ASFV P30

African Swine Fever Virus P30
Shipped with Ice Packs
In Stock

Description

Introduction

ASFV P30 (African Swine Fever Virus p30 protein) is a critical structural phosphoprotein encoded by the CP204L gene of ASFV, a highly virulent pathogen causing hemorrhagic fever in domestic pigs and wild boars. With a molecular weight of ~30 kDa, P30 is expressed early during infection and exhibits strong antigenicity, making it a key target for diagnostic assays and host-virus interaction studies . Its roles span viral internalization, immune evasion, and modulation of host cellular processes .

Key Features:

  • Gene: CP204L

  • Expression: Early-phase synthesis (detected 2–4 hours post-infection), persisting throughout infection

  • Localization: Membrane-associated, secreted extracellularly during infection

  • Post-translational modifications: Phosphorylated at serine residues, with predicted glycosylation sites

Functional Roles:

FunctionMechanismSupporting Evidence
Viral EntryFacilitates ASFV internalization via host cell membrane interactionsAntibodies against P30 inhibit >95% viral entry
Immune ModulationTriggers early antibody response (detectable by 8–12 days post-infection)Dominant antigen in ELISA assays
Host Protein InteractionBinds to DAB2, RPSA, OAS1, PARP9, CAPG, ARPC5, and VBP1, influencing endocytosis and cytoskeletal dynamicsYeast two-hybrid and co-IP validation

Comparative Performance of P30-Based Assays:

MethodSensitivityTime to DetectionKey Advantage
P30-qPCRCt = 22.14 ± 4.51 (vs. P72-qPCR: 24.04 ± 3.84) 3–5 days post-infection (dpi)Earlier detection in lymphoid tissues
P30-IHC85% detection at 3 dpi (vs. 40% for P72-IHC) 3 dpi in spleen, 5 dpi in lymph nodesSuperior antigen visibility in acute lesions
ELISA100% seroconversion by 15 dpi 8–12 dpiHigh-throughput screening for antibodies
Aptamer AssaysLimit of detection (LOD) = 0.61 ng/mL <24 hoursNo cross-reactivity with PRRSV or IgG

P30-Host Protein Interactions:

Host ProteinBiological RoleInteraction Confirmed Via
DAB2Endocytosis regulationYeast two-hybrid, GST pulldown
RPSARibosomal functionsCo-immunoprecipitation
OAS1Antiviral innate immunityLaser confocal microscopy
PARP9DNA damage repairCo-IP validation
CAPGActin cytoskeleton remodelingGST pulldown

Functional enrichment analysis links these interactions to pathways in endocytosis (GO:0006897), actin regulation (KEGG:04810), and interferon signaling .

Therapeutic and Vaccine Implications

  • Antibody Neutralization: Anti-P30 antibodies inhibit viral entry but fail to confer full protection, suggesting complementary targets are needed .

  • Aptamer Development: High-affinity aptamers (Kd = 140 ± 10 pM) enable rapid, low-cost biosensors for field diagnostics .

  • Subunit Vaccines: P30’s immunogenicity supports its use in multi-antigen formulations, though alone it insufficiently induces neutralizing immunity .

Evolutionary and Epidemiological Significance

P30 sequences show high conservation across ASFV isolates, with <5% amino acid variability in global strains . This stability enhances its reliability as a diagnostic marker in diverse outbreaks, including recent epidemics in Asia and Europe .

Research Gaps and Future Directions

  • Mechanistic Studies: Clarify P30’s role in immune evasion via PARP9 and OAS1 interactions .

  • Field Validation: Scale aptamer-based assays for point-of-care use in resource-limited settings .

  • Multi-Antigen Strategies: Combine P30 with CD2v or MGF505 proteins to improve diagnostic specificity and vaccine efficacy .

