Recombinant Frog virus 3 Uncharacterized protein 004R (FV3-004R)

Shipped with Ice Packs
In Stock

Description

Key Features:

  • Conserved Motifs: Contains hydrophobic regions indicative of membrane association .

  • Post-Translational Modifications: Predicted myristylation sites, common in viral proteins for host-cell membrane binding .

  • Functional Uncharacterization: Despite its classification, no direct enzymatic or immune-modulatory roles have been experimentally validated .

Genomic and Evolutionary Context

FV3-004R is encoded by ORF 004R in the FV3 genome (GenBank: NC_005946.1), which spans 957 nucleotides . FV3 is notable for its recombinant evolution, with studies revealing widespread genetic exchange between FV3 and Common midwife toad virus (CMTV) . While FV3-004R itself has not been linked to recombination events, its genomic neighbors (e.g., ORFs 003R and 005L) show interspecies recombination patterns .

Genomic FeatureDetails
Genome Position4443–5399 bp (FV3 strain Goorha)
Conserved DomainsDUF230 (pfam03003)
HomologsMyristylated membrane proteins (70% identity)
Recombinant PotentialNot directly observed, but neighboring ORFs show CMTV-FV3 recombination

Research Gaps and Future Directions

Despite its availability, FV3-004R remains understudied:

  1. Functional Role: No in vivo studies confirm its role in viral replication or immune evasion.

  2. Host Interactions: Potential binding partners or pathways remain unidentified .

  3. Structural Biology: Lack of crystallography or cryo-EM data limits mechanistic insights.

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order remarks. We will accommodate your request whenever possible.
Lead Time
Delivery times vary depending on the purchasing method and location. Please contact your local distributor for precise delivery estimates.
Note: Our proteins are shipped with standard 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
Before opening, briefly centrifuge the vial 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 guideline.
Shelf Life
Shelf life depends on several factors: storage conditions, buffer composition, temperature, and protein 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 manufacturing.
Tag type is determined during production. If you require a specific tag, please inform us; we will prioritize its development.
Synonyms
FV3-004R; Uncharacterized protein 004R
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-60
Protein Length
full length protein
Species
Frog virus 3 (isolate Goorha) (FV-3)
Target Names
FV3-004R
Target Protein Sequence
MNAKYDTDQGVGRMLFLGTIGLAVVVGGLMAYGYYYDGKTPSSGTSFHTASPSFSSRYRY
Uniprot No.

Target Background

Database Links

KEGG: vg:2947776

Subcellular Location
Host membrane; Single-pass membrane protein.

Q&A

What is the structural characterization of the FV3-004R protein?

FV3-004R is a small uncharacterized protein (60 amino acids) encoded by the frog virus 3 genome. The full amino acid sequence is MNAKYDTDQGVGRMLFLGTIGLAVVVGGLMAYGYYYDGKTPSSGTSFHTASPSFSSRYRY . Based on sequence analysis, the protein contains hydrophobic regions suggesting potential membrane association, though its specific structure-function relationship remains under investigation.

For structural studies, researchers typically express the recombinant protein with an N-terminal His-tag in E. coli expression systems . The protein is generally purified to >90% purity as determined by SDS-PAGE before being used in structural analyses such as circular dichroism or crystallography attempts.

What is the temporal expression pattern of FV3-004R during viral infection?

FV3 genes are expressed in a coordinated fashion leading to the sequential appearance of immediate early (IE), delayed early (DE), and late (L) viral transcripts . Transcriptome analyses using oligonucleotide microarrays containing 70-mer probes corresponding to each of the 98 FV3 ORFs have been crucial in determining the temporal expression patterns .

Based on comprehensive temporal classification studies of FV3 genes, researchers have identified:

  • 33 immediate early (IE) genes

  • 22 delayed early (DE) genes

  • 36 late (L) genes

While the specific classification of FV3-004R was not explicitly mentioned in the search results, the temporal expression pattern can be determined through similar microarray analyses or RT-PCR validation approaches used for other FV3 genes .

How should recombinant FV3-004R protein be properly stored and handled in laboratory settings?

