Recombinant African swine fever virus Virus attachment protein p12 (Mal-106)

Shipped with Ice Packs
In Stock

Description

Definition and Nomenclature

Recombinant ASFV p12 (Mal-106) is a laboratory-produced protein derived from the African swine fever virus, a highly contagious disease-causing virus in pigs. The protein is a 17 kDa (under non-reducing conditions) glycoprotein localized in the viral envelope, critical for initiating host cell attachment . Synonyms include Virus attachment protein p12 and Protein p12. The "Mal-106" designation refers to its origin from the Malawi/Lil 20-1/1983 virus isolate .

Key Properties

PropertyDetail
Molecular Weight17 kDa (non-reduced), 12–10 kDa (intermediate forms in infected cells)
Amino Acid SequenceMALDGSSGGGSNVETLLIVAIIVVIMAIMLYYFWWMPRQQQKKCSKAEECTCNNGSCSLK TS
LocalizationExternal envelope layer; requires partial disruption for epitope exposure
Purity>90% by SDS-PAGE
Source OrganismsE. coli (His-tagged), Spodoptera frugiperda insect cells

The protein’s structure includes a hydrophobic region (positions 1–62) and a glycosylated domain critical for receptor binding .

Mechanism of Action

  1. Attachment Initiation: p12 binds to host cell receptors, enabling virion adhesion .

  2. Receptor Specificity: Demonstrates preferential binding to Vero cells (susceptible) over L cells (insusceptible) .

  3. Inhibition of Infection: Recombinant p12 blocks viral attachment and infection in in vitro models .

Research Findings

  • Neutralization Limitations: Antibodies against p12 fail to neutralize ASFV or inhibit attachment in pigs, despite inducing immune responses .

  • Viral Isolate Variability: p12 inhibits infection across ASFV strains, including attenuated and virulent isolates .

Expression Systems

SystemYield & PurityAdvantages/Challenges
E. coliHigh yield (>90% purity), low costLacks post-translational modifications
Spodoptera frugiperda>50 mg/L, >10% total proteinsAuthentic glycosylation; complex scalability

Purification Steps

  1. Aqueous Phase Partitioning: Separates cytoplasmic proteins from insect cell debris .

  2. Immunoaffinity Chromatography: Uses p12-specific monoclonal antibodies for high-purity isolation .

Vaccine Development

  • Potential: Targeted disruption of p12-mediated attachment could prevent infection .

  • Challenges: Pigs immunized with recombinant p12 show no protective immunity, suggesting incomplete antigenic coverage .

Challenges and Limitations

  1. Antibody Efficacy: Despite inducing immune responses, anti-p12 antibodies fail to neutralize ASFV .

  2. Vaccine Design: p12 alone may not elicit broad immunity; combination with other antigens (e.g., p72, p54) may be required .

  3. Production Complexity: Insect cell systems yield authentic glycosylation but require specialized infrastructure .

Product Specs

Form
Lyophilized powder
Please note: We will prioritize shipping the format currently in stock. However, if you have a specific format preference, please indicate your requirement in the order notes, and we will fulfill your request if possible.
Lead Time
Delivery time may vary depending on the purchasing method and location. Please consult your local distributor for specific delivery timelines.
Note: All of our proteins are shipped with standard blue ice packs. If you require dry ice shipping, please inform us in advance as additional charges will apply.
Notes
Repeated freezing and thawing is not recommended. For optimal preservation, store working aliquots at 4°C for up to one week.
Reconstitution
We recommend briefly centrifuging the vial before opening to ensure the contents settle at the bottom. Reconstitute the protein in deionized sterile 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%, which can be used as a reference.
Shelf Life
Shelf life is influenced by various factors, including storage conditions, buffer composition, temperature, and the inherent stability of the protein.
Generally, the shelf life of liquid form is 6 months at -20°C/-80°C. For lyophilized form, the shelf life is 12 months at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquot for multiple uses to minimize freeze-thaw cycles.
Tag Info
Tag type is determined during the manufacturing process.
The specific tag type will be established during production. If you have a particular tag preference, please inform us, and we will prioritize its development if possible.
Synonyms
Mal-106; Virus attachment protein p12; Protein p12
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-62
Protein Length
full length protein
Species
African swine fever virus (isolate Tick/Malawi/Lil 20-1/1983) (ASFV)
Target Names
Mal-106
Target Protein Sequence
MALDGSSGGGSNVETLLIVAIIVVIMAIMLYYFWWMPRQQQKKCSKAEECTCNNGSCSLK TS
Uniprot No.