Product Specs

Introduction
African swine fever virus (ASFV) causes African swine fever (ASF), a highly contagious and deadly disease affecting pigs. ASFV is a large DNA virus classified under the Asfarviridae family. Its complex virion structure comprises up to 54 polypeptides, including the early viral protein p30. Encoded by the CP204L gene, ASFV P30 is a major structural protein and plays a crucial role in the virus's entry mechanism.
Description
This recombinant African Swine Fever Virus P30 protein, expressed in E. coli, consists of 194 amino acids (40-315 aa) and has a molecular weight of 22kDa. A 6xHis tag is fused to the protein to facilitate purification, which is carried out using a proprietary chromatographic technique.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The African Swine Fever Virus P30 protein solution is formulated in 25mM K2CO3 and PBS.
Stability
For short-term storage (2-4 weeks), the protein can be stored at 4°C. For extended storage, it is recommended to freeze the protein at -20°C. Adding a carrier protein such as 0.1% HSA or BSA is advisable for long-term storage. Avoid repeated freezing and thawing.
Purity
The purity of the protein is greater than 90%, as determined by 10% SDS-PAGE analysis with Coomassie blue staining.
Source

Escherichia Coli.

Q&A

What is ASFV P30 and what role does it play in viral infection?

ASFV P30 is a structural membrane phosphoprotein encoded by the CP204L gene with a molecular weight of approximately 30 kDa. As a phosphorylated protein, P30 likely plays regulatory roles in signal transduction during infection . P30 is expressed during the early stages of viral infection, appearing on the infected cell membrane shortly after infection, and can be secreted outside the cell during the infection process .

Research methodologies for studying P30's role in viral infection typically involve protein expression systems, immunofluorescence assays, and viral inhibition studies with specific antibodies targeting P30 epitopes.

How does P30 expression compare chronologically to other ASFV structural proteins?

P30 exhibits a distinct chronological expression pattern compared to other ASFV structural proteins, particularly P72. Experimental data from qRT-PCR and immunohistochemistry (IHC) analyses have demonstrated significant differences in expression levels between P30 and P72, especially during the early stages of infection when acute ASF lesions develop .

Studies have revealed that P30 protein can be detected within 2-4 hours after infection, whereas P72 protein is typically detected around 24 hours post-infection . This differential expression has been confirmed at both transcriptional (mRNA) and translational (protein) levels, with a significant positive correlation between the two levels .

The chronological expression differences are particularly prominent in lymphoid tissues. For example, in experimentally infected pigs, the number of ASFV-positive cells based on P30-IHC was significantly higher than P72-IHC at early infection stages (3 dpi in spleen, 5 dpi in lymph node, and 5-7 dpi in thymus) . This chronological difference in protein expression provides valuable insights for developing early detection methods.

To study these chronological differences, researchers typically employ time-course experiments with synchronized infections, followed by molecular and immunological detection methods to track the appearance and accumulation of different viral proteins.

What are the key antigenic regions of P30 and their significance?

P30 contains four distinct antigenic regions that have been defined through extensive monoclonal antibody (mAb) studies. Research involving 21 mAbs against ASFV P30 has identified at least 4 linear epitopes distributed across these regions . Among these, antigenic regions 3 and 4 are particularly significant as they are highly conserved and immunodominant in host antibody responses .

Nine of 14 mAbs that mapped to antigenic regions 3 and 4 demonstrated reactivity with P30 across multiple serological methods, including indirect immunofluorescence assay (IFA), ELISA, and Western blot . This consistent reactivity across different detection platforms makes these regions valuable targets for diagnostic test development.

The antigenic properties of P30 are essential for improving P30-based serologic tests. Current P30 protein-based enzyme-linked immunosorbent assays (ELISAs) are widely used for ASFV antibody screening, but their sensitivity is not comparable to the more complex immunoperoxidase test (IPT) . Understanding these antigenic regions provides a foundation for developing enhanced diagnostic tools with improved sensitivity.

Methodologically, researchers map these antigenic regions through epitope mapping techniques including peptide arrays, phage display libraries, and competitive binding assays with characterized monoclonal antibodies.

How can P30-based detection methods be optimized for early diagnosis of ASFV?