For optimal stability and activity, recombinant FV3-004R protein should be stored according to the following protocol:

  • Upon receipt, briefly centrifuge the vial before opening to bring contents to the bottom

  • Reconstitute the lyophilized protein in deionized sterile water to a concentration of 0.1-1.0 mg/mL

  • Add glycerol to a final concentration of 5-50% (with 50% being the standard recommendation)

  • Aliquot for long-term storage at -20°C/-80°C to avoid repeated freeze-thaw cycles

  • For short-term use, working aliquots can be stored at 4°C for up to one week

It's important to note that repeated freezing and thawing is not recommended as it may lead to protein degradation and loss of activity . For experimental reproducibility, researchers should document the storage conditions and number of freeze-thaw cycles when reporting results using this protein.

What are the recommended protocols for expressing and purifying recombinant FV3-004R protein?

The expression and purification of recombinant FV3-004R protein typically follows this methodological approach:

  • Construct Design: The full-length gene (encoding amino acids 1-60) is cloned into a bacterial expression vector with an N-terminal His-tag .

  • Expression System: Transform the construct into an appropriate E. coli strain. BL21(DE3) or similar strains are commonly used for recombinant viral protein expression .

  • Induction Conditions:

    • Culture bacteria in LB medium supplemented with appropriate antibiotics

    • Grow at 37°C until OD600 reaches 0.6-0.8

    • Induce with IPTG (typically 0.5-1.0 mM)

    • Continue growth at lower temperature (16-25°C) for 4-16 hours

  • Cell Harvest and Lysis:

    • Harvest cells by centrifugation (5,000 × g, 15 min, 4°C)

    • Resuspend in lysis buffer containing protease inhibitors

    • Lyse cells using sonication or high-pressure homogenization

  • Purification:

    • Affinity chromatography using Ni-NTA resin to capture His-tagged protein

    • Wash with increasing imidazole concentrations

    • Elute with high imidazole buffer

    • Optional further purification by size exclusion chromatography

  • Quality Control:

    • Assess purity by SDS-PAGE (should be >90%)

    • Confirm identity by Western blot or mass spectrometry

  • Buffer Exchange and Storage:

    • Exchange into Tris/PBS-based buffer containing 6% Trehalose, pH 8.0

    • Lyophilize or store as described in the storage protocol

How can researchers design experiments to investigate potential functions of the uncharacterized FV3-004R protein?

Investigating the function of uncharacterized viral proteins like FV3-004R requires a multi-faceted experimental approach:

  • Bioinformatic Analysis:

    • Sequence homology searches against characterized proteins

    • Prediction of structural motifs and domains

    • Comparative analysis with related ranavirus proteins

  • Localization Studies:

    • Express fluorescently-tagged FV3-004R in infected cells

    • Track subcellular localization using confocal microscopy

    • Perform co-localization studies with cellular compartment markers

  • Protein-Protein Interaction Studies:

    • Yeast two-hybrid screens with host cell proteins

    • Co-immunoprecipitation experiments

    • Proximity labeling approaches (BioID or APEX)

  • Functional Assays:

    • Gene knockout or knockdown studies using CRISPR-Cas9

    • Overexpression studies and phenotypic analysis

    • Effects on viral replication kinetics

  • Structural Biology:

    • X-ray crystallography or NMR studies

    • Cryo-EM analysis in context of viral particles

  • Host Response Analysis:

    • Transcriptome analysis of host cells in presence/absence of FV3-004R

    • Analysis of immune response modulation

A systematic approach combining these methodologies would help elucidate the function of this currently uncharacterized protein.

What analytical techniques are most effective for studying interactions between FV3-004R and host cell proteins?

Several complementary analytical techniques can be employed to study FV3-004R interactions with host proteins:

  • Affinity Purification-Mass Spectrometry (AP-MS):

    • Express tagged FV3-004R in host cells

    • Purify protein complexes using affinity tags

    • Identify interacting partners by LC-MS/MS

    • Quantify enrichment against appropriate controls

  • Proximity-Dependent Biotin Identification (BioID):

    • Fuse FV3-004R to a biotin ligase (BirA*)

    • Express in cells and allow biotinylation of proximal proteins

    • Purify biotinylated proteins and identify by MS

    • Advantage: captures transient interactions

  • Surface Plasmon Resonance (SPR):