Target Background

Function
This protein serves as the virus attachment protein.
Protein Families
Asfivirus attachment protein p12 family
Subcellular Location
Virion membrane; Single-pass membrane protein.

Q&A

What is the ASFV p12 (Mal-106) protein and what is its origin?

The p12 protein is a virus attachment protein originating from African swine fever virus (ASFV), specifically from the isolate Tick/Malawi/Lil 20-1/1983. It functions as a transmembrane protein that promotes the adsorption of virus particles to host cells by binding to specific receptors on the host cell membrane, mediating ASFV entry into cells. The recombinant form of this protein can be produced in expression systems such as E. coli for research purposes, as indicated by its UniProt ID: P0C9Y2 . This protein represents one of the key viral components involved in the initial stages of ASFV infection, which is endemic to sub-Saharan Africa and maintained in nature through cycles between ticks and wild pigs, bushpigs, and warthogs.

How does p12 function in the viral infection cycle?

The p12 protein functions primarily as an attachment protein that promotes the adsorption of ASFV particles to host cells. It achieves this by binding to specific protein receptors on the host cell membrane, which represents the initial step in viral entry . Experimental evidence shows that recombinant p12 can block specific binding of ASFV to susceptible cells, confirming its role in viral attachment . Once attachment occurs, other viral proteins like p30 appear to be involved in the subsequent internalization process. In the broader infection cycle context, p12 works in coordination with other attachment proteins including p54 and the major capsid protein p72, which also participate in the virus attachment process, though through potentially different mechanisms or receptor interactions .

How can researchers produce and purify recombinant p12 protein for experimental studies?

Recombinant p12 protein can be produced using E. coli expression systems . The methodology typically involves:

  • Cloning the p12 gene sequence from ASFV (isolate Tick/Malawi/Lil 20-1/1983) into an appropriate expression vector

  • Transforming the construct into a suitable E. coli strain optimized for protein expression

  • Inducing protein expression under controlled conditions (temperature, induction agent concentration, duration)

  • Cell lysis to release the expressed protein

  • Purification using affinity chromatography (typically via a His-tag or other fusion tag)

  • Further purification steps such as ion-exchange chromatography or size-exclusion chromatography

  • Quality control assessment via SDS-PAGE, Western blotting, and functional binding assays

The purified recombinant protein should be stored under appropriate conditions to maintain its stability and functionality for subsequent experimental use. Researchers should verify that the recombinant p12 retains its native binding properties before proceeding with attachment or receptor studies.

What methods are used to study p12-receptor interactions on host cells?

Several methodological approaches can be employed to study p12-receptor interactions:

  • Binding assays: Measuring the binding of labeled recombinant p12 to cells, which has revealed both saturable (specific) and non-saturable (non-specific) binding components .

  • Competitive inhibition studies: Using excess unlabeled p12 to compete with labeled p12, demonstrating the specificity of binding.

  • Enzymatic treatment of cell surfaces: Treatment with various enzymes has shown that:

    • Proteases inhibit p12 binding, indicating protein receptors

    • Glycosidases do not affect binding, suggesting carbohydrates are not involved

    • Lipases have no effect, indicating lipids are not part of the receptor interaction

  • Cross-linking studies: To identify the specific cellular proteins that interact with p12.

  • Surface plasmon resonance: For real-time binding kinetics analysis and affinity determination.

  • Virus competition assays: Measuring how recombinant p12 affects ASFV binding and infection, which has confirmed that recombinant p12 can block specific binding of the virus to cells .

These methods collectively provide insights into the nature of the cellular receptors and the specificity of the p12-receptor interaction.

