The Ba71V-126 Antibody Pair refers to a reagent system targeting the African Swine Fever Virus (ASFV) inner membrane protein p54, derived from the attenuated BA71V strain. This antibody pair is primarily used in immunoassays for detecting ASFV antigens, leveraging the conserved nature of p54 across viral isolates. Below is a comprehensive analysis of its structure, applications, and research findings, supported by peer-reviewed studies and product specifications.
The antibody pair binds to p54 (Ba71V-126), a 20.94 kDa recombinant protein expressed in insect cells (Table 1). This protein is part of the ASFV envelope and plays a critical role in viral entry and replication .
Feature | Description |
---|---|
Protein Length | Met1-Leu183 |
Expression System | Insect cells |
Purity | >90% (SDS-PAGE) |
Applications | ELISA, Immunogen, Western Blotting, Bioactivity Testing |
The Ba71V-126 Antibody Pair is employed in ELISA kits to detect ASFV antigens in infected cells or tissues. Studies demonstrate its utility in:
ASFV p54 is a key target in subunit vaccine designs. For example, a blend of p30, p54, and p72 fused with bacterial lipoprotein OprI induced robust immune responses in pigs . Similarly, a DNA prime-recombinant vaccinia vector regime reduced viremia by 70% in challenged animals .
A conserved B-cell epitope (52DPLASQRDIYY62) on pB125R was identified as critical for immune recognition . While this epitope is distinct from p54, its discovery underscores the broader utility of ASFV epitope-based diagnostics and vaccines.
Proteomic analyses of p54 interactions revealed associations with host proteins like Rab5, Rab7, and PI4Kβ, highlighting its role in viral entry via macropinocytosis .
KEGG: vg:22220355
BA71V is an attenuated tissue-culture adapted strain of African Swine Fever Virus that has been extensively used in laboratory research. Unlike virulent field isolates such as the Georgia 2007/1 (GRG) strain, BA71V can be grown in cell culture systems rather than requiring primary macrophages. The key differences between BA71V and virulent isolates lie in their gene complement, particularly in multigene family (MGF) members, which are often implicated in host immune response suppression .
Transcriptome analysis has revealed that despite these genomic differences, the expression patterns of conserved genes between BA71V and virulent isolates show surprising similarities in both expression levels and temporal regulation. This indicates that virulence determinants are primarily driven by virulent isolate-specific genes rather than differential regulation of shared genes .
Several ASFV proteins serve as important antigenic targets for antibody development, including:
CD2v protein (EP402R gene product) - A heavily glycosylated membrane protein that mediates hemadsorption and is implicated in virulence and host range
p72 capsid protein (B646L) - The major structural protein and commonly used diagnostic target
p30 (CP204L) - An early protein frequently used for diagnostic purposes
p54 (E183L) - A membrane protein involved in virus attachment
The selection of appropriate antigens for antibody development depends on the research objectives. CD2v extracellular domain expressed in mammalian systems (like 293F cells) offers advantages for generating antibodies against conformational epitopes due to proper glycosylation .
Verification of antibody specificity requires multiple complementary approaches:
Verification Method | Application | Key Considerations |
---|---|---|
Western Blot (WB) | Confirms binding to denatured protein | May miss conformational epitopes |
Immunoperoxidase Monolayer Assay (IPMA) | Confirms binding to infected cells | Allows visualization of cellular localization |
Indirect Immunofluorescence Assay (IFA) | Confirms binding in transfected or infected cells | Good for colocalization studies |
ELISA | Quantitative binding assessment | Useful for measuring antibody titers |
Dot Blot | Rapid screening for epitope mapping | Useful with peptide arrays |
For comprehensive validation, test antibodies against both recombinant proteins and virus-infected cells. Additionally, include appropriate controls such as uninfected cells and isotype control antibodies to ensure specificity .
The choice of expression system significantly impacts the quality of antibodies generated against ASFV proteins:
Expression System | Advantages | Limitations | Best For |
---|---|---|---|
293F Mammalian System | Proper glycosylation and folding | Higher cost, lower yield | Membrane proteins like CD2v |
E. coli | High yield, cost-effective | Lacks post-translational modifications | Non-glycosylated proteins, epitope mapping |
Baculovirus | Intermediate yield and modifications | Insect glycosylation patterns differ | Structural proteins |
For heavily glycosylated proteins like CD2v, mammalian expression systems are strongly recommended. Research has shown that the 293F expression system provides proper glycosylation of the CD2v extracellular region, which is critical for generating antibodies that recognize native viral proteins . For non-glycosylated structural proteins, bacterial expression systems may be sufficient.
When designing transcriptome experiments comparing BA71V and virulent ASFV strains:
Standardize infection conditions: Use a high multiplicity of infection (MOI of ~5) to ensure synchronized infection cycles and minimize the complication of variable proportions of uninfected cells .
Select appropriate time points: Include both early (~5 hpi) and late (~16 hpi) time points to capture the temporal dynamics of gene expression .
Choose appropriate sequencing methodology: Cap Analysis of Gene Expression sequencing (CAGE-seq) allows precise mapping of transcription start sites and quantification of transcript levels .
Normalize data appropriately: For direct comparisons, use reads per million (RPM) to account for differences in sequencing depth across samples .
Validate key findings: Confirm RNA-seq results using qRT-PCR or protein expression analysis.
Data analysis should include both differential expression analysis for individual genes and cluster analysis to identify co-regulated gene groups .
Developing effective monoclonal antibodies against BA71V proteins requires careful attention to:
Immunogen design: Express the full extracellular domain or functional regions of membrane proteins in mammalian systems to ensure proper folding and glycosylation .
