Note: While we prioritize shipping the format currently in stock, please specify your preferred format in order notes for fulfillment according to your requirements.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
The tag type is determined during the production process. If you require a specific tag, please inform us, and we will prioritize its development.
FV3-007R is one of the many uncharacterized proteins encoded by the Frog virus 3 genome. Based on patterns observed in other FV3 proteins, we can estimate that FV3-007R likely has characteristics similar to other viral proteins in the same family. FV3 proteins typically range from small proteins (like FV3-004R at 60 amino acids with a molecular weight of 7.58 kDa) to much larger proteins (such as FV3-003R at 438 amino acids with a molecular weight of 56.17 kDa) . Without specific data on FV3-007R, researchers should first determine basic parameters including:
Amino acid sequence length
Molecular weight
Isoelectric point (pI)
Hydrophobicity profile
Predicted secondary structure
For reference, other FV3 proteins show varying properties as demonstrated in this comparative data table:
| Protein | Length (aa) | Molecular weight (kDa) | Isoelectric point | Nature | Hydrophobicity % |
|---|---|---|---|---|---|
| FV3-001R | 256 | 34.33 | 1.61 | Hydrophobic | 42% |
| FV3-002L | 320 | 40.40 | 1.92 | Hydrophobic | 39% |
| FV3-003R | 438 | 56.17 | 2.62 | Hydrophobic | 47% |
| FV3-004R | 60 | 7.58 | 0.36 | Hydrophobic | 43% |
Initial characterization should begin with comprehensive bioinformatic analysis:
Perform sequence alignment with other iridovirus proteins to identify conserved domains
Use prediction tools to identify potential functional motifs
Compare with the core set of 72 genes conserved across ranaviruses
Determine if FV3-007R is likely an immediate early (IE), delayed early (DE), or late (L) gene based on promoter analysis
Analyze the genomic context of FV3-007R to predict potential functions
Many FV3 genes show marked sequence conservation within the family, particularly among structural proteins like the major capsid protein (MCP) . Determining whether FV3-007R belongs to the conserved core genes or is unique to FV3 will provide initial clues to its function.
The choice of expression system depends on research goals:
Bacterial expression systems (E. coli):
Advantages: Rapid growth, high yield, cost-effective
Limitations: Lack of post-translational modifications, potential issues with folding
Recommended for: Initial structural studies, antibody production
Insect cell expression (Baculovirus):
Advantages: Post-translational modifications, better folding of complex proteins
Limitations: Longer production time, higher cost
Recommended for: Functional studies requiring proper protein folding
Mammalian expression systems:
Advantages: Native-like post-translational modifications, proper folding
Limitations: Highest cost, lower yields
Recommended for: Critical functional analyses
When designing expression constructs, consider incorporating:
Affinity tags (His-tag, GST) for purification
Fluorescent protein fusions for localization studies
Cleavable tags to remove fusion partners after purification
Purification strategy should be tailored to the protein's properties:
Determine solubility first - many viral proteins can be insoluble when overexpressed
For soluble proteins:
Affinity chromatography based on incorporated tags
Ion exchange chromatography (consider predicted pI)
Size exclusion chromatography for final polishing
For insoluble proteins:
Gentle solubilization with non-ionic detergents
Refolding from inclusion bodies if necessary
Consider fusion partners that enhance solubility
Monitor protein quality using:
SDS-PAGE for purity assessment
Western blotting for identity confirmation
Circular dichroism to assess secondary structure
Dynamic light scattering for aggregation analysis
Multiple complementary approaches should be employed:
Temporal expression analysis:
Localization studies:
Protein-protein interaction studies:
Co-immunoprecipitation with viral and host proteins
Yeast two-hybrid or proximity labeling approaches
Mass spectrometry to identify interaction partners
Loss-of-function approaches:
To determine essentiality:
Generate knockout mutants using homologous recombination to replace FV3-007R with a selectable marker
Assess viral replication kinetics in permissive cell lines
Compare single-step and multi-step growth curves between wild-type and mutant viruses
Examine plaque morphology and size
Conduct complementation studies with ectopically expressed FV3-007R
If the knockout is lethal, conditional expression systems can be employed to further study the essential function:
Tetracycline-inducible expression
Temperature-sensitive mutants
Complementing cell lines expressing the protein of interest
Structural determination requires a multi-faceted approach:
In silico structure prediction:
Use AlphaFold2 or RoseTTAFold for initial structural models
Molecular dynamics simulations to predict flexibility
Compare with structural homologs if identified
Experimental structure determination:
X-ray crystallography (requires protein crystallization)
Cryo-electron microscopy (particularly if part of larger complexes)
NMR spectroscopy (if protein size permits)
Small-angle X-ray scattering (SAXS) for solution structure
Limited proteolysis:
Identify stable domains and flexible regions
Guide construct design for structural studies
Information on protein folding and accessibility
Post-translational modifications (PTMs) can significantly impact protein function:
Mass spectrometry-based approaches:
Bottom-up proteomics for identification of specific modifications
Top-down proteomics for intact protein analysis
Targeted analysis for specific modifications (phosphorylation, methylation, etc.)
Modification-specific detection methods:
Phospho-specific antibodies for phosphorylation
Pro-Q Diamond staining for phosphoprotein detection
Glycoprotein-specific staining methods
Functional impact assessment:
Site-directed mutagenesis of modified residues
Comparison of activity before and after treatment with modification-removing enzymes
In vitro enzymatic assays to identify responsible modification enzymes
FV3 is known to encode multiple proteins that modulate host immune responses . To determine if FV3-007R plays a role:
Immunomodulation screening:
Assess impact on host interferon responses
Evaluate effects on NF-κB signaling pathway
Determine if FV3-007R affects apoptosis pathways
Host protein binding studies:
Identify host binding partners through pull-down assays
Validate interactions through co-immunoprecipitation
Map interaction domains through deletion constructs
Comparative analysis:
Selection of appropriate experimental systems is crucial:
Cell lines:
Fathead minnow (FHM) cells for permissive growth
Xenopus laevis cell lines for amphibian host studies
BHK-21 (baby hamster kidney) cells for mammalian expression
Primary cell cultures:
Amphibian hepatocytes or kidney cells
Xenopus peritoneal macrophages for immune studies
Fish cell lines for comparative host range studies
Animal models:
Xenopus laevis tadpoles for in vivo studies
Ambystoma tigrinum (tiger salamander) for alternate host studies
Consider developmental stage (tadpole vs. adult) for age-dependent effects
Antibody production requires careful planning:
Epitope selection:
Analyze predicted surface-exposed regions
Avoid highly conserved domains if specificity for FV3-007R is required
Consider multiple epitopes for comprehensive protein detection
Production strategy:
Recombinant protein immunization for polyclonal antibodies
Synthetic peptide approach for region-specific detection
Monoclonal antibody development for reproducible detection
Validation methods:
Western blot against recombinant protein
Immunoprecipitation efficiency testing
Pre-absorption controls
Testing in FV3-007R knockout/knockdown systems
CRISPR/Cas9 genome editing for viral studies requires specific considerations:
Guide RNA design:
Target FV3-007R coding sequence while avoiding essential neighboring genes
Design multiple gRNAs targeting different regions
Check for potential off-target effects in both viral and host genomes
Delivery methods:
Transfection of CRISPR/Cas9 components before viral infection
Construction of recombinant virus through homologous recombination
Use of Cas9-expressing cell lines for improved efficiency
Screening and validation:
PCR-based screening for genomic modifications
Sequencing confirmation of edited regions
Western blot verification of protein knockout
Functional complementation to confirm phenotype specificity