistical approach integrates multiple data sources to predict functional associations between genes. As demonstrated in the development of MoReNet (Mouse Retinal Network), Bayesian statistics can be applied to information from gene expression databases, protein-protein interaction networks, and gene ontology annotations to construct comprehensive retinal gene networks .

  • Expression correlation analysis: Analyze correlation patterns across multiple retina-specific microarray datasets to identify genes with expression patterns similar to RCVRN. Important methodological consideration: analyze each dataset separately rather than merging them, as co-expression might be condition-specific and signals in one condition might be overwhelmed by noise in others .

  • Performance evaluation: Use Receiver Operating Characteristic (ROC) curves to evaluate prediction performance, implementing 5-fold cross-validation where datasets are divided into training and testing sets .

  • Independence analysis: Ensure dataset independence by calculating Pearson correlation coefficients (PCCs) of L-values between datasets. Values below 0.15 (as observed in 83% of comparisons in retinal network studies) suggest dataset independence .

  • Functional cluster identification: Apply network clustering algorithms to identify functional modules within the RCVRN-associated network. The MoReNet approach identified five major gene clusters in the retinal network, each enriched for different biological functions .

  • Hub gene identification: Calculate network centrality measures to identify hub genes in the RCVRN-associated network. Approximately 50 hub genes were identified in the mouse retinal network that are predicted to play particularly important roles in retinal function .

How do temporal expression patterns of RCVRN correlate with other photoreceptor genes in development and disease?

Temporal expression analysis of RCVRN with other photoreceptor genes reveals important correlations in both development and disease conditions:

Development correlations:

  • In normal retinal development, RCVRN expression patterns closely align with transcription factors regulating photoreceptor differentiation. The RCVRN-eGFP reporter system shows that recoverin expression faithfully replicates the developmental characteristics of photoreceptors during retinal differentiation .

  • Quantitative analysis of gene expression during retinal organoid differentiation shows coordinated upregulation of RCVRN with other photoreceptor-specific genes including opsins (OPN1SW, OPN1MW), phototransduction components (PDE6H, CNGA3), and photoreceptor-specific arrestin (ARR3) .

Disease correlations:

  • In rod-cone dystrophy models (Rho−/−OPN1-GFP mice), gene expression analysis reveals coordinated temporal changes in multiple photoreceptor genes. While RCVRN expression remains stable in wild-type OPN1-GFP mice over time, Rho−/−OPN1-GFP mice show significant reductions in multiple photoreceptor genes .

  • Temporal expression analysis shows differential decline rates of various genes:

    • CRX declines significantly from baseline starting at postnatal week 6 (PNW6)

    • OPN1SW and CNGA3 decline from baseline by PNW17

    • Multiple genes including OPN1SW, OPN1MW, ARR3, CNGA3, and PDE6H show significant reduction by PNW25

The table below summarizes the temporal pattern of gene expression changes in the Rho−/−OPN1-GFP mouse model compared to wild-type controls:

GeneInitial DeclineSignificant ReductionDifference Between Genotypes
CRXPNW6PNW25Significant at PNW17 (p<0.001) and PNW25 (p<0.001)
OPN1SWPNW17PNW25Significant at PNW17 (p<0.05) and PNW25 (p<0.001)
OPN1MWLaterPNW25Not significant at any time point
ARR3LaterPNW25Significant at PNW25 (p<0.05)
CNGA3PNW17PNW25Not specifically mentioned
PDE6HLaterPNW25Not specifically mentioned

This temporal correlation analysis provides insights into the sequence of molecular events during photoreceptor degeneration and identifies potential early biomarkers of disease progression.

What are the methodological challenges in distinguishing rod versus cone expression of RCVRN in mouse models?

Distinguishing rod versus cone expression of RCVRN in mouse models presents several methodological challenges that require sophisticated experimental approaches:

  • Overlapping expression patterns: RCVRN is expressed in both rod and cone photoreceptors, making simple RCVRN reporter systems insufficient for distinguishing between these cell types. Researchers must implement additional methodologies to differentiate between rod and cone expression.