    • Immobilize purified FV3-004R on sensor chip

    • Flow potential interacting proteins over surface

    • Measure binding kinetics and affinity constants

    • Advantage: provides quantitative interaction data

  • Microscale Thermophoresis (MST):

    • Label FV3-004R or potential binding partners

    • Monitor thermophoretic movement upon binding

    • Determine binding constants in solution

    • Advantage: requires small sample amounts

  • Cross-linking Mass Spectrometry (XL-MS):

    • Cross-link protein complexes in cells

    • Digest and analyze by MS

    • Identify cross-linked peptides to map interaction sites

    • Advantage: provides structural information about the complex

  • Förster Resonance Energy Transfer (FRET):

    • Tag FV3-004R and potential partner with fluorophore pairs

    • Monitor energy transfer as indicator of proximity

    • Advantage: can be performed in living cells

Each technique has strengths and limitations; therefore, a combination of approaches is recommended for comprehensive interaction characterization.

How does recombination between FV3 and other ranaviruses potentially affect the FV3-004R gene region?

Recombination between FV3 and other ranaviruses, particularly common midwife toad virus (CMTV), has been widely documented and can significantly impact viral genes including potentially FV3-004R . Analysis of recombination events in ranavirus genomes reveals several key patterns:

  • Recombination Breakpoints: Most recombination breakpoints are located within open reading frames (ORFs), generating new ORFs and proteins that are mosaics between FV3 and CMTV . This creates chimeric proteins with potentially altered functions.

  • Protein Composition Ratios: The FV3/CMTV ratio within recombinant ORFs varies from 0 (entirely CMTV-like) to 0.98 (mostly FV3-like), depending on where the recombination events occur .

  • Impact on Protein Function: When recombination occurs within an ORF, it can generate a novel protein with domains from both parental viruses, potentially altering function, host interactions, or virulence properties.

While the search results don't specifically mention FV3-004R in the context of recombination, the widespread nature of recombination events throughout the FV3 genome suggests this gene could be affected. Researchers investigating FV3-004R should consider analyzing sequence variations across isolates to identify potential recombination events affecting this specific region.

What methodological approaches are best for analyzing potential recombination events affecting the FV3-004R gene?

To analyze potential recombination events affecting FV3-004R, researchers should employ a systematic approach combining:

  • Genome Sequencing:

    • Full genome sequencing of multiple FV3 isolates from different geographic regions

    • Use of next-generation sequencing with sufficient depth (>30×)

    • Assembly and annotation focusing on the FV3-004R region

  • Recombination Detection Algorithms:

    • RDP4 suite (incorporating methods like GENECONV, BootScan, MaxChi)

    • GARD (Genetic Algorithm for Recombination Detection)

    • HyPhy package for detecting selection pressures in recombinant regions

  • Comparative Genomic Analysis:

    • Multiple sequence alignment of FV3-004R across isolates

    • Comparison with homologs in related ranaviruses

    • Phylogenetic analysis to detect incongruences indicative of recombination

  • Breakpoint Analysis:

    • Precise mapping of potential recombination breakpoints

    • Analysis of ORF integrity and protein coding potential

    • Assessment of FV3/CMTV ratios in recombinant regions

  • Functional Validation:

    • Expression of putative recombinant FV3-004R variants

    • Comparative functional assays

    • Structural analysis of protein changes

The analysis should include appropriate statistical validation and multiple hypothesis testing correction to minimize false positives in recombination detection.

How does the temporal expression pattern of FV3-004R compare with other genes in the ranavirus genome?

Understanding the temporal expression pattern of FV3-004R requires comparing its expression with the established patterns of other FV3 genes. FV3 gene expression follows a coordinated cascade with three temporal classes:

  • Immediate Early (IE) Genes:

    • Expressed very early in infection

    • Do not require de novo protein synthesis

    • Typically involved in regulatory functions

    • Can be identified by expression in presence of cycloheximide (CHX)

    • 33 genes fall into this category in FV3

  • Delayed Early (DE) Genes:

    • Require prior IE gene expression

    • Often involved in nucleic acid metabolism

    • 22 genes identified in this category

  • Late (L) Genes:

    • Expressed after viral DNA replication begins

    • Often encode structural proteins

    • Can be identified using temperature-sensitive mutants defective in DNA synthesis

    • 36 genes identified in this category

To determine the specific temporal class of FV3-004R, researchers should:

  • Perform time-course RT-PCR or qRT-PCR analysis (2, 4, and 9 hours post-infection)

  • Analyze expression in presence of cycloheximide (CHX)

  • Test expression using temperature-sensitive mutants at non-permissive temperatures

  • Compare results with microarray data from other FV3 genes

This methodological approach allows precise classification of FV3-004R into one of the three temporal classes, providing insights into its potential role during viral replication.