The early expression of P30 during ASFV infection makes it an excellent target for developing highly sensitive early detection methods. P30-qPCR targeting the CP204L gene has demonstrated superior effectiveness compared to conventional P72-qPCR in detecting ASFV at the onset of infection . The optimization of such detection methods involves several methodological considerations:

First, primer and probe design is critical. Researchers should target highly conserved regions within the CP204L gene to ensure broad detection across various ASFV strains. Multiplex qPCR approaches that simultaneously target both P30 and P72 can provide complementary information and increase detection reliability.

Second, sample selection significantly impacts detection sensitivity. Studies have shown that P30 is detected earlier and with lower cycle threshold (Ct) values in lymphoid tissues, particularly in the spleen and lymph nodes . In a comparative study of experimentally infected pigs, P30-qPCR identified ASFV in spleen and lymph nodes of pigs at 3- and 5-days post-infection when P72-qPCR results were still negative . The table below summarizes the comparative detection efficacy:

Time Post-InfectionTissue TypeP30-qPCR DetectionP72-qPCR Detection
1 dayLymphoidNegativeNegative
3 daysSpleenPositive (3/3 pigs)Negative
3 daysLymph nodesPositive (3/3 pigs)Negative
5 daysSpleenPositive (4/4 pigs)Negative
5 daysLymph nodesPositive (4/4 pigs)Negative
7-9 daysAll tissuesPositivePositive

Third, validation with field samples is essential. In a field outbreak study, P30-qPCR demonstrated significantly lower Ct values (22.14 ± 4.51) compared to P72-qPCR (24.04 ± 3.84) when testing blood samples from clinically infected pigs , confirming its superior sensitivity in real-world scenarios.

For immunological detection, focusing on antigenic regions 3 and 4 of P30 can enhance the sensitivity of antibody-based assays. These regions show consistent reactivity across different serological methods and are immunodominant in host responses .

What host cell proteins interact with P30, and what are the implications for ASFV pathogenesis?

P30 interacts with several host cell proteins, forming a complex interaction network that influences viral entry, replication, and immune evasion. A membrane yeast two-hybrid system study identified seven porcine alveolar macrophage (PAM) cellular proteins that interact with P30 .

These interactions have been verified through multiple experimental approaches. GST-pull down assays confirmed that GST-tagged P30 directly binds to DAB2, OAS1, RPSA, PARP9, and VBP1, while interactions with CAPG and ARPC5 appear to be indirect . Colocalization studies using confocal microscopy in both transfected cells and ASFV-infected cells further validated these interactions .

The functional implications of these interactions are significant for understanding ASFV pathogenesis:

  • Viral Entry Mechanism: P30 interacts with DAB2 and RPSA on the cell membrane, potentially facilitating ASFV endocytosis. DAB2 recruits clathrin, mediating clathrin-dependent endocytosis .

  • Cytoskeletal Rearrangement: Interactions with ARPC5 and CAPG suggest P30 may trigger actin rearrangement, facilitating macropinocytosis as an alternative entry pathway .

  • Immune Evasion: P30's interaction with DAB2 may inhibit TLR4 entry into endosomes, while its binding to PARP9 could inhibit PI3K/AKT3 activation, ultimately reducing type I interferon production .

  • Viral Replication Enhancement: By interacting with RPSA, P30 may eliminate RPSA's inhibitory effect on the MAPK pathway, potentially promoting viral replication .

  • Antiviral Response Modulation: P30's interaction with OAS1 may prevent RNase L-dependent or independent antiviral responses .

Methodologically, these interactions can be studied through techniques such as co-immunoprecipitation, proximity ligation assay, FRET/BRET analyses, and targeted mutagenesis to disrupt specific interaction domains.

How does P30's RNase-like activity contribute to viral evasion of host immunity?

Recent research has identified a novel RNA-destroying function in the P30 protein, suggesting an additional mechanism by which ASFV may evade host defenses . This RNase-like activity represents a significant finding that may explain aspects of ASFV's ability to manipulate host cell functions and evade immune responses.

