Recombinant Equine herpesvirus 1 Major DNA-binding protein (31), partial

Shipped with Ice Packs
In Stock

Description

Overview of Recombinant Equine Herpesvirus 1 Major DNA-Binding Protein (31), Partial

The Recombinant Equine Herpesvirus 1 Major DNA-binding protein (31), partial, also known as UL31 protein (UL31P) of equine herpesvirus 1 (EHV-1), is a protein encoded by the early UL31 gene of EHV-1 . EHV-1 is a significant pathogen in horses, closely related to human pathogens like herpes simplex virus types 1 and 2 (HSV-1 and HSV-2) . The UL31 protein exhibits homology to the ICP8 protein of HSV-1, which is a single-stranded DNA-binding protein crucial for DNA replication .

Role in Viral DNA Replication

The UL31P's characteristics suggest it plays a vital role in EHV-1 DNA replication . As a single-stranded DNA-binding protein, UL31P may be involved in the DNA replicative complex, similar to how ICP8 functions in HSV-1 .

Genetic Variations and Pathogenicity

Genetic variations within EHV-1, such as deletions in the U segment or mutations in the DNA polymerase gene (ORF30), can impact the virus's pathogenicity . For instance, deletions in genes US6, US7, and US8 in the Kentucky A (KyA) strain lead to attenuation . Similarly, mutations in ORF30 have been associated with neuropathogenicity, although this association is not definitive .

Table 1: Deletions in EHV-1 Strains

Gene(s) DeletedStrainEffect on Pathogenicity
US6, US7, US8KyAAttenuation

EHV-1 Replication Cycle

The EHV-1 lytic replication cycle includes several key steps: entry into host cells, viral nucleocapsid uncoating, viral gene expression, DNA replication, virion assembly, and egress . The virus can infect various cell types, including endothelial cells, epithelial cells, and mononuclear cells . Attachment to host cells is mediated by glycoproteins such as gC and gB, followed by gD binding to specific receptors .

Product Specs

Form
Lyophilized powder

Note: We will prioritize shipping the format currently in stock. If you require a specific format, please specify this in your order notes. We will fulfill requests based on availability.

Lead Time
Delivery times vary depending on the purchasing method and location. Please contact your local distributor for precise delivery estimates.

Note: All 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
Centrifuge the vial briefly before opening to collect the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our standard glycerol concentration is 50% and can be used as a guideline.
Shelf Life
Shelf life depends on several factors: storage conditions, buffer components, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C. Lyophilized formulations have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquot for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
The tag type is determined during the manufacturing process.

Tag type is determined during production. If you require a specific tag, please inform us, and we will prioritize its development.

Synonyms
DBP; 31Major DNA-binding protein
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Protein Length
Partial
Purity
>85% (SDS-PAGE)
Species
Equine herpesvirus 1 (strain V592) (EHV-1) (Equine abortion virus)
Target Names
DBP
Uniprot No.

Target Background

Function
This protein plays several critical roles in viral infection. It participates in the unwinding of the viral DNA origin to initiate replication through interaction with the origin-binding protein. It may also disrupt secondary structures (loops, hairpins, etc.) in single-stranded DNA (ssDNA), thus mitigating polymerase pausing during elongation. Furthermore, it promotes viral DNA recombination via strand transfer, enabling the transfer of a DNA strand from a linear duplex to a complementary single-stranded DNA circle. It can also catalyze the renaturation of complementary single strands. Finally, it reorganizes the host cell nucleus, facilitating the formation of prereplicative sites and replication compartments. This process is mediated by the protein's ability to form double-helical filaments in the absence of DNA.
Protein Families
Herpesviridae major DNA-binding protein family
Subcellular Location
Host nucleus.

Q&A

What is the UL31 protein of EHV-1 and how is it characterized?

The UL31 protein (UL31P) of equine herpesvirus 1 (EHV-1) is a single-stranded DNA-binding protein with significant homology to the ICP8 protein of herpes simplex virus type 1 (HSV-1). The protein consists of 1209 amino acids and has a molecular mass of approximately 120 kDa as detected by SDS-PAGE analysis. Structurally, the UL31P contains functional domains that enable it to preferentially bind to single-stranded DNA over double-stranded DNA, which is critical for its role in viral DNA replication .