How can researchers visualize the location of p12 in the virion structure?

Visualizing p12's location within the ASFV virion can be achieved through several advanced imaging techniques:

  • Immunoelectron microscopy: This has been used to localize p12 to the inner envelope of the virus in some studies . The method involves:

    • Fixed and sectioned virions or infected cells

    • Incubation with specific anti-p12 antibodies

    • Detection with gold-conjugated secondary antibodies

    • Electron microscopy imaging to visualize the gold particles in relation to viral structures

  • Cryo-electron microscopy: This technique can provide high-resolution structural information of the intact virion without fixation artifacts.

  • Immunofluorescence confocal microscopy: For visualizing p12 in infected cells during different stages of infection.

  • Super-resolution microscopy techniques: Like STORM or PALM for nanoscale localization of p12 relative to other viral proteins.

  • Biochemical fractionation: Isolation of different viral components (outer envelope, inner envelope, capsid, core) followed by Western blot analysis to detect p12.

The contradictory findings regarding p12 localization highlight the importance of using multiple complementary techniques and careful controls when studying viral structural proteins.

How do researchers explain the contradictory findings regarding p12 localization in the virus particle?

The contradictory findings regarding p12 localization (outer envelope versus inner envelope) may be explained by several factors that warrant further investigation:

  • Methodological differences: Different fixation, embedding, or immunolabeling protocols might affect epitope accessibility or protein localization.

  • Virus strain variations: Different ASFV isolates might display structural differences or variations in protein incorporation into the virion.

  • Dynamic relocalization: p12 might relocalize during virion maturation or in response to environmental triggers.

  • Protein processing: Post-translational modifications or proteolytic processing might generate different forms of p12 with distinct localizations.

  • Antibody specificity: Different antibodies might recognize distinct epitopes or conformations of p12 that are differentially accessible in various virion compartments.

A systematic comparative study using standardized methods across multiple ASFV isolates would help resolve these contradictions. Additionally, temporal studies of p12 localization during virion assembly could reveal whether its position changes during the maturation process. Researchers should also consider employing multiple labeling approaches simultaneously to verify localization results .

What is the molecular mechanism of p12-mediated attachment to host cells?

The molecular mechanism of p12-mediated attachment involves specific protein-protein interactions between p12 and cellular receptors. Research has revealed that:

  • The interaction is primarily protein-based, as protease treatment of cells inhibits binding, while glycosidase and lipase treatments do not affect binding .

  • The binding exhibits both saturable (specific) and non-saturable (non-specific) components, similar to what has been observed for whole ASFV virions .

  • The specific cellular receptor proteins for p12 have not been fully characterized, representing a significant knowledge gap.

  • The binding does not appear to require carbohydrate or lipid components on the cellular side of the interaction.

  • The binding affinity and kinetics parameters (kon, koff, KD) for the p12-receptor interaction require further characterization.

Future research directions should include receptor identification through techniques such as affinity purification coupled with mass spectrometry, CRISPR screens to identify essential host factors for binding, and structural studies of the p12-receptor complex to identify critical interaction residues.

How does p12 function in relation to other ASFV attachment proteins?

ASFV employs multiple proteins in the attachment and entry process, with p12 functioning as part of a complex system:

  • Cooperative attachment model: p12, p54, and p72 (major capsid protein) all participate in virus attachment, potentially binding to different cellular receptors or to different regions of the same receptor complex .

  • Sequential action model: Different viral proteins may function at different stages of entry:

    • p72 and p54 appear to be involved primarily in virus attachment

    • p30 seems to function in the subsequent step of virus internalization

  • Differential neutralization effects:

    • Antibodies against p54 inhibit ASFV binding to macrophages

    • Antibodies against p30 block virus internalization even after attachment

    • Antibodies against p12 fail to neutralize viral infectivity despite p12's role in attachment

This complex interplay suggests that ASFV has evolved redundant or complementary attachment mechanisms, perhaps explaining why antibodies against individual attachment proteins often fail to provide complete protection. Understanding the coordinated functions of these proteins requires systems-level experimental approaches and may reveal potential combinatorial targets for intervention strategies.