The RNA-degrading activity of P30 likely targets host cell RNA, potentially disrupting cellular gene expression and immune signaling pathways. This function may be particularly important in countering the host's innate antiviral responses that rely on RNA-sensing pathways and interferon production .

Methodologically, studying this RNase-like activity involves several experimental approaches:

  • In vitro RNA degradation assays: Purified recombinant P30 protein can be incubated with various RNA substrates to assess its degradation activity and substrate specificity.

  • Mutational analysis: Generating P30 variants with mutations in predicted catalytic domains can help identify the specific residues responsible for the RNase activity.

  • Cellular RNA analysis: RNA-seq and other transcriptomic approaches can be used to identify specific host RNA targets in P30-expressing cells compared to controls.

  • Functional immunology assays: Measuring the impact of wild-type versus RNase-deficient P30 on interferon production, NF-κB activation, and other immune signaling pathways can elucidate the functional consequences of this activity.

While significant progress has been made in characterizing this activity, critical questions remain unanswered, including the precise role of P30's RNase activity in ASFV infection in pigs and whether it targets specific host RNA molecules . Future research should focus on these aspects to fully understand the contribution of this function to ASFV pathogenesis.

How can P30 research inform the development of improved diagnostic tools for ASFV?

P30 research offers several avenues for developing enhanced diagnostic tools for ASFV detection. The early expression of P30 during infection and its distinct antigenic properties make it particularly valuable for diagnostic applications.

For molecular diagnostics, targeting the CP204L gene encoding P30 provides earlier detection capability compared to conventional P72-based methods. The p30-qPCR assay has demonstrated superior sensitivity in detecting ASFV at the onset of infection, with studies showing significantly lower Ct values compared to p72-qPCR in both experimentally infected animals and field samples . This improved sensitivity is critical for early intervention during outbreaks.

For serological diagnostics, focusing on the immunodominant antigenic regions 3 and 4 of P30 can enhance the sensitivity of antibody-based assays . While P30-based ELISAs are already widely used for ASFV antibody screening, their sensitivity remains lower than the immunoperoxidase test (IPT) . Knowledge of these specific antigenic domains can guide the development of next-generation ELISAs with improved sensitivity.

Methodologically, researchers can approach diagnostic development through:

  • Multiplex PCR systems: Combining P30 and P72 targets in a single assay to provide complementary detection capabilities.

  • Peptide-based ELISAs: Focusing on specific epitopes within antigenic regions 3 and 4 rather than using the whole P30 protein.

  • Point-of-care testing: Adapting P30-based detection to rapid test formats suitable for field use, such as lateral flow assays targeting P30 antigens or antibodies.

  • Sequential testing algorithms: Implementing P30-qPCR as an initial screening tool followed by P72-qPCR confirmation to optimize detection sensitivity while maintaining specificity.

What experimental approaches can elucidate the differences in P30 expression between ASFV strains of varying virulence?

The differential expression of P30 may contribute to variations in virulence among ASFV strains. Experimental approaches to investigate these differences require systematic comparisons across strains with varying virulence profiles.

Time-course experiments comparing P30 expression kinetics in cells infected with different ASFV strains are fundamental to this research. Previous findings suggest that the timing of maximal P30 expression may vary significantly between virulent and attenuated strains, with a delay of 2-4 days observed in attenuated viral infections compared to virulent infections .

A comprehensive experimental approach would include:

  • In vitro expression studies: Infecting susceptible cell lines (like porcine macrophages) with different ASFV strains and analyzing P30 expression at multiple time points using qRT-PCR and Western blot.

  • In vivo comparative studies: Experimentally infecting pigs with virulent versus attenuated ASFV strains and collecting tissues at defined intervals to assess P30 expression patterns using qPCR, IHC, and proteomic approaches.

  • Promoter activity analysis: Cloning the CP204L promoter regions from different strains and measuring their activity using reporter assays to identify regulatory differences that might influence expression timing and levels.