The amino acid sequences of UL31P from different EHV-1 strains (Ab4p and RacL11) show very high sequence conservation with 99.8% identity, differing in only three amino acid positions. This high conservation suggests the protein's essential function in the viral life cycle. The protein's single-stranded DNA-binding function has been established through gel shift assays using purified recombinant protein, demonstrating its sequence-independent binding capability similar to that observed with HSV-1 ICP8 .

How is the UL31 gene regulated during EHV-1 infection?

The UL31 gene of EHV-1 is regulated as an early gene during viral infection. Its expression is synergistically trans-activated by two viral regulatory proteins: the immediate-early protein (IEP) and the UL5P (EICP27). Promoter assays have demonstrated that optimal expression of UL31 requires the coordinated action of both these regulatory proteins .

Temporal expression analysis shows that UL31 RNA transcripts and protein are first detectable at approximately 6 hours post-infection (hpi) in EHV-1-infected cells, with expression levels increasing significantly at later times of infection (8 and 12 hpi). The early temporal class designation has been confirmed through metabolic inhibition assays using cycloheximide (CHX) to block protein synthesis and phosphonoacetic acid (PAA) to inhibit viral DNA synthesis. These experiments reveal that UL31 expression follows patterns consistent with other early genes such as thymidine kinase (TK) .

A consensus IEP-binding site (IEBS; 5′-ATCGT-3′) has been identified approximately 455 bp upstream of the tentative TATA box within the UL31 promoter region, which likely serves as a cis-acting element required for IEP-mediated trans-activation of the UL31 gene .

What are the DNA-binding properties of UL31P and how can they be experimentally assessed?

The UL31P of EHV-1 exhibits preferential binding to single-stranded DNA over double-stranded DNA, a property that can be experimentally assessed through gel shift assays. These assays have demonstrated that the protein binds to single-stranded DNA in a sequence-independent manner, consistent with its putative role in stabilizing single-stranded regions during viral DNA replication .

To experimentally assess the DNA-binding properties of UL31P, researchers can:

  • Express and purify recombinant UL31P, typically as a GST-fusion protein, from bacterial expression systems

  • Prepare labeled single-stranded and double-stranded DNA probes

  • Perform electrophoretic mobility shift assays (EMSA) with varying concentrations of purified protein and DNA probes

  • Analyze the resulting shifts to determine binding specificity and affinity

Results from such experiments show that UL31P forms stable complexes with single-stranded DNA, resulting in retarded band migration during electrophoresis. Competition assays with unlabeled DNA can further demonstrate the specificity of these interactions. Comparisons with double-stranded DNA probes reveal the protein's preference for single-stranded substrates .

Which regions of UL31P are responsible for its nuclear localization, and how can these be identified?

The nuclear localization of EHV-1 UL31P is primarily determined by a nuclear localization signal (NLS) located within the C-terminal 32 amino acid residues of the protein. This has been demonstrated through subcellular localization studies of green fluorescent protein (GFP)-UL31 fusion proteins .

To identify the NLS domains experimentally, researchers can:

  • Create a series of GFP-UL31 fusion proteins with progressive C-terminal deletions

  • Express these fusion constructs in appropriate cell lines (e.g., RK13 cells)

  • Visualize the subcellular localization using fluorescence microscopy

  • Compare localization patterns to identify which deletion constructs fail to accumulate in the nucleus

Studies have shown that the C-terminal 32 amino acid sequence of UL31P is both necessary and sufficient for nuclear targeting. Multiple sequence alignments of this region with homologous proteins from other alphaherpesviruses reveal significant conservation, particularly with HSV-1 ICP8 (42.6% identity) and PRV UL29 (44% identity), although lower homology (10%) is observed with VZV ORF29, which contains its NLS within the N-terminus .

How do the DNA-binding domains of UL31P compare with those of other herpesvirus single-stranded DNA-binding proteins?

The DNA-binding domains of EHV-1 UL31P show significant structural and functional homology to other herpesviral single-stranded DNA-binding proteins, particularly HSV-1 ICP8 and VZV ORF29P. Sequence alignment and functional studies highlight several conserved regions that mediate the protein's interaction with single-stranded DNA .

