Recombinant Frog virus 3 Uncharacterized protein 084R (FV3-084R)

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 customized fulfillment.
Lead Time
Delivery times vary depending on the purchase method and location. Please contact your local distributor for precise delivery estimates.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires advance notice 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 consolidate 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%, but this can be adjusted based on customer requirements.
Shelf Life
Shelf life depends on several factors including storage conditions, buffer components, temperature, and the protein's inherent stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during the manufacturing process.
The tag type will be determined during production. If a specific tag type is required, please inform us in advance to facilitate preferential development.
Synonyms
FV3-084RUncharacterized protein 084R
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-245
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Frog virus 3 (isolate Goorha) (FV-3)
Target Names
FV3-084R
Target Protein Sequence
MLWEAVTDKP VKLKGLLELL LNNMDSARLV VTSQSVSVVD YQSNMAVTAS MPSSVFTSYV YKSDAECLYA GLPHAALPDL KSFKAKCNVT LRLMGDPECG QYTMKIIIAN ASHMSTSINM VVDHGKKEAD RGHPEGAGKP FTLTQQEFNT LCKTFKQGPV NLGVFGGVLV ASGGVDGIKV KEVAFGAPDC VTPHVKLCVH AEKMSRLVKM GPFSAGSLTV CVAQGSVTVS TCGHLGSLTV TLFEG
Uniprot No.

Q&A

What is FV3-084R and how does it fit into the Frog Virus 3 genomic structure?

FV3-084R is one of approximately 98 annotated open reading frames (ORFs) in the Frog Virus 3 genome. As a large double-stranded DNA virus, FV3 contains coding genes that have been classified into temporal expression categories (immediate early, delayed early, and late viral transcripts) . The 084R designation indicates its position in the viral genome sequence. Like many FV3 proteins, 084R remains largely uncharacterized, but whole transcriptomic analyses have confirmed its expression during viral infection in multiple host tissues .

Comprehensive transcriptomic studies have mapped reads spanning the full FV3 genome at approximately 10× depth on both positive and negative strands in infected tissues including intestine, liver, spleen, lung, and kidney, while reads were only mapped to partial genomic regions in infected thymus, skin, and muscle . This tissue-dependent expression pattern suggests varied roles for viral proteins, including 084R, in different host environments.

What expression patterns have been observed for FV3-084R in infected tissues?

Transcriptomic analyses of FV3-infected Xenopus laevis tissues have documented differential expression patterns of viral genes, including 084R, across various tissues. The most robust viral gene expression was observed in kidney tissues, with substantial expression also detected in intestine, liver, spleen, and lung . This tissue-dependent expression suggests that FV3-084R may have specialized functions in specific host environments.

When comparing wild-type FV3 (FV3-WT) with an ORF64R-deleted recombinant strain (FV3-Δ64R), researchers observed differential expression of multiple viral genes across tissue types . This indicates that the expression of viral proteins, potentially including 084R, can be influenced by mutations in other parts of the viral genome, suggesting complex regulatory networks within the virus.

Are there any predicted functional domains in FV3-084R?

While specific information about FV3-084R domains is limited in the available literature, recent transcriptomic analyses of FV3 have identified several putative ORFs that encode hypothetical proteins containing viral mimicking conserved domains found in host interferon (IFN) regulatory factors (IRFs) and IFN receptors . These findings suggest that some uncharacterized FV3 proteins might play roles in immune evasion by mimicking host immune components.

Researchers investigating FV3-084R should perform domain prediction analyses using tools such as SMART, Pfam, or InterProScan to identify potential functional regions. Additionally, sequence comparison with other ranaviral proteins and proteins from more distantly related viruses might reveal conserved motifs that could indicate functional significance.

What expression systems are most suitable for recombinant FV3-084R production?

Multiple expression systems can be employed for FV3-084R recombinant protein production, each with distinct advantages depending on research objectives:

Expression SystemAdvantagesConsiderations for FV3-084R
E. coliHigh yield, cost-effective, rapidMay lack post-translational modifications; potential folding issues
Yeast (S. cerevisiae, P. pastoris)Eukaryotic processing, higher folding fidelityModerate yield, longer production time
Insect cells (Sf9, Sf21, High Five)Near-native eukaryotic processingHigher cost, complex methodology
Mammalian cells (293, 293T, CHO)Most authentic processing and foldingHighest cost, lowest yield, longest production time

For initial characterization studies, bacterial expression using strains like BL21(DE3), JM115, or Rosetta-GAMI is recommended as a starting point . If protein functionality requires eukaryotic post-translational modifications, insect cell systems using Sf9 or Sf21 cells may provide a better balance between authentic processing and reasonable yield .

Which fusion tags optimize solubility and purification of FV3-084R?