  • Viral mutants: Generating recombinant viruses with altered P30 expression characteristics to directly test the relationship between P30 expression patterns and virulence.

It is important to note that strain-dependent differences may be more pronounced in cases of subacute or chronic ASF compared to acute ASF. As highlighted in one study, "significant differences between p30 and p72 expression were found only in the early stage of acute ASF, different results may appear in experiments using lower virulent strains causing subacute or chronic ASF" . Therefore, experimental designs must incorporate strains representing the full spectrum of disease manifestations.

How might P30's role in ASFV infection inform novel therapeutic approaches?

Understanding P30's multifunctional role in ASFV infection opens several avenues for therapeutic intervention. Based on the current knowledge of P30's functions, several therapeutic strategies can be explored:

  • Entry Inhibitors: Since P30 is essential for virus internalization, with anti-P30 antibodies capable of inhibiting over 95% of viral entry , developing small molecule inhibitors or peptide mimetics that block P30-mediated entry represents a promising approach. These could target either P30 directly or its interactions with host receptors like DAB2 and RPSA.

  • Disruption of Host-Protein Interactions: The identified interactions between P30 and host proteins (DAB2, OAS1, RPSA, PARP9, VBP1) provide specific targets for therapeutic intervention . Compounds that disrupt these interactions could potentially inhibit viral replication or enhance host immune responses.

  • RNase Activity Inhibition: P30's RNA-destroying function could be targeted with specific inhibitors, potentially restoring normal host cell RNA processing and immune signaling .

  • Enhanced Vaccines: Knowledge of P30's immunodominant epitopes, particularly in antigenic regions 3 and 4 , can guide the design of subunit or epitope-based vaccines that elicit more effective neutralizing antibody responses.

Methodological approaches to developing these therapeutics include:

  • High-throughput screening: Testing compound libraries for inhibitors of P30 functions or P30-host protein interactions.

  • Structure-based drug design: Using structural information about P30 and its interaction domains to design targeted inhibitors.

  • Peptide therapeutics: Developing peptides that mimic binding interfaces to competitively inhibit P30-host protein interactions.

  • RNA aptamer technology: Designing aptamers that specifically bind to P30 and neutralize its functions.

  • Combination approaches: Targeting multiple viral functions simultaneously to minimize the development of resistance.

While these approaches hold promise, it's important to note that effective translation to clinical applications requires systematic evaluation in both cell culture and animal models to assess efficacy, safety, and the potential for resistance development.

What methodological approaches are most effective for studying P30's multiple functions in different experimental systems?

Investigating P30's diverse functions requires a multi-faceted experimental approach combining molecular, cellular, and in vivo techniques. Based on current research, the following methodological strategies have proven effective:

For studying P30's role in viral entry and host cell interactions:

  • Recombinant protein systems: Expression of GST-tagged P30 in E. coli for pull-down assays has successfully identified direct interactions with host proteins such as DAB2, OAS1, RPSA, PARP9, and VBP1 .

  • Co-localization studies: Confocal microscopy using co-transfection of plasmids expressing HA-tagged host proteins and EGFP-P30 has confirmed the spatial overlap of P30 with interacting partners in cellular contexts .

  • Viral internalization assays: Pretreatment with anti-P30 antibodies followed by quantification of viral entry provides direct evidence of P30's role in ASFV internalization .

For analyzing P30's expression kinetics:

  • Time-course experiments: Comparing P30 and P72 expression at defined intervals post-infection (1, 3, 5, 7, and 9 days) in experimentally infected animals provides comprehensive data on expression patterns .

  • Multi-technique validation: Combining qRT-PCR for transcriptional analysis with immunohistochemistry for protein detection ensures robust verification of expression patterns .

  • Tissue-specific analysis: Examining multiple tissues (spleen, lymph nodes, thymus) separately reveals important differences in expression patterns across anatomical sites .