Comparative studies reveal:

  • EHV-1 UL31P shares binding mechanism characteristics with HSV-1 ICP8, recognizing single-stranded DNA in a sequence-independent manner

  • The protein contains conserved motifs common to the herpesvirus single-stranded DNA-binding protein family

  • Despite sequence divergence, functional conservation exists among these proteins, suggesting similar roles in viral replication

To experimentally compare these proteins, researchers can perform domain-swapping experiments, creating chimeric proteins where the DNA-binding domains from one viral protein replace those in another. Such approaches can identify functionally equivalent regions and unique features specific to each viral protein. Additionally, structural analysis through X-ray crystallography or cryo-electron microscopy can provide detailed insights into the three-dimensional arrangement of DNA-binding domains and their interaction with nucleic acid substrates .

What experimental approaches can be used to identify functional domains within the UL31 protein?

Multiple complementary experimental approaches can be employed to identify and characterize functional domains within the EHV-1 UL31 protein:

  • Deletion analysis: Creating a series of N-terminal and C-terminal deletion mutants to map regions essential for specific functions (nuclear localization, DNA binding, protein-protein interactions)

  • Site-directed mutagenesis: Introducing point mutations in conserved residues to assess their contribution to protein function

  • Domain swapping: Exchanging putative functional domains with corresponding regions from homologous proteins to evaluate functional conservation

  • Protein fragmentation: Testing isolated protein fragments for retained activities, such as DNA binding or protein interactions

  • Yeast two-hybrid screening: Identifying protein interaction domains through systematic analysis of protein fragments

  • Structural biology approaches: X-ray crystallography or cryo-EM to determine three-dimensional structure

  • Immunoprecipitation coupled with mass spectrometry: Identifying interaction partners that may reveal functional roles

Experimental results using these approaches have already established that the C-terminal 32 amino acids of UL31P contain the nuclear localization signal, while DNA-binding activity resides in domains that recognize single-stranded DNA preferentially over double-stranded DNA. Similar approaches applied to homologous proteins like HSV-1 ICP8 can provide additional insights into the functional organization of UL31P .

What are the optimal methods for expressing and purifying recombinant EHV-1 UL31P for functional studies?

Optimal expression and purification of recombinant EHV-1 UL31P involves several critical considerations:

Expression Systems:

  • Bacterial expression: The UL31 gene can be cloned into prokaryotic expression vectors such as pGEX to produce GST-fusion proteins in E. coli. This approach has been successfully used to generate functionally active protein for DNA-binding studies .

  • Mammalian expression: For studies requiring post-translational modifications, the UL31 gene can be cloned into vectors like pCMV or pEGFP for expression in mammalian cells such as RK13. This approach is particularly valuable for localization studies or when studying protein-protein interactions in a cellular context .

Purification Strategy:

  • Affinity chromatography using GST-tagged proteins bound to glutathione-agarose beads

  • Size exclusion chromatography to separate full-length protein from degradation products

  • Ion exchange chromatography for further purification based on charge properties

Protocol Outline:

  • Clone the UL31 gene into an appropriate expression vector

  • Transform/transfect the construct into the selected expression system

  • Induce protein expression (IPTG for bacterial systems)

  • Lyse cells in buffer containing appropriate protease inhibitors

  • Perform affinity purification using the fusion tag

  • Consider tag removal if necessary for downstream applications

  • Perform quality control via SDS-PAGE and western blot analysis

  • Assess functional activity through DNA-binding assays

Purified UL31P should be tested for its ability to bind single-stranded DNA through gel shift assays to confirm retention of biological activity. For structural studies or crystallization attempts, additional purification steps and buffer optimization may be necessary .

What are the common challenges in producing functional recombinant UL31P and how can they be addressed?

Producing functional recombinant UL31P presents several challenges that researchers should anticipate and address:

Challenge 1: Protein Solubility

  • Problem: Large proteins like UL31P (120 kDa) often form inclusion bodies in bacterial expression systems.

  • Solutions:

    • Use lower induction temperatures (16-18°C)

    • Include solubility-enhancing fusion tags (MBP, SUMO)

    • Optimize expression conditions (IPTG concentration, induction time)

    • Consider refolding protocols if necessary

Challenge 2: Protein Stability

  • Problem: UL31P may be susceptible to proteolytic degradation.

  • Solutions:

    • Include protease inhibitor cocktails during purification

    • Optimize buffer conditions (pH, salt concentration)

    • Minimize freeze-thaw cycles

    • Store with glycerol or other stabilizing agents

Challenge 3: Maintaining DNA-Binding Activity

  • Problem: Recombinant protein may lose DNA-binding activity during purification.