How might FV3-004R contribute to viral pathogenesis and what experimental designs could test this?

Though FV3-004R remains uncharacterized, investigating its potential role in viral pathogenesis requires systematic experimental approaches:

  • Gene Knockout Studies:

    • Generate FV3 variants with FV3-004R deletions or mutations using reverse genetics

    • Compare replication kinetics in cell culture

    • Assess virulence in amphibian models

    • Measure viral loads in different tissues

  • Host Response Analysis:

    • Expose host cells to purified recombinant FV3-004R

    • Perform RNA-seq to identify transcriptional changes

    • Analyze immune signaling pathway activation/suppression

    • Compare with responses to whole virus infection

  • Cellular Localization and Trafficking:

    • Create fluorescently-tagged FV3-004R constructs

    • Track protein localization during infection

    • Identify potential co-localization with cellular organelles

    • Examine timing of expression relative to pathogenic events

  • Interaction with Virulence Factors:

    • Investigate potential interactions with known ranavirus virulence genes

    • Compare expression patterns with US22 family proteins that counter antiviral responses

    • Analyze co-expression networks during infection

  • Recombinant Virus Studies:

    • Create chimeric viruses with FV3-004R variants from different isolates

    • Test for altered host range or tissue tropism

    • Examine differences in replication efficiency

    • Assess impact on host mortality rates

  • Comparative Analysis Across Isolates:

    • Compare FV3-004R sequences from isolates with different virulence profiles

    • Identify potential correlations between sequence variations and pathogenicity

    • Test hypotheses using recombinant proteins or viruses

The experimental design should include appropriate controls and multiple host cell types or species to account for potential host-specific effects.

What role might FV3-004R play in evading host immune responses, and how can this be investigated?

Despite being uncharacterized, several experimental approaches can determine if FV3-004R plays a role in immune evasion:

  • Protein Localization During Infection:

    • Express tagged FV3-004R in infected cells

    • Determine if it localizes to immune signaling compartments

    • Track potential co-localization with immune receptors or adaptors

  • Host Protein Interaction Screening:

    • Perform yeast two-hybrid or AP-MS screens against immune signaling proteins

    • Validate interactions using co-immunoprecipitation

    • Determine functional consequences of identified interactions

  • Immune Signaling Pathway Analysis:

    • Express FV3-004R in reporter cell lines for key immune pathways (NF-κB, IRF3, etc.)

    • Stimulate cells with immune agonists and measure pathway inhibition

    • Compare with known viral immune antagonists

  • Comparative Analysis with Immune Evasion Genes:

    • Compare sequence features with known ranavirus immune antagonists

    • Look for similarity to US22 family proteins that counter antiviral responses

    • Investigate if FV3-004R is conserved in more virulent recombinant strains

  • Immunological Assays:

    • Measure cytokine responses in presence/absence of FV3-004R

    • Analyze effects on antigen presentation pathways

    • Test impact on interferon production or signaling

  • Temporal Expression Analysis:

    • Correlate FV3-004R expression timing with immune response dynamics

    • Determine if it's expressed early like other immune evasion genes

    • Analyze if expression changes in response to immune activation

A comprehensive approach using these methodologies would help elucidate any potential role of FV3-004R in immune evasion strategies employed by FV3.

How can researchers validate the function of FV3-004R using CRISPR-Cas9 genome editing of the viral genome?