For investigating P30's antigenic properties:

  • Monoclonal antibody panels: Developing comprehensive mAb collections targeting different P30 epitopes enables detailed mapping of antigenic regions .

  • Multi-format serological testing: Testing reactivity across different platforms (IFA, ELISA, Western blot) ensures identification of consistently reactive epitopes suitable for diagnostic applications .

For studying P30's RNase-like activity:

  • RNA degradation assays: In vitro assays with purified recombinant P30 and defined RNA substrates provide direct evidence of enzymatic activity .

  • Cell-based RNA stability assays: Comparing RNA profiles in cells expressing wild-type versus mutant P30 can reveal the impact of this activity in cellular contexts .

Each experimental system presents distinct advantages and limitations. In vitro approaches offer controlled conditions for mechanistic studies but may not fully recapitulate the complexity of in vivo infection. Animal models provide physiologically relevant contexts but involve greater ethical considerations and technical challenges. Balancing these approaches is essential for comprehensive understanding of P30's functions.

What are the key challenges in translating P30 research findings into practical applications?

Translating P30 research into practical applications faces several significant challenges that researchers must address:

  • Strain Variation: ASFV exhibits considerable genetic diversity, potentially affecting P30 sequence, structure, and function across strains. Research has focused primarily on reference strains, and the degree to which findings translate to field isolates remains uncertain. Studies comparing P30 across diverse strains are needed to ensure broad applicability of any P30-based diagnostics or therapeutics.

  • Host Species Differences: While domestic pigs are the primary research focus, ASFV also infects wild boars and various African wild suids. Potential variations in P30-host protein interactions across these species could influence diagnostic sensitivity or therapeutic efficacy. Comparative studies examining P30 function in different host species would address this challenge.

  • Complexity of P30's Multiple Functions: P30's involvement in viral entry, immune evasion, and potential RNA degradation creates challenges for targeting specific functions therapeutically without disrupting others. This complexity necessitates careful design of intervention strategies to achieve desired outcomes without unintended consequences.

  • Technical Limitations in Field Settings: While P30-qPCR shows superior early detection capabilities compared to P72-qPCR , implementing such molecular techniques in resource-limited field settings presents logistical challenges. Adapting P30-based detection to point-of-care formats suitable for field use requires significant technological innovation.

  • Incomplete Understanding of P30's Role in Pathogenesis: Despite progress in characterizing P30's interactions and functions, significant knowledge gaps remain. For example, the precise role of P30's RNase activity in ASFV infection in pigs and whether it targets specific host RNA molecules are still unknown . These gaps complicate the development of targeted interventions.

  • Integration with Other ASFV Proteins: P30 functions as part of a complex viral system, and its activities may depend on interactions with other viral proteins. Research approaches that consider P30 in isolation may miss important contextual factors that influence its behavior during actual infection.

Addressing these challenges requires multidisciplinary collaboration among virologists, immunologists, structural biologists, and bioengineers to develop comprehensive approaches that account for the complexity of ASFV biology while delivering practical solutions for diagnosis, prevention, and control.

What emerging technologies could advance our understanding of P30's role in ASFV pathogenesis?

Several cutting-edge technologies hold promise for deepening our understanding of P30's multifaceted roles in ASFV pathogenesis:

CRISPR-Cas9 Genome Editing: Precise editing of the CP204L gene in the viral genome could create mutant viruses with altered P30 expression or function. This approach would allow direct assessment of how specific P30 domains contribute to viral replication, host interactions, and pathogenesis. Additionally, CRISPR-based screening of host factors could identify previously unknown cellular proteins that interact with P30.

Cryo-Electron Microscopy: High-resolution structural analysis of P30 alone and in complex with its binding partners could reveal critical interaction interfaces. Understanding the three-dimensional organization of P30 within the virion and at the cell membrane during entry would provide insights into its functional mechanisms.

Proteomics and Interactomics: Advanced mass spectrometry techniques, including proximity labeling methods like BioID or APEX, could map the complete P30 interactome under different conditions. Temporal proteomics approaches would reveal how these interactions change throughout the viral life cycle.