  • Solutions:

    • Avoid harsh purification conditions

    • Validate activity with gel shift assays after each purification step

    • Include reducing agents to maintain cysteine residues

    • Optimize storage conditions

Challenge 4: Post-Translational Modifications

  • Problem: Bacterial systems lack the machinery for mammalian-type post-translational modifications.

  • Solutions:

    • Use mammalian or insect cell expression systems when modifications are critical

    • Consider phosphorylation status (unlike HSV-1 ICP8, EHV-1 UL31P does not appear to be phosphorylated)

Challenge 5: Protein Yield

  • Problem: Large proteins often express at lower yields.

  • Solutions:

    • Optimize codon usage for the expression host

    • Scale up culture volumes

    • Consider expressing functional domains separately

By anticipating these challenges and implementing appropriate strategies, researchers can successfully produce functional recombinant UL31P for structural and functional studies .

What is the role of UL31P in EHV-1 DNA replication and how can this be experimentally determined?

The UL31P of EHV-1 is thought to play a crucial role in viral DNA replication as a single-stranded DNA-binding protein. Based on its homology to HSV-1 ICP8 and its DNA-binding properties, UL31P likely functions in several aspects of the viral replication process :

  • Stabilization of single-stranded DNA: UL31P preferentially binds to single-stranded DNA, potentially stabilizing replication intermediates.

  • Recruitment of replication factors: By analogy to HSV-1 ICP8, UL31P may serve as a scaffold for assembling the viral replication complex.

  • Unwinding of DNA duplex: UL31P may assist in the melting of duplex DNA during replication fork progression.

  • Regulation of gene expression: Some single-stranded DNA-binding proteins also function as transcriptional regulators.

These functions can be experimentally determined through various approaches:

  • Temperature-sensitive mutants or conditional knockdowns: Creating viruses with mutations in UL31 that affect protein function at restrictive temperatures or under specific conditions.

  • Pulse-label experiments: Measuring viral DNA synthesis rates in the presence of wild-type versus mutant UL31P.

  • Chromatin immunoprecipitation (ChIP): Determining the association of UL31P with viral replication origins and other genomic regions during infection.

  • Protein-protein interaction studies: Identifying UL31P interaction partners within the viral replication complex using co-immunoprecipitation or proximity labeling approaches.

  • In vitro replication assays: Reconstituting viral DNA replication with purified components including UL31P to assess its specific contributions.

  • Live-cell imaging: Visualizing the dynamics of fluorescently tagged UL31P during viral replication to determine its spatiotemporal relationships with replication compartments.

Research has already established that UL31P is expressed as an early gene product, consistent with its putative role in DNA replication, and that it preferentially binds to single-stranded DNA, further supporting its involvement in replication processes .

How does UL31P interact with other viral proteins during EHV-1 replication?

While the specific interactions of EHV-1 UL31P with other viral proteins during replication have not been fully characterized, inferences can be made based on homologous proteins in related herpesviruses and the limited available data on EHV-1:

Known and Predicted Interactions:

  • Regulatory Proteins: UL31P expression is synergistically trans-activated by the immediate-early protein (IEP) and UL5P (EICP27), suggesting these proteins function in a coordinated manner during the viral replication cycle .

  • DNA Replication Complex: By analogy to HSV-1 ICP8, UL31P likely interacts with other components of the viral DNA replication machinery, potentially including:

    • Viral DNA polymerase (ORF30P) and its accessory protein (ORF18P)

    • Origin-binding protein (gene 53 product)

    • Helicase-primase complex

  • Nuclear Compartmentalization: UL31P contains a C-terminal nuclear localization signal that targets it to the nucleus where viral DNA replication occurs .

Experimental Approaches to Study Interactions:

  • Co-immunoprecipitation (Co-IP): Using antibodies against UL31P to pull down protein complexes from infected cells, followed by Western blot or mass spectrometry analysis to identify interaction partners.

  • Proximity-based labeling: Expressing UL31P fused to enzymes like BioID or APEX2 that biotinylate nearby proteins, enabling the identification of the UL31P interactome.

  • Yeast two-hybrid screening: Systematically testing interactions between UL31P and other viral proteins.