Using CRISPR-Cas9 to edit the FV3 genome and validate FV3-004R function requires a careful experimental design:

  • Guide RNA Design and Validation:

    • Design multiple guide RNAs targeting the FV3-004R locus

    • Test guide RNA efficiency in reporter systems

    • Ensure specificity by checking for off-target sites in the FV3 genome

  • Viral Genomic Modification Strategies:

    • Knockout Approach: Create complete gene deletions or frameshift mutations

    • Tagging Approach: Insert epitope tags or fluorescent proteins for localization studies

    • Point Mutations: Introduce specific amino acid changes to test functional hypotheses

    • Promoter Modification: Alter expression timing to test temporal importance

  • Delivery System Optimization:

    • Transfect CRISPR-Cas9 components into permissive cells

    • Infect with wild-type FV3

    • Harvest and screen for edited viruses

  • Mutant Virus Screening and Isolation:

    • PCR and sequencing to identify desired mutations

    • Plaque purification to isolate clonal mutant viruses

    • Verify genome integrity beyond the target site

  • Phenotypic Characterization:

    • Growth Curves: Compare replication kinetics with wild-type virus

    • Cell Tropism: Test infection efficiency in different cell types

    • Virulence Assays: Assess pathogenicity in suitable amphibian models

    • Transcriptome Analysis: Examine global changes in viral/host gene expression

  • Complementation Studies:

    • Express FV3-004R in trans to rescue mutant phenotypes

    • Create domain-specific mutants for structure-function analysis

    • Test cross-complementation with homologs from related ranaviruses

  • Data Analysis and Interpretation:

    Analysis TypeWild-type FV3FV3-004R KnockoutFV3-004R Point Mutant
    Viral Titer (log10 PFU/ml)[baseline][observed change][observed change]
    Plaque Size (mm)[baseline][observed change][observed change]
    Time to CPE (hours)[baseline][observed change][observed change]
    Host Gene Expression[baseline pattern][differential pattern][differential pattern]
    Viral Gene Expression[baseline pattern][differential pattern][differential pattern]

This comprehensive approach would provide robust evidence for the function of FV3-004R in the viral life cycle.

What statistical approaches are most appropriate for analyzing experimental data related to FV3-004R function?

When analyzing experimental data related to FV3-004R function, researchers should consider these statistical approaches:

  • For Growth Curve Analysis:

    • Area under the curve (AUC) calculations

    • Repeated measures ANOVA for time course differences

    • Non-linear regression for growth rate parameters

    • Sample data structure:

    Time (h)WT FV3 (log10 PFU/ml)FV3-004R Mutant (log10 PFU/ml)p-value
    02.3 ± 0.22.3 ± 0.2n.s.
    124.7 ± 0.33.9 ± 0.4<0.05
    246.8 ± 0.45.2 ± 0.5<0.01
    487.5 ± 0.36.1 ± 0.4<0.01
    727.3 ± 0.46.0 ± 0.5<0.01
  • For Protein Interaction Studies:

    • Significance Analysis of INTeractome (SAINT) for AP-MS data

    • Permutation tests for co-immunoprecipitation enrichment

    • Calculation of enrichment factors with confidence intervals

  • For Transcriptomics/Proteomics:

    • Differential expression analysis (DESeq2, limma)

    • Gene Set Enrichment Analysis (GSEA)

    • Pathway analysis with multiple testing correction

    • Volcano plot visualization of significant changes

  • For Functional Assays:

    • Student's t-test or ANOVA with appropriate post-hoc tests

    • Non-parametric alternatives when normality cannot be assumed

    • Effect size calculations (Cohen's d or similar)

  • For Recombination Analysis:

    • Statistical tests implemented in recombination detection software

    • Phylogenetic incongruence tests

    • Breakpoint distribution analysis

    • Multiple test correction (FDR or Bonferroni)

  • For Replication Studies:

    • Meta-analysis approaches for combining multiple experiments

    • Power analysis for determining adequate sample sizes

    • Bayesian approaches for updating confidence with new data

Each analytical approach should include appropriate controls, biological replicates (minimum n=3), and clear reporting of statistical methods, significance thresholds, and effect sizes.

How can researchers integrate data from multiple experimental approaches to build a comprehensive model of FV3-004R function?