Why do antibodies against p12 fail to neutralize ASFV infectivity despite its role in attachment?

Although p12 plays a critical role in ASFV attachment, antibodies directed against this protein fail to neutralize viral infectivity or protect pigs from ASF infection . Several hypotheses may explain this counterintuitive finding:

  • Redundant attachment mechanisms: ASFV employs multiple attachment proteins (p12, p54, p72) that may provide functional redundancy, allowing the virus to attach via alternative mechanisms when p12 is blocked.

  • Inaccessibility during infection: The conformation or position of p12 in the intact virion might differ from the recombinant protein used for immunization, rendering antibodies ineffective against the native viral form.

  • Rapid internalization: The virus may be internalized too quickly after initial attachment for antibodies to effectively block the process.

  • Post-attachment neutralization escape: Once attachment has occurred, subsequent steps in viral entry may become antibody-resistant.

  • Epitope masking: Critical functional domains of p12 might be masked by other viral structures or by the interaction with the receptor itself.

This phenomenon highlights the challenges in developing antibody-based interventions against ASFV and suggests that effective vaccines might require targeting multiple viral components simultaneously or focusing on post-attachment stages of infection.

What experimental approaches can determine if recombinant p12 could be valuable in ASFV vaccine development?

Despite the limitations of p12 antibodies in neutralizing infectivity, several experimental approaches can evaluate whether recombinant p12 might still contribute to vaccine development:

  • T-cell response studies: Even if antibodies are not protective, p12 might elicit protective T-cell responses that could be evaluated using:

    • ELISPOT assays to measure T-cell activation

    • Cytotoxicity assays to assess CD8+ T-cell function

    • Adoptive transfer experiments to test protective capacity

  • Multi-antigen approaches: Testing p12 in combination with other ASFV antigens (p54, p30, p72) to identify synergistic protective effects:

    • DNA vaccines expressing multiple antigens

    • Recombinant viral vectors delivering p12 alongside other antigens

    • Virus-like particles incorporating p12 and other structural proteins

  • Challenge studies in pigs: Comprehensive protection assessment after immunization with p12-containing formulations:

    • Monitoring clinical signs, viremia, and survival

    • Comparing single-antigen versus multi-antigen approaches

    • Evaluating different adjuvants and delivery systems

  • Receptor competition strategies: Evaluating whether recombinant p12 could be used therapeutically to block infection:

    • Dose-response studies of infection inhibition

    • Timing experiments to determine the window of effectiveness

    • Delivery method optimization for in vivo applications

These approaches acknowledge that while antibodies against p12 alone may not be protective, the protein might still play a valuable role in comprehensive vaccine strategies through other immunological mechanisms .

How do the binding properties of p12 compare between different ASFV isolates?

Comparing p12 binding properties across different ASFV isolates is crucial for understanding strain-specific variations in attachment and host range. Research approaches should include:

  • Sequence analysis: Comparing p12 sequences from multiple isolates to identify:

    • Conserved regions that might be essential for function

    • Variable regions that might influence binding specificity

    • Phylogenetic relationships between functional variants

  • Comparative binding studies: Assessing binding characteristics of recombinant p12 from different isolates:

    • Affinity measurements using surface plasmon resonance

    • Cell tropism differences across various host cell types

    • Competition assays between p12 variants

  • Cross-protective potential: Evaluating whether p12 from one isolate can block infection by heterologous strains.

  • Structure-function analysis: Mapping functional domains through:

    • Deletion mutants to identify binding regions

    • Site-directed mutagenesis of conserved versus variable residues

    • Structural studies (X-ray crystallography or cryo-EM)

Such comparative studies would be particularly valuable given that ASFV has distinct isolates with potentially different properties, including the hemadsorption (HAD) and non-hemadsorption (non-HAD) phenotypes that correlate with virulence differences . Understanding these strain-specific variations in p12 function could reveal important insights into ASFV host range, tissue tropism, and pathogenicity.

What cellular factors interact with p12 during ASFV attachment and entry?