  • Fluorescence resonance energy transfer (FRET): Detecting protein-protein interactions in living cells using fluorescently tagged proteins.

  • Bimolecular fluorescence complementation (BiFC): Visualizing protein interactions through the reconstitution of fluorescent protein fragments.

Current research indicates that EHV-1 UL31P, like its homologs in other herpesviruses, likely forms part of a complex network of protein-protein interactions that collectively drive viral DNA replication. Further studies are needed to fully elucidate these interaction networks and their functional significance in the viral life cycle .

How does UL31P contribute to the pathogenesis of EHV-1 infection?

The contribution of UL31P to EHV-1 pathogenesis is largely inferred from its role in viral replication and the broader understanding of EHV-1 disease progression:

Replication Efficiency:
As a single-stranded DNA-binding protein likely involved in viral genome replication, UL31P would directly impact viral load and spread within the host. Efficient replication in the respiratory epithelium, the primary site of infection, enables the virus to establish infection and disseminate to secondary sites .

Viremia and Spread:
EHV-1 pathogenesis involves a cell-associated viremia where infected peripheral blood mononuclear cells (PBMCs) transport the virus throughout the body. The ability of the virus to replicate efficiently in these cells, potentially facilitated by UL31P's role in DNA replication, contributes to successful dissemination to the pregnant uterus or central nervous system .

Latency Establishment:
Single-stranded DNA-binding proteins like UL31P may play roles in the balance between lytic replication and latency establishment. By analogy to HSV-1 ICP8, UL31P might influence the efficiency of lytic replication versus latency establishment in neurons of the trigeminal ganglion .

Experimental Approaches:

  • Viral mutants: Generate and characterize UL31 mutants to assess their impact on viral replication in various cell types and in animal models.

  • Comparative pathogenesis: Compare the pathogenicity of wild-type virus versus UL31 mutants in experimental infections.

  • Ex vivo models: Study the replication of UL31 mutants in respiratory mucosal explants or endothelial cell cultures.

  • Transcriptome analysis: Examine how alterations in UL31 affect viral and cellular gene expression during infection.

How does UL31P compare between different strains of EHV-1, and what are the functional implications of any variations?

Comparisons between UL31P sequences from different EHV-1 strains reveal a high degree of conservation, with limited variation that may still have functional implications:

Sequence Conservation:
Studies comparing the UL31 genes of EHV-1 strains Ab4p and RacL11 found 99.8% nucleotide identity, resulting in only three amino acid differences among the 1209 amino acids of the protein . This high conservation suggests strong selective pressure to maintain UL31P function, consistent with its presumed essential role in viral replication.

Strain Variation Analysis:

  • The limited amino acid substitutions identified between strains could potentially affect:

    • Protein-protein interactions

    • DNA-binding affinity or specificity

    • Nuclear localization efficiency

    • Protein stability or folding

  • The positions of these substitutions relative to functional domains could provide insights into their potential impact on protein function.

Functional Implications:
While the functional consequences of these limited variations have not been directly studied, they may contribute to subtle differences in replication efficiency between strains. Given that EHV-1 strains vary in their pathogenic potential, particularly regarding neurological disease (neuropathogenic vs. non-neuropathogenic strains), even subtle differences in replication efficiency could potentially influence disease outcomes .

Experimental Approaches:

  • Site-directed mutagenesis: Introducing strain-specific variations into a common genetic background to assess their functional impact

  • Chimeric protein analysis: Creating hybrid proteins with domains from different strains to map functional differences

  • Comparative replication studies: Assessing replication efficiency in various cell types with different strain variants

  • Structural analysis: Determining how amino acid substitutions might affect protein structure and function

The high conservation of UL31P across EHV-1 strains underscores its essential function in the viral life cycle, while the limited variations observed might contribute to strain-specific differences in replication kinetics or cell tropism .

How has UL31P evolved among different herpesviruses, and what does this reveal about its function?