Selection of appropriate fusion tags can significantly impact recombinant FV3-084R solubility, folding, and purification efficiency:

Fusion TagPrimary BenefitRecommended Application for FV3-084R
His-TagEfficient purification via metal affinityStandard initial approach; can be positioned at N or C terminus
MBP (Maltose Binding Protein)Enhances solubilityRecommended if initial His-tagged constructs show poor solubility
GST (Glutathione S-Transferase)Improves folding and solubilityAlternative to MBP; enables pull-down assays
FLAGMinimal size, high specificity detectionUseful for co-immunoprecipitation studies
TrxA (Thioredoxin)Enhances disulfide bond formationBeneficial if FV3-084R contains multiple cysteines

Tag position (N-terminal vs. C-terminal) can significantly impact protein folding and function . For uncharacterized proteins like FV3-084R, preparing constructs with tags at both positions is advisable to determine optimal configuration for soluble, functional protein production.

What protein reprocessing techniques are critical for obtaining functional FV3-084R?

Depending on expression results, various reprocessing techniques may be necessary:

  • Protein renaturation: If FV3-084R forms inclusion bodies in bacterial systems, denaturation with urea or guanidine HCl followed by controlled refolding through dialysis may be required.

  • Endotoxin removal: Critical for immunological studies, particularly if using E. coli expression systems.

  • Filtration sterilization: Necessary for cell culture applications and to prevent contamination during storage.

  • Lyophilization: For long-term storage, particularly when high protein concentrations are needed .

The optimal reprocessing strategy should be determined empirically, with protein activity assays performed after each step to ensure maintenance of functional integrity.

How can researchers determine the temporal expression class of FV3-084R?

The temporal expression pattern of FV3-084R (immediate early, delayed early, or late) provides important clues about its function in the viral replication cycle. To determine this:

  • Time-course experiments: Infect host cells (e.g., Xenopus cell lines) with FV3 and collect RNA samples at defined intervals post-infection (e.g., 1h, 3h, 6h, 12h, 24h).

  • Inhibitor studies: Use cycloheximide (protein synthesis inhibitor) to identify immediate early genes expressed without de novo protein synthesis, or phosphonoacetic acid (viral DNA synthesis inhibitor) to distinguish between delayed early and late genes.

  • RT-qPCR analysis: Design primers specific to FV3-084R and quantify expression relative to known immediate early (e.g., 18K), delayed early (e.g., vPol), and late (e.g., MCP) viral genes.

  • RNA-Seq comparison: Compare expression profiles with established temporal class patterns identified in previous transcriptomic studies .

This classification would place FV3-084R into the broader context of the viral replication cycle and provide initial clues about its potential function.

What approaches should be used to investigate potential immune evasion functions of FV3-084R?

Recent transcriptomic analyses have identified several FV3 proteins with potential immune evasion functions through molecular mimicry of host interferon regulatory factors (IRFs) and interferon receptors . To investigate whether FV3-084R shares these properties:

  • Sequence analysis: Perform comprehensive bioinformatic comparisons with known viral immune evasion proteins and host immune components.

  • Reporter gene assays: Transfect cells with IFN-responsive promoter reporters (e.g., ISRE-luciferase) with and without FV3-084R expression to determine effects on IFN signaling.

  • Co-immunoprecipitation studies: Investigate physical interactions between FV3-084R and host immune signaling components.

  • Knockout/knockdown experiments: Generate FV3-084R-deficient virus through CRISPR/Cas9 genome editing and assess changes in viral pathogenesis and host immune response.

  • Structural studies: If molecular mimicry is suspected, crystallization and structural determination would provide definitive evidence of structural homology with host immune components.

These approaches would help determine whether FV3-084R contributes to the virus's ability to evade host immune responses, which is particularly relevant given the virus's impact on vulnerable amphibian populations.

How can differential tissue tropism of FV3-084R expression be experimentally verified?

Transcriptomic data suggests tissue-dependent expression patterns for FV3 genes . To specifically verify FV3-084R tissue tropism:

  • Tissue-specific RT-qPCR: Quantify FV3-084R transcript levels across multiple tissues from infected animals, normalizing to viral genome copy number to account for differences in infection levels.

  • In situ hybridization: Develop FV3-084R-specific probes to visualize expression patterns within tissue sections, providing spatial context for expression.

  • Custom antibody development: Generate antibodies against recombinant FV3-084R for immunohistochemistry studies of protein localization in infected tissues.

  • Tissue-specific transcriptomics: Perform comparative RNA-Seq on multiple tissues with varying infection levels to identify correlations between FV3-084R expression and tissue-specific host factors.

A standardized experimental setup is crucial for meaningful comparisons:

Tissue TypeSample Collection TimingProcessing MethodAnalysis Techniques
Kidney3, 6, 9 days post-infectionFlash frozen/RNAlaterRT-qPCR, RNA-Seq
Liver3, 6, 9 days post-infectionFlash frozen/RNAlaterRT-qPCR, RNA-Seq
Spleen3, 6, 9 days post-infectionFlash frozen/RNAlaterRT-qPCR, RNA-Seq
Intestine3, 6, 9 days post-infectionFlash frozen/RNAlaterRT-qPCR, RNA-Seq
Skin3, 6, 9 days post-infectionFFPEIHC, ISH
Muscle3, 6, 9 days post-infectionFFPEIHC, ISH

What protein interaction networks might involve FV3-084R during infection?