  • Spatiotemporal resolution: The mouse retina contains approximately 97% rods and only 3% cones, making cone-specific RCVRN expression difficult to analyze without specific enrichment strategies.

  • Single-cell resolution techniques: To address these challenges, researchers can employ:

    • Single-cell RNA sequencing of RCVRN-positive cells to identify rod- versus cone-specific transcriptional signatures

    • Combined fluorescent reporters where RCVRN reporters are used alongside cone-specific (e.g., OPN1SW-driven) or rod-specific (e.g., rhodopsin-driven) reporters

    • Laser capture microdissection approaches similar to those used to isolate photoreceptor, inner nuclear, and ganglion cell layers

  • Dual immunohistochemistry: Co-labeling techniques using antibodies against RCVRN and rod- or cone-specific markers can help distinguish expression patterns at the protein level.

  • Developmental tracking: Monitoring RCVRN expression throughout development in combination with known temporal patterns of rod versus cone development can help distinguish cell type-specific expression.

  • Quantitative analysis in degeneration models: In rod-cone dystrophy models such as Rho−/−OPN1-GFP mice, researchers can assess relative changes in RCVRN expression compared to specific rod markers (e.g., rhodopsin) or cone markers (e.g., OPN1SW, OPN1MW) .

  • Mouse model selection: Choosing appropriate models for study is critical. For instance, the OPN1-GFP mouse model has been used to specifically label and study cone photoreceptors, while Rho−/−OPN1-GFP mice provide a model where rod photoreceptors degenerate while cone markers can still be tracked .

How can RCVRN mouse models contribute to the identification of novel therapies for retinal degenerative diseases?

RCVRN mouse models offer sophisticated platforms for the identification of novel therapies for retinal degenerative diseases through several methodological approaches:

  • High-throughput screening systems: RCVRN-reporter mouse models provide quantifiable readouts for photoreceptor survival and function, enabling:

    • Screening of compound libraries for molecules that preserve RCVRN-expressing cells

    • Evaluation of gene therapy vectors for efficiency in targeting and rescuing photoreceptors

    • Assessment of cell replacement therapies using transplanted RCVRN-positive cells

  • Mechanism-based therapeutic development: Transcriptomic analysis of RCVRN-positive cells from healthy and degenerating retinas identifies:

    • Dysregulated pathways that could be targeted therapeutically

    • Potential neuroprotective factors that preserve photoreceptor function

    • Novel CD biomarkers (such as CD133) that could be utilized for therapeutic cell selection and enrichment

  • Temporal intervention studies: RCVRN mouse models allow examination of therapeutic windows by:

    • Tracking the temporal progression of photoreceptor degeneration

    • Identifying early molecular changes before visible degeneration occurs

    • Determining optimal timing for therapeutic intervention

  • Combinatorial therapy evaluation: Using comprehensive gene network analysis approaches like those applied in MoReNet , researchers can:

    • Identify hub genes in the RCVRN-associated network as primary therapeutic targets

    • Predict potential synergistic therapeutic approaches based on network interactions

    • Use "guilt by association" methods to predict novel gene functions and disease associations

  • Personalized medicine approaches: RCVRN mouse models can be combined with patient-specific mutations to:

    • Create personalized disease models for testing mutation-specific therapies

    • Develop reporter systems in patient-derived iPSCs to monitor treatment responses

    • Validate mutation-specific therapeutic approaches before clinical application

A key methodological advantage of RCVRN mouse models in therapeutic development is their ability to provide quantifiable readouts of photoreceptor health across the entire spectrum of photoreceptor development and degeneration, from early precursors to mature cells .

What are the optimal controls for experiments using RCVRN reporter mouse models?