The specific cellular factors that interact with p12 during ASFV attachment have not been fully characterized, representing a critical knowledge gap. Current understanding indicates:

  • Protein-based receptors: Enzymatic studies using proteases, glycosidases, and lipases suggest that the cellular receptor for p12 is primarily protein-based, with no significant carbohydrate or lipid components involved in the interaction .

  • Cell type specificity: p12 binds to permissive cells in both saturable and non-saturable interactions, suggesting specific receptor engagement alongside non-specific membrane interactions .

  • Receptor identification approaches:

    • Affinity purification using recombinant p12 as bait

    • Crosslinking studies followed by mass spectrometry

    • CRISPR screens to identify essential host factors

    • Comparative proteomics between permissive and non-permissive cells

  • Known exclusions: Research has demonstrated that CD163, previously considered a potential ASFV receptor, is not necessary for ASFV infection, at least for genotype 2 ASFVs .

Identifying the specific cellular receptor(s) for p12 would significantly advance our understanding of ASFV tropism and potentially reveal new targets for antiviral intervention. This represents an important area for continued research using contemporary receptor identification technologies.

What methodological approaches can resolve the contradictions in p12 localization within the virion?

Resolving the contradictions regarding p12 localization (outer envelope versus inner envelope) requires systematic application of complementary methodologies:

  • Comparative immunoelectron microscopy:

    • Using multiple antibodies targeting different p12 epitopes

    • Implementing various sample preparation techniques

    • Quantitative analysis of gold particle distribution

  • Advanced structural approaches:

    • Cryo-electron tomography of intact virions

    • Sub-tomogram averaging for higher resolution

    • Correlative light and electron microscopy

  • Biochemical verification:

    • Viral particle fractionation with detergents of varying strengths

    • Protease protection assays to determine exposed regions

    • Cross-linking experiments to map protein neighborhoods

  • Dynamic studies:

    • Pulse-chase labeling to track p12 during virion assembly

    • Live-cell imaging with fluorescently tagged p12

    • Time-course analysis of virion maturation

  • Genetic approaches:

    • Epitope tagging at different positions within p12

    • Mutational analysis of trafficking signals

    • Domain swapping with proteins of known localization

A multi-technique approach would provide the most definitive resolution to this contradiction, potentially revealing that p12 occupies different locations at different stages of the viral lifecycle or that distinct pools of p12 exist within the virion structure .

Table 6.1: Effects of Enzymatic Treatments on p12 Binding to Host Cells

Enzyme TypeEffect on p12 BindingImplication for Receptor Nature
ProteasesSignificant inhibitionReceptor has critical protein component
GlycosidasesNo significant effectCarbohydrates not essential for binding
LipasesNo significant effectLipids not essential for binding

Data derived from binding studies using recombinant p12 on Vero cells

Table 6.2: Comparative Functions of ASFV Attachment Proteins

Viral ProteinPrimary FunctionAntibody EffectLocation
p12Virus attachmentNon-neutralizingContradictory (outer/inner envelope)
p54Virus attachmentInhibits viral bindingStructural protein
p30 (p32)Virus internalizationBlocks virus after attachmentStructural protein
p72Virus attachmentInduces neutralizing antibodiesMajor capsid protein

Table synthesized from comparative studies of ASFV structural proteins and their roles in viral entry

Table 6.3: ASFV Proteins Involved in Host Immune Modulation

Viral ProteinFunctionMechanismRelevance to Infection
5HLInhibits apoptosisBinds pro-apoptotic Bcl-2 family proteinsPromotes infected cell survival
4CLInhibits apoptosisInhibits caspase activityEnhances cell survival
C-type lectinEnhances cell survivalUnknownPrevents early cell death
5ELInhibits immune responsePrevents NF-κB binding to DNA; Inhibits NFATModulates host response
CD2v (8DR)Mediates hemadsorptionSimilar to T-lymphocyte adhesion receptorAssociated with virulence

Table summarizing ASFV proteins involved in modulating host responses, providing context for p12 function within the broader viral infection strategy

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.