Integrating data from diverse experimental approaches to model FV3-004R function requires a systematic multi-omics strategy:

  • Data Integration Framework:

    • Establish a centralized database for all FV3-004R experimental data

    • Standardize data formats for cross-experiment comparability

    • Implement data quality control and normalization procedures

  • Multi-omics Data Fusion:

    • Integrate genomics, transcriptomics, proteomics, and interactomics data

    • Use computational approaches like Similarity Network Fusion (SNF)

    • Apply machine learning for pattern discovery across datasets

    • Example integration table:

    Data TypeObservationConfidenceSupporting Evidence
    LocalizationMembrane-associatedHighFluorescence microscopy, fractionation studies
    InteractomeBinds host protein XMediumAP-MS, Y2H, co-IP validation
    ExpressionIE gene classHighRT-PCR, microarray, CHX treatment
    StructureContains transmembrane domainMediumPrediction algorithms, CD spectroscopy
    FunctionInhibits pathway YMediumReporter assays, knockout phenotype
  • Network Analysis Approaches:

    • Construct protein-protein interaction networks

    • Identify functional modules and pathways

    • Map FV3-004R within the viral-host interaction landscape

    • Predict functional roles based on network position

  • Temporal Dimension Integration:

    • Align data across infection time points

    • Create dynamic models of FV3-004R activity

    • Correlate expression timing with functional events

  • Comparative Analysis Framework:

    • Integrate data from FV3-004R homologs in related viruses

    • Compare recombinant variants with different functions

    • Build evolutionary context for functional predictions

  • Validation Strategy:

    • Design experiments to test model predictions

    • Implement iterative cycles of prediction and validation

    • Quantify model confidence and identify knowledge gaps

  • Visual and Computational Representation:

    • Develop interactive visualizations of integrated data

    • Create computational models of FV3-004R function

    • Update models as new data becomes available

This integrated approach transforms isolated experimental findings into a comprehensive functional model, revealing emergent properties not apparent from individual datasets.

What are the key challenges in interpreting recombination data for FV3 genes like FV3-004R, and how can researchers address them?

Interpreting recombination data for FV3 genes presents several key challenges that researchers must address through careful methodological approaches:

  • Distinguishing Recombination from Convergent Evolution:

    • Challenge: Similar sequences may arise through convergent evolution rather than recombination

    • Solution: Use multiple recombination detection methods and require consensus across algorithms

    • Analysis Approach: Compare evolutionary rates across different gene regions and examine spatial patterns of sequence similarity

  • Breakpoint Precision:

    • Challenge: Determining exact recombination breakpoints can be difficult

    • Solution: Implement bootscanning methods with small window sizes and perform sensitivity analyses

    • Analysis Approach: Create breakpoint distribution plots with confidence intervals

  • Functional Interpretation of Recombinants:

    • Challenge: Understanding how recombination affects protein function

    • Solution: Map recombination breakpoints onto protein domain structures

    • Analysis Approach: Calculate FV3/CMTV ratios for each recombinant ORF and correlate with phenotypic changes

  • Sampling Bias:

    • Challenge: Limited sampling may miss key recombinant lineages

    • Solution: Include diverse geographical and temporal sampling

    • Analysis Approach: Perform rarefaction analysis to estimate sampling completeness

  • Recombination vs. Sequencing Artifacts:

    • Challenge: Sequencing errors or assembly issues may mimic recombination signals

    • Solution: Use high-quality sequencing with sufficient depth and validate unusual patterns

    • Analysis Approach: Apply quality filters and compare results across technical replicates

  • Temporal Dynamics of Recombination:

    • Challenge: Understanding when recombination events occurred

    • Solution: Implement Bayesian dating methods and construct temporally-aware phylogenies

    • Analysis Approach: Create time-scaled trees and map recombination events onto chronological frameworks

  • Distinguishing Direct vs. Indirect Recombination:

    • Challenge: Determining if recombination occurred directly between two strains or through intermediates

    • Solution: Sample potential intermediate hosts and perform coalescent analyses

    • Analysis Approach: Network-based representations of recombination patterns rather than simple trees

A robust analysis workflow addressing these challenges would include:

  • High-quality genome sequencing of diverse isolates

  • Application of multiple recombination detection algorithms

  • Statistical validation with appropriate null models

  • Mapping of recombination breakpoints to protein structures

  • Functional testing of recombinant variants

  • Integration with epidemiological and ecological data

This comprehensive approach would provide a more accurate picture of recombination affecting FV3-004R and its potential functional consequences.

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.