Designing rigorous controls for experiments using RCVRN reporter mouse models requires careful consideration of several factors:

  • Genetic background controls:

    • Use littermates whenever possible to minimize genetic background variations

    • For CRISPR/Cas9-generated RCVRN reporter mice, include control animals that underwent the same genome editing process but without insertion of reporter sequences

    • For comparative studies, ensure all mouse lines share the same genetic background or backcross for at least 6-10 generations

  • Reporter expression controls:

    • Include immunostaining for endogenous RCVRN protein to confirm faithful recapitulation of expression patterns by the reporter

    • Compare RCVRN-reporter expression with other established photoreceptor markers to validate specificity

    • For FACS experiments, include fluorescence-minus-one (FMO) controls and isotype controls for antibody staining

  • Developmental controls:

    • Establish baseline expression at multiple developmental timepoints (e.g., PNW2, PNW6, PNW17, PNW25) as was done in the Rho−/−OPN1-GFP study

    • Include age-matched controls for all experimental timepoints

    • Consider circadian variations in gene expression by standardizing tissue collection times

  • Technical controls for transcriptome analysis:

    • Include unsorted cells alongside FACS-sorted RCVRN-positive and RCVRN-negative populations

    • Perform technical replicates (at least 2 independent experiments) for RNA sequencing as described in the RCVRN-eGFP reporter hiPSC study

    • Include spike-in controls for RNA-seq normalization

  • Environmental controls:

    • Standardize housing conditions including light cycles, especially important for photoreceptor studies

    • Control for potential environmental factors affecting retinal development (temperature, diet, stress)

  • Statistical controls:

    • Use appropriate statistical methods such as non-linear regression models for analyzing temporal changes in gene expression

    • Ensure sufficient biological replicates for adequate statistical power (minimum n=3 per group)

How can researchers integrate RCVRN mouse data with human retinal disease information?

Integrating RCVRN mouse model data with human retinal disease information requires sophisticated methodological approaches:

  • Comparative transcriptomics:

    • Create parallel datasets of RCVRN-expressing cells from mouse models and human retinal organoids or post-mortem tissues

    • Perform cross-species normalization to identify conserved and divergent gene expression patterns

    • Use ortholog mapping tools to systematically compare mouse and human datasets

  • Network-based disease gene prediction:

    • Utilize interspecies gene networks like MoReNet to identify hub genes conserved between mouse and human retinas

    • Apply Bayesian network approaches to integrate mouse experimental data with human genetic data

    • Use "guilt by association" methods in the retinal network to predict novel disease-associated genes

  • Translational validation approaches:

    • Test findings from RCVRN mouse models in human iPSC-derived retinal organoids

    • Create equivalent RCVRN-reporter systems in human cells to validate mouse findings

    • Compare expression changes in mouse models with patterns observed in human patient samples

  • Cross-species phenotyping:

    • Develop standardized phenotyping protocols that can be applied to both mouse models and human patients

    • Correlate RCVRN expression patterns with functional and structural changes in both species

    • Use advanced imaging techniques (OCT, adaptive optics) that provide comparable data between mouse and human studies

  • Systems biology integration:

    • Incorporate data from RCVRN mouse models into larger systems biology frameworks

    • Use computational approaches to predict how mouse phenotypes might translate to human disease progression

    • Apply machine learning algorithms to identify biomarkers with cross-species predictive value

This integrative approach enhances the translational relevance of RCVRN mouse model findings and accelerates the development of therapies for human retinal diseases.

What are the most reliable methods for quantifying RCVRN expression changes in mouse models?

Reliable quantification of RCVRN expression changes in mouse models requires a multi-modal approach:

  • RT-qPCR analysis:

    • Design primers spanning exon-exon junctions to avoid genomic DNA amplification

    • Use multiple reference genes (at least 3) that are stable across experimental conditions

    • Apply statistical methods like non-linear regression models to analyze temporal changes

    • Include technical triplicates and biological replicates (minimum n=3)

  • RNA sequencing approaches:

    • For bulk RNA-seq, collect cells in TRIzol reagent and assess RNA quality using Agilent 2100 Bioanalyzer

    • For single-cell RNA-seq, optimize dissociation protocols to maintain cell viability and RNA integrity