Evolutionary analysis of UL31P and its homologs across different herpesviruses provides valuable insights into its functional conservation and adaptation:

Cross-Species Comparison:
UL31P homologs are found throughout the alphaherpesvirus subfamily, demonstrating its ancient origin and essential function. Specific amino acid identity comparisons reveal:

VirusAmino Acid Identity with EHV-1 UL31PFunctional Conservation
HSV-1 ICP857.7%Single-stranded DNA binding
PRV UL3167.9%Nuclear localization, DNA binding
VZV ORF2962.5%DNA replication
EHV-4 UL31Higher similarity (exact % not provided)DNA binding, similar temporal expression

This substantial sequence conservation across evolutionarily divergent viruses strongly suggests conservation of critical functions in viral DNA replication .

Domain Conservation and Divergence:

  • DNA-binding domains: Highly conserved, reflecting the fundamental importance of this function

  • Nuclear localization signals: Variable in position but functionally conserved (C-terminal in EHV-1, HSV-1, and PRV; N-terminal in VZV)

  • Protein interaction domains: More variable, possibly reflecting adaptation to species-specific factors

Evolutionary Insights:

  • Functional constraints: The high degree of conservation in DNA-binding regions suggests strong selective pressure to maintain this function

  • Host adaptation: Variations in regulatory regions and protein interaction domains may reflect adaptation to different host environments

  • Recombination events: Analysis of EHV-1 and EHV-4 genomes reveals evidence of widespread natural recombination among field isolates of EHV-4, which could potentially impact UL31P function if such recombination occurs within the UL31 gene

Methodological Approaches:

  • Phylogenetic analysis: Constructing evolutionary trees based on UL31P sequences

  • Selection pressure analysis: Identifying regions under positive or negative selection

  • Functional complementation studies: Testing whether UL31P from one virus can complement defects in another

  • Computational structural analysis: Predicting conservation of structural features across homologs

This evolutionary perspective reveals that while UL31P has maintained its core function as a single-stranded DNA-binding protein essential for viral replication, subtle variations have evolved that may contribute to virus- and host-specific adaptations .

What are the most effective techniques for studying the interactions between UL31P and viral DNA?

Several advanced techniques can effectively characterize the interactions between UL31P and viral DNA:

Electrophoretic Mobility Shift Assays (EMSA):

  • The most direct method for studying UL31P-DNA interactions in vitro

  • Can demonstrate preferential binding to single-stranded versus double-stranded DNA

  • Can determine sequence preferences and binding affinities

  • Allows competition assays to assess binding specificity

  • Has been successfully used to show that UL31P preferentially binds single-stranded DNA

Surface Plasmon Resonance (SPR):

  • Provides real-time, label-free measurements of binding kinetics

  • Determines association and dissociation rate constants

  • Enables precise affinity measurements (KD values)

  • Can assess the effects of buffer conditions on binding

Fluorescence Anisotropy:

  • Monitors changes in the rotational diffusion of fluorescently labeled DNA upon protein binding

  • Allows equilibrium binding measurements in solution

  • Provides direct measurement of binding affinities

  • Can be performed in various buffer conditions to assess ionic strength dependencies

Chromatin Immunoprecipitation (ChIP):

  • Maps binding of UL31P to specific regions of the viral genome during infection

  • Can be coupled with sequencing (ChIP-seq) for genome-wide binding profiles

  • Reveals temporal changes in DNA binding during the viral replication cycle

  • Identifies potential cooperativity with other viral or cellular factors

DNA Footprinting:

  • Identifies specific nucleotides contacted by UL31P

  • Can be performed using chemical (e.g., hydroxyl radical) or enzymatic (e.g., DNase I) approaches

  • Provides high-resolution mapping of binding sites

Single-Molecule Techniques:

  • Fluorescence resonance energy transfer (FRET) to monitor protein-DNA interactions at the single-molecule level

  • Optical or magnetic tweezers to assess the effects of UL31P on DNA mechanical properties

  • Total internal reflection fluorescence (TIRF) microscopy to visualize protein-DNA interactions in real-time

Cryo-Electron Microscopy:

  • Structural characterization of UL31P-DNA complexes

  • Reveals the molecular architecture of larger nucleoprotein complexes

  • Can be combined with image reconstruction for high-resolution analysis

These techniques can be applied individually or in combination to comprehensively characterize how UL31P interacts with viral DNA, potentially revealing mechanisms by which it contributes to DNA replication and other aspects of the viral life cycle .

How can CRISPR-Cas9 genome editing be utilized to study UL31P function in EHV-1?

