Recombinant Frog virus 3 Uncharacterized protein 007R (FV3-007R)

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder

Note: While we prioritize shipping the format currently in stock, please specify your preferred format in order notes for fulfillment according to your requirements.

Lead Time
Delivery times vary depending on the purchasing method and location. Please contact your local distributor for precise delivery estimates.

Note: Standard shipping includes blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.

Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to collect the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50%, provided as a reference for your consideration.
Shelf Life
Shelf life depends on various factors including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized formulations have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquot for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.

The tag type is determined during the production process. If you require a specific tag, please inform us, and we will prioritize its development.

Synonyms
FV3-007RUncharacterized protein 007R
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-128
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Frog virus 3 (isolate Goorha) (FV-3)
Target Names
FV3-007R
Target Protein Sequence
MRSIKPLRCC NAHGRHVSQE YGRCTLLLFR EKLFLQTGLV CNKQCNAPNN DGAESKHHGI HHGSRGALAL RGAGVHLLAS AALGPRVLAG LVPTGRSVQG SVGQCGRVAQ IGRARDVAAR KQESYCEK
Uniprot No.

Q&A

What is currently known about the fundamental properties of FV3-007R?

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:

ProteinLength (aa)Molecular weight (kDa)Isoelectric pointNatureHydrophobicity %
FV3-001R25634.331.61Hydrophobic42%
FV3-002L32040.401.92Hydrophobic39%
FV3-003R43856.172.62Hydrophobic47%
FV3-004R607.580.36Hydrophobic43%

How should researchers approach initial sequence analysis of FV3-007R?

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.

What expression systems are most suitable for recombinant FV3-007R production?

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

What are the optimal purification strategies for recombinant FV3-007R?

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

What experimental approaches are most effective for determining the function of FV3-007R?

Multiple complementary approaches should be employed:

  • Temporal expression analysis:

    • Determine whether FV3-007R is an immediate early (IE), delayed early (DE), or late (L) gene

    • Use RT-qPCR to measure transcript levels during infection

    • Correlate expression timing with viral replication phases

  • Localization studies:

    • Generate fluorescently tagged FV3-007R to track localization

    • Co-localization with cellular organelles can suggest function

    • Assess whether the protein localizes to viral assembly sites (VAS)

  • 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:

    • Antisense morpholino oligonucleotides (asMO) for knockdown

    • siRNA-mediated silencing

    • CRISPR/Cas9 gene knockout if working with recombinant virus

How can researchers determine if FV3-007R is essential for viral replication?

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

What approaches should be used to determine the structure of FV3-007R?

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

How can post-translational modifications of FV3-007R be identified and characterized?

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

How does FV3-007R potentially interact with host immune responses?

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:

    • Examine if FV3-007R has homologs in other ranaviruses

    • Compare host responses to wild-type vs. FV3-007R-deficient virus

    • Consider if FV3-007R belongs to the virus-encoded immune evasion proteins like vCARD or βHSD

What cell culture systems are most appropriate for studying FV3-007R function?

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

What are the key considerations when designing antibodies against FV3-007R?

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

How can CRISPR/Cas9 technology be optimized for studying FV3-007R?

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

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.