    • Use spike-in controls for normalization

    • Apply appropriate bioinformatic pipelines for differential expression analysis

  • Protein quantification:

    • Western blot analysis with validated antibodies against RCVRN

    • Quantitative immunohistochemistry with appropriate controls

    • ELISA or other immunoassays for high-throughput protein quantification

    • Consider mass spectrometry-based approaches for unbiased protein quantification

  • Live imaging of reporter expression:

    • For RCVRN-GFP or similar reporter models, establish standardized imaging parameters

    • Use internal controls to normalize for variations in imaging conditions

    • Apply automated image analysis algorithms for unbiased quantification

    • Track individual cells over time when possible to account for cell-to-cell variability

  • Flow cytometry:

    • Standardize dissociation protocols to ensure consistent single-cell suspensions

    • Use consistent gating strategies across experiments

    • Quantify both percentage of RCVRN-positive cells and mean fluorescence intensity

    • Include compensation controls and fluorescence-minus-one controls

Table: Comparison of Methods for RCVRN Expression Quantification

MethodAdvantagesLimitationsBest Application
RT-qPCRHigh sensitivity, quantitative, relatively simpleCannot distinguish cell types in mixed populationsTemporal expression changes, validation of RNA-seq
RNA-seqComprehensive, unbiased, provides contextExpensive, complex analysisGlobal transcriptome analysis, network identification
Protein assaysMeasures functional product, post-translational informationAntibody specificity issues, semi-quantitativeValidation of transcript findings, functional studies
Reporter imagingReal-time, longitudinal, cell-type specificPotential reporter artifacts, qualitativeDevelopmental studies, live cell tracking
Flow cytometrySingle-cell resolution, quantitative, cell isolationRequires tissue dissociation, potential selection biasCell type identification, population analysis

Using multiple complementary approaches provides the most reliable assessment of RCVRN expression changes in mouse models.

How should researchers interpret discrepancies between RCVRN transcript levels and protein expression?

Interpreting discrepancies between RCVRN transcript levels and protein expression requires careful methodological consideration:

  • Temporal delay factors:

    • Protein synthesis and degradation rates may differ from mRNA dynamics

    • Establish time-course experiments to determine the temporal relationship between transcript and protein changes

    • In photoreceptors specifically, consider the specialized protein transport mechanisms to the outer segment that may delay protein localization

  • Methodological considerations:

    • Evaluate sensitivity differences between transcript detection (e.g., qPCR, RNA-seq) and protein detection methods (e.g., Western blot, immunohistochemistry)

    • Employ absolute quantification methods (e.g., digital PCR, quantitative Western blot with recombinant protein standards) to directly compare transcript and protein quantities

    • Consider using ribosome profiling to assess translation efficiency of RCVRN mRNA

  • Regulatory mechanisms:

    • Investigate post-transcriptional regulatory mechanisms including:

      • microRNA-mediated regulation

      • RNA-binding protein interactions

      • Alternative splicing events

    • Assess post-translational modifications that may affect protein stability or antibody detection

    • Consider protein localization changes that might affect detection without changing total expression

  • Cell-type specific analysis:

    • Use single-cell approaches to determine whether discrepancies are due to changes in specific photoreceptor subpopulations

    • Consider the possibility that transcript and protein changes may occur in different cell populations

    • Employ spatial transcriptomics alongside protein imaging to correlate expression patterns at the cellular level

  • Functional validation:

    • Determine whether transcript or protein levels better correlate with functional measures of photoreceptor activity

    • Use reporter systems that reflect different levels of regulation (e.g., transcriptional vs. translational reporters)

    • Consider the biological context of the discrepancy - which measure is more relevant to the specific research question?

A comprehensive approach incorporating these considerations will help researchers accurately interpret RCVRN expression discrepancies and their biological significance.

What novel research directions can be pursued using RCVRN mouse models?