CRISPR-Cas9 genome editing offers powerful approaches to study UL31P function in the context of EHV-1 infection:

Targeting Strategies:

  • Complete UL31 Gene Knockout:

    • Design gRNAs targeting critical regions of the UL31 gene

    • Introduce frameshift mutations or large deletions to completely disrupt protein expression

    • Assess the impact on viral replication to determine essentiality

    • Note: If UL31 is essential (as predicted), complete knockouts may not yield viable virus, necessitating conditional approaches

  • Domain-Specific Mutations:

    • Target specific functional domains (DNA-binding region, nuclear localization signal)

    • Use homology-directed repair to introduce precise mutations

    • Create point mutations that alter specific functions while preserving protein expression

    • Example: Mutate the C-terminal nuclear localization signal to study its importance in viral replication

  • Tagged Protein Generation:

    • Introduce epitope tags or fluorescent protein fusions at the C- or N-terminus

    • Enable visualization of UL31P localization during infection

    • Facilitate immunoprecipitation studies to identify interaction partners

    • Create split-GFP constructs to study protein-protein interactions in living cells

Experimental Design Considerations:

  • BAC Mutagenesis Pipeline:

    • Many EHV-1 strains are available as bacterial artificial chromosomes (BACs)

    • Perform CRISPR editing in bacterial systems before virus reconstitution

    • Screen for desired mutations using colony PCR and sequencing

    • Reconstitute mutant viruses in permissive cell lines

  • Conditional Systems:

    • If UL31 is essential, implement conditional approaches:

      • Create cell lines expressing wild-type UL31P to complement viral mutants

      • Develop inducible expression systems to control UL31P levels

      • Utilize degron tags for temporal control of protein stability

  • Phenotypic Analyses:

    • Compare growth kinetics of wild-type and mutant viruses

    • Assess viral DNA replication using qPCR or click chemistry approaches

    • Examine changes in the formation of replication compartments

    • Measure effects on gene expression patterns using RNA-seq

    • Analyze changes in virus morphogenesis using electron microscopy

  • Rescue Experiments:

    • Demonstrate specificity by rescuing phenotypes through complementation

    • Express wild-type UL31P in trans to verify that observed defects are directly attributed to UL31P mutations

    • Use homologs from related viruses to assess functional conservation

CRISPR-Cas9 approaches provide unprecedented precision in manipulating the viral genome, enabling detailed functional studies of UL31P that were previously challenging or impossible with traditional mutagenesis methods. When combined with advanced imaging, biochemical, and genomic analyses, these approaches can comprehensively define the role of UL31P in the EHV-1 life cycle .

What high-throughput approaches can be used to identify host factors that interact with UL31P?

Several high-throughput approaches can efficiently identify host cellular factors that interact with EHV-1 UL31P:

1. Proximity-Based Labeling Proteomics:

  • BioID: Fusion of UL31P with a promiscuous biotin ligase (BirA*) that biotinylates proteins in close proximity

  • APEX2: Fusion of UL31P with an engineered peroxidase that catalyzes biotin-phenol labeling of nearby proteins

  • TurboID/miniTurboID: Faster variants of BioID with improved labeling kinetics

  • Advantages: Captures transient interactions; works in native cellular environments; can be temporally controlled during infection

2. Affinity Purification-Mass Spectrometry (AP-MS):

  • Express tagged versions of UL31P (FLAG, HA, or Strep-tag) in infected cells

  • Immunoprecipitate protein complexes using tag-specific antibodies

  • Identify co-purifying proteins using liquid chromatography-tandem mass spectrometry (LC-MS/MS)

  • Quantitative approaches (SILAC, TMT) can distinguish specific from non-specific interactions

  • Advantages: Well-established methodology; can identify stable protein complexes

3. Yeast Two-Hybrid (Y2H) Screening:

  • Use UL31P as bait against human or equine cDNA libraries

  • Can screen full-length UL31P or specific domains separately

  • Follow up positive interactions with validation in mammalian cells

  • Advantages: Can screen large libraries; doesn't require specialized equipment

4. Protein Microarrays:

  • Probe arrays containing thousands of purified human proteins with labeled UL31P

  • Identify direct protein-protein interactions

  • Advantages: Identifies direct binary interactions; high-throughput format

5. CRISPR Screens:

  • Perform genome-wide CRISPR knockout or activation screens in cells expressing UL31P