Immunization protocol: Use a prime-boost strategy with Freund's complete adjuvant for primary immunization, followed by incomplete adjuvant for boosters at 21 and 42 days .
Hybridoma screening strategy:
Clone selection criteria: Select hybridomas based on:
Antibody titer and affinity
Specificity in multiple assay formats
Recognition of native viral protein in infected cells
Stability in culture
Epitope mapping: Characterize the binding epitopes through truncation mutants or peptide arrays to understand antibody specificity .
Interpretation of ASFV gene expression patterns requires understanding of both temporal regulation and relative abundance:
Early vs. Late genes: Categorize genes based on expression patterns:
Expression clusters: More refined analysis reveals five distinct expression patterns:
Functional correlation: Genes with similar functions often share expression patterns. For example:
Strain comparisons: When comparing BA71V to virulent strains like GRG, focus on:
For comprehensive epitope mapping:
Initial mapping with truncated proteins:
Fine mapping with overlapping peptides:
Confirmation with point mutations:
Introduce alanine substitutions at key positions
Test impact on antibody binding
Identify critical contact residues
Structural context analysis:
Map epitope onto protein structure (if available)
Evaluate surface exposure and accessibility
Assess conservation across ASFV strains
For example, in studies with CD2v protein, researchers identified a linear epitope (154SILE157) using a combination of truncation constructs followed by testing with overlapping peptides in Dot-Blot, ELISA, and IFA tests .
Analysis of ASFV promoter architecture reveals distinct patterns that control gene expression timing:
Early Promoter Motifs (EPM):
Late Promoter Features:
Contextual analysis:
Examine the relationship between promoter sequences and expression timing
Compare motifs across different ASFV strains (BA71V vs. virulent isolates)
Correlate with functional gene categories
Biological implications:
Understanding these promoter elements is essential for predicting gene expression timing and designing experiments to target specific stages of the viral lifecycle.
When troubleshooting specificity issues:
Analyze potential cross-reactive targets:
Perform sequence homology searches
Test against related viral proteins
Include appropriate controls (uninfected cells, isotype controls)
Optimize assay conditions:
Adjust antibody concentration
Modify blocking reagents (try different blockers like BSA, milk, or commercial blockers)
Increase washing stringency (higher salt, or addition of detergents)
Epitope engineering approaches:
Select unique regions for immunization
Avoid conserved domains shared with host proteins
Consider affinity maturation techniques
Purification strategies:
Perform negative selection against cross-reactive antigens
Use epitope-specific affinity purification
Consider subclass switching if isotype may contribute to non-specific binding
For detailed temporal analysis:
High-resolution time course:
Nascent RNA labeling:
Use metabolic labeling with 4-thiouridine (4sU)
Capture newly synthesized RNA at each time point
Distinguish primary transcription from RNA accumulation
Single-cell approaches:
Apply scRNA-seq to capture cell-to-cell variability
Identify distinct infection phases within population
Correlate with viral protein expression by flow cytometry
Quantitative analysis frameworks:
For a comprehensive understanding, integrate transcriptomic data with measurements of viral genome replication, as the ratio of viral transcripts increases approximately 2-fold in GRG and 8-fold in BA71V from early to late infection, which correlates with a 15-fold increase in GRG genome copy numbers and a 30-fold increase in BA71V genomes .
Antibodies against BA71V proteins can be applied in multiple advanced research contexts:
Application | Methodology | Key Considerations |
---|---|---|
Live-cell imaging | Use Fab fragments or non-neutralizing antibodies | Confirm antibody doesn't interfere with infection |
Super-resolution microscopy | Direct fluorophore conjugation | Optimize signal-to-noise ratio |
Immunoprecipitation for protein interaction studies | Crosslink antibodies to beads | Test multiple lysis conditions |
ChIP-seq for viral protein-DNA interactions | Validate antibody specificity in ChIP | Optimize crosslinking conditions |
Viral neutralization assays | Compare neutralizing vs. non-neutralizing epitopes | Include isotype controls |
FACS for infected cell isolation | Confirm antibody access to target in intact cells | Consider fixation impact on epitopes |
Each application requires specific validation steps to ensure antibodies perform optimally in the selected experimental context.
Single-cell technologies offer powerful new insights into ASFV infection:
Single-cell RNA sequencing (scRNA-seq):
Reveals heterogeneity in cellular responses to infection
Identifies distinct cell states during infection progression
Allows precise temporal ordering of infection events
Spatial transcriptomics:
Maps infection dynamics within tissue context
Identifies local microenvironmental factors influencing infection
Correlates with immune cell infiltration patterns
CyTOF and spectral flow cytometry:
Enables high-dimensional profiling of protein expression
Allows simultaneous detection of multiple viral and host proteins
Reveals rare cell populations with distinct infection phenotypes
Integration with BA71V-126 antibody applications:
Use antibodies to sort infected cells at different stages
Combine protein and RNA detection in the same cells
Track infection progression at single-cell resolution
These approaches can address fundamental questions about why some cells support productive infection while others are resistant, even with standardized MOI.
Understanding ASFV promoter architecture enables several synthetic biology applications:
Engineered expression systems:
Design synthetic promoters with predictable temporal activity
Create attenuated virus strains with altered gene expression
Develop reporter viruses for tracking infection
Vaccine development strategies:
Modulate antigen expression timing and levels
Engineer strains with enhanced immunogenicity
Create temperature-sensitive variants through promoter modification
ASFV-based vector development:
Utilize ASFV promoters in heterologous expression systems
Develop large-capacity vectors for gene delivery
Create chimeric promoters with novel properties
The detailed understanding of early promoter motifs (EPM) with their conserved sequence signatures positioned 9-10 nucleotides upstream of the transcription start site provides a foundation for rational design of synthetic regulatory elements .