RCVRN mouse models open numerous novel research directions with significant potential impact:

  • Single-cell trajectory mapping:

    • Use RCVRN-reporter mice to isolate photoreceptors at different developmental stages

    • Apply single-cell RNA sequencing to construct developmental trajectories

    • Identify branch points and decision factors in photoreceptor specification

    • Map the molecular events leading to rod versus cone differentiation

  • Circuit integration studies:

    • Combine RCVRN reporters with trans-synaptic tracers to map photoreceptor connectivity

    • Investigate how photoreceptor degeneration affects downstream retinal circuits

    • Develop optogenetic approaches targeting RCVRN-expressing cells to manipulate specific photoreceptor populations

  • Regenerative medicine applications:

    • Use RCVRN reporters to track integration and maturation of transplanted photoreceptor precursors

    • Identify factors that enhance survival and integration of RCVRN-positive cells

    • Develop methods to direct stem cell differentiation toward RCVRN-expressing photoreceptors

    • Explore the potential of identified CD markers like CD133 for clinical applications in cell selection

  • Neuroprotective pathway discovery:

    • Perform comprehensive transcriptomic profiling of RCVRN-positive cells under stress conditions

    • Identify molecular pathways that promote photoreceptor survival

    • Use RCVRN mouse models to test neuroprotective compounds in vivo

    • Explore the role of non-cell-autonomous factors in photoreceptor survival

  • Integrative multi-omics approaches:

    • Combine transcriptomic, proteomic, and metabolomic analyses of RCVRN-expressing cells

    • Construct comprehensive molecular networks using Bayesian approaches

    • Identify key regulatory nodes that could serve as therapeutic targets

    • Develop predictive models of photoreceptor degeneration and response to therapy

  • Comparative evolutionary studies:

    • Compare RCVRN expression and function across species with different visual systems

    • Investigate the evolution of photoreceptor-specific regulatory networks

    • Explore the conservation of RCVRN-associated pathways between mouse models and human retina

These novel research directions leverage the unique capabilities of RCVRN mouse models to address fundamental questions in retinal biology and develop new therapeutic approaches for retinal diseases.

Product Science Overview

Introduction

Recoverin is a low molecular-weight, neuronal calcium sensor (NCS) protein primarily located in the photoreceptor outer segments of the vertebrate retina . It plays a crucial role in the recovery phase of visual excitation and adaptation to background light . This article delves into the historical background, structure, function, and significance of Recoverin, particularly focusing on the mouse recombinant version.

Historical Background

The discovery of Recoverin dates back to 1989 when P. Philippov’s group from M.V. Lomonosov Moscow State University developed a method for purifying visual G-protein transducin and other G-proteins . During this process, they identified an unknown protein with an apparent molecular weight of 26 kDa, which they named "p26" . This protein was later found to be specific to the retina, particularly the photoreceptor layer, and was capable of binding calcium ions (Ca²⁺) due to its EF-hand type calcium-binding sites . The protein was subsequently renamed “Recoverin” due to its role in photoreceptor recovery .

Structure

Recoverin contains several EF-hand type Ca²⁺-binding sites, which are crucial for its function . The binding of calcium to these sites induces a conformational change in the protein, enabling it to interact with other molecules . Recoverin also undergoes N-terminal myristoylation, a lipid modification that facilitates its attachment to cellular membranes .

Function

Recoverin plays a pivotal role in the visual cycle by acting as a calcium sensor . In the dark, calcium-bound Recoverin inhibits G-protein-coupled receptor kinase (GRK), preventing the phosphorylation of rhodopsin, a visual receptor . This inhibition is lifted when light reduces intracellular calcium levels, allowing GRK to phosphorylate rhodopsin and initiate the recovery phase of visual excitation . Recoverin’s ability to bind to phospholipid membranes further enhances its regulatory functions .

Significance

Recoverin is not only essential for normal visual function but also serves as a marker for evaluating the differentiation of pluripotent stem cells into rod and cone photoreceptors or cone bipolar cells . Additionally, Recoverin is classified as a cancer-retina antigen due to its aberrant expression in tumors of the lung and other tissues . This makes it a valuable tool in both basic research and clinical diagnostics.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.