Single-Cell RNA Sequencing: This technology could elucidate how P30 affects gene expression in individual cells during infection, potentially revealing cell-type-specific responses and heterogeneity in host cell reactions to P30-mediated processes.

Organoid Models: Porcine intestinal or lung organoids could provide more physiologically relevant systems for studying P30 function than traditional cell lines while avoiding some of the ethical and practical limitations of animal studies.

Nanobody Technology: Developing single-domain antibodies (nanobodies) against specific P30 epitopes could provide highly specific tools for functional inhibition studies and potentially for therapeutic applications.

In Situ Structural Analysis: Techniques like correlative light and electron microscopy (CLEM) could visualize P30 localization and interactions within infected cells with unprecedented precision, revealing spatiotemporal aspects of its function.

These technologies, particularly when used in combination, could address current knowledge gaps regarding P30's RNase activity specificity, the structural basis of its host protein interactions, and its precise role in modulating immune responses during infection.

How might comparative studies of P30 across different ASFV strains inform evolutionary perspectives on viral pathogenesis?

Comparative studies of P30 across diverse ASFV strains could provide valuable insights into the evolution of viral pathogenesis strategies and host adaptation mechanisms:

Sequence analysis of the CP204L gene across historical and contemporary ASFV isolates could reveal evolutionary patterns in P30 conservation versus diversification. Such studies would help identify which regions have remained stable over time (suggesting essential functions) versus those that show greater variability (potentially indicating adaptation to different hosts or immune pressures).

Methodologically, this research would benefit from:

  • Phylogenetic analysis: Constructing evolutionary trees based on CP204L sequences from diverse ASFV genotypes to map the historical development of P30 variants.

  • Structure-function correlations: Relating sequence variations to functional differences in P30 activities such as host protein binding, RNase activity, or antigenic properties.

  • Host adaptation studies: Comparing P30 from ASFV strains that primarily circulate in domestic pigs versus those from wild suids to identify adaptations to different host environments.

  • Experimental evolution approaches: Passaging ASFV through different host systems and tracking changes in the CP204L gene could reveal how selection pressures shape P30 evolution.

  • Recombinant protein comparisons: Expressing and functionally characterizing P30 variants from different strains to directly assess how sequence differences impact protein function.

These comparative approaches could reveal whether changes in P30 represent adaptive strategies that ASFV employs to optimize its replication in different host contexts or in response to evolving immune pressures. Understanding these evolutionary dynamics could inform predictions about how ASFV might continue to evolve and adapt to new geographical regions or host populations.

Product Science Overview

Introduction

African Swine Fever Virus (ASFV) is a highly contagious and lethal virus that affects domestic pigs and wild boars. It is the only member of the Asfarviridae family and is known for causing a hemorrhagic fever with mortality rates approaching 100% . ASFV is a large, complex virus with a double-stranded DNA genome that encodes numerous structural and non-structural proteins .

Structure and Function of P30

One of the key structural proteins of ASFV is P30, also known as CP204L. P30 is a membrane phosphoprotein that plays a crucial role in the virus’s ability to infect host cells . It is involved in the early stages of viral infection and is one of the most immunogenic proteins produced during ASFV infection . This makes P30 a valuable target for serological detection and surveillance of ASFV .

Interaction with Host Proteins

P30 interacts with several host cellular proteins, which suggests it may play a regulatory role in signal transduction and other cellular processes . Research has identified seven host proteins that interact with P30, including DAB2, RPSA, OAS1, PARP9, CAPG, ARPC5, and VBP1 . These interactions are believed to be involved in endocytosis, actin cytoskeleton regulation, and innate immunity .

Importance in ASFV Pathogenesis

The P30 protein is essential for the internalization of ASFV into host cells, making it a critical factor in the virus’s ability to establish infection . Its immunogenic properties also make it a key target for the development of diagnostic tools and potential vaccines .

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 2024 Thebiotek. All Rights Reserved.