Understanding the protein interaction networks involving FV3-084R is crucial for deciphering its function. Several complementary approaches should be employed:

  • Yeast Two-Hybrid screening: Using FV3-084R as bait against both viral and host cDNA libraries to identify potential interacting partners.

  • Proximity labeling: Expressing FV3-084R fused to BioID or APEX2 in host cells to label proximal proteins for mass spectrometry identification.

  • Co-immunoprecipitation followed by mass spectrometry: Pull-down experiments using tagged FV3-084R to identify interaction partners.

  • Protein microarrays: Probing arrays containing host cellular proteins with labeled recombinant FV3-084R.

Based on transcriptomic studies of FV3, special attention should be given to potential interactions with components of host interferon signaling pathways, as several FV3 proteins have been implicated in immune evasion through molecular mimicry of host immune components .

How can CRISPR/Cas9 genome editing be utilized to study FV3-084R function in viral replication?

CRISPR/Cas9 technology offers powerful approaches for functional studies of FV3-084R:

  • Knockout studies: Create FV3-084R-deficient virus by targeting the gene with CRISPR/Cas9, followed by phenotypic characterization:

    • Viral replication kinetics in cell culture

    • Host range alterations

    • Pathogenesis in animal models

    • Transcriptomic changes in infected cells

  • Knockin studies: Insert reporter genes (e.g., fluorescent proteins) in-frame with FV3-084R to track expression dynamics and localization.

  • Domain mapping: Create targeted mutations in specific domains to identify functional regions without removing the entire protein.

  • Complementation experiments: Re-introduce wild-type or modified versions of FV3-084R to FV3-084R-deficient virus to confirm phenotype specificity.

When designing guide RNAs for FV3-084R editing, careful analysis of the genomic context is essential to avoid off-target effects on overlapping genes or regulatory elements.

What structural biology approaches are most promising for determining FV3-084R structure?

Determining the three-dimensional structure of FV3-084R would provide invaluable insights into its function. Several complementary approaches should be considered:

For any structural studies, it's critical to ensure that recombinant FV3-084R retains native folding and function through activity assays appropriate to its predicted function.

How does FV3-084R expression compare between different FV3 strains?

Comparing FV3-084R expression between viral strains can provide insights into its role in virulence and host adaptation:

  • Comparative transcriptomics: RNA-Seq analysis comparing FV3-WT and FV3-Δ64R strains has already revealed differential expression patterns for many viral genes across various tissues . Similar approaches can be extended to other FV3 variants.

  • Sequence analysis: Compare the FV3-084R sequence across different FV3 isolates to identify conserved regions (likely functional domains) and variable regions (potential adaptation to different hosts or environments).

  • Promoter analysis: Examine the upstream regulatory regions of FV3-084R across strains to identify potential differences in expression control.

  • RT-qPCR validation: Quantify FV3-084R transcript levels in cells infected with different FV3 strains under standardized conditions.

These comparative approaches might reveal correlations between FV3-084R expression patterns and strain-specific properties such as virulence, host range, or tissue tropism.

What are the key knowledge gaps regarding FV3-084R that require further investigation?

Despite recent advances in FV3 transcriptomics , significant knowledge gaps remain regarding FV3-084R:

  • Functional characterization: The biological function of FV3-084R remains unknown. Priority should be given to knockout studies and phenotypic characterization.

  • Structural information: No structural data exists for FV3-084R, limiting our understanding of its mechanism of action.

  • Evolutionary conservation: Comparative analysis across ranaviruses could reveal the evolutionary importance of this protein.

  • Host interactions: Identification of host cell factors that interact with FV3-084R would provide insights into its role during infection.

  • Temporal regulation: Precise classification of FV3-084R into temporal expression classes (immediate early, delayed early, or late) would help contextualize its function.

Addressing these knowledge gaps through methodical research will contribute to our understanding of ranaviral pathogenesis and potentially identify new targets for intervention strategies.

How might research on FV3-084R contribute to broader understanding of ranaviral pathogenesis?

Research on FV3-084R has potential implications beyond basic characterization:

  • Immune evasion mechanisms: If FV3-084R contains viral mimicking domains similar to those found in other FV3 proteins , it may contribute to understanding how ranaviruses evade host immune responses.

  • Viral evolution: Comparative studies of FV3-084R across ranaviral species could illuminate evolutionary adaptations to different hosts.

  • Diagnostic applications: If FV3-084R is consistently expressed during infection, it could serve as a diagnostic marker for FV3 infection.

  • Conservation implications: Understanding the molecular mechanisms of FV3 pathogenesis, including the role of FV3-084R, could inform conservation strategies for vulnerable amphibian populations affected by this virus.

  • Biotechnological applications: Uncharacterized viral proteins sometimes possess unique enzymatic or regulatory properties that can be harnessed for biotechnology applications.

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.