  • Identify genes that affect UL31P localization, stability, or function

  • Advantages: Identifies functional relationships beyond physical interactions

6. RNA-Seq of UL31P-Expressing Cells:

  • Compare transcriptomes of cells expressing wild-type versus mutant UL31P

  • Identify pathways affected by UL31P expression

  • Advantages: Provides insight into broader functional impact

Data Analysis and Validation:

  • Filtering Strategies:

    • Compare against negative controls (empty vector, unrelated viral protein)

    • Filter against common contaminant databases

    • Prioritize proteins enriched in multiple independent experiments

    • Focus on proteins with relevant cellular functions (DNA replication, nuclear organization)

  • Validation Approaches:

    • Co-immunoprecipitation in infected cells

    • Proximity ligation assay (PLA) to visualize interactions in situ

    • Functional validation through siRNA knockdown or CRISPR knockout

    • Co-localization studies using fluorescence microscopy

  • Network Analysis:

    • Integrate data into protein interaction networks

    • Identify enriched cellular pathways and processes

    • Compare with interactomes of homologous proteins from other herpesviruses

These high-throughput approaches, particularly when used in combination, can provide comprehensive insights into how UL31P interfaces with the host cell machinery during viral infection, potentially revealing new targets for antiviral intervention .

How might understanding UL31P function contribute to developing antivirals or vaccines against EHV-1?

Understanding UL31P function has significant implications for developing novel antivirals and improved vaccines against EHV-1:

Antiviral Development Strategies:

  • Direct Inhibition of UL31P Function:

    • Design small molecules that interfere with UL31P DNA-binding activity

    • Develop peptide inhibitors that disrupt critical protein-protein interactions

    • Target the interface between UL31P and other viral replication factors

    • Advantage: High specificity for viral processes with potentially low host toxicity

  • Structure-Based Drug Design:

    • Utilize structural information about UL31P DNA-binding domains

    • Identify binding pockets amenable to small molecule inhibition

    • Employ in silico screening followed by biochemical validation

    • Focus on conserved regions that might inhibit multiple herpesviruses

  • Nucleic Acid-Based Approaches:

    • Design antisense oligonucleotides or siRNAs targeting UL31 mRNA

    • Develop CRISPR-Cas strategies to cleave viral genomic sequences

    • Create aptamers that specifically bind and inhibit UL31P function

Vaccine Development Applications:

  • Attenuated Vaccine Strains:

    • Engineer EHV-1 strains with modified UL31P that maintain immunogenicity but have reduced replication capacity

    • Create temperature-sensitive mutants of UL31P for attenuated live vaccines

    • Ensure mutations are stable and cannot revert to wild-type during replication

  • Subunit or Vector Vaccines:

    • Include UL31P or its immunogenic epitopes in subunit vaccine formulations

    • Express UL31P along with other viral antigens in viral vector platforms

    • Target both humoral and cell-mediated immune responses

  • Improved Protection Assessment:

    • Develop assays based on UL31P function to evaluate vaccine efficacy

    • Monitor viral replication efficiency as a correlate of protection

    • Assess the impact of vaccination on viral DNA replication

Therapeutic Considerations:

Current EHV-1 vaccines provide incomplete protection, particularly against neurological disease (EHM) . Understanding UL31P's role in viral replication could identify new targets or approaches to enhance vaccine efficacy. For antiviral development, targeting essential viral replication proteins like UL31P might provide broader protection against various clinical manifestations of EHV-1 infection.

The high conservation of UL31P across EHV-1 strains suggests that therapeutics targeting this protein might be effective against diverse viral isolates, potentially including those with the neuropathogenic marker (D752 variant) associated with neurological disease .

These approaches could contribute to addressing the significant ongoing impact of EHV-1 on equine health and the horse industry, where current control measures remain inadequate for preventing serious disease outbreaks .

What are the most promising research directions for further understanding the role of UL31P in EHV-1 biology?

Several promising research directions could significantly advance our understanding of UL31P's role in EHV-1 biology:

1. Structural Biology Approaches:

  • Determine the three-dimensional structure of UL31P alone and in complex with DNA

  • Map the structural basis for preferential binding to single-stranded DNA

  • Identify potential sites for small molecule inhibitor development

  • Use cryo-electron microscopy to visualize UL31P within viral replication complexes

2. Systems Biology of Viral Replication:

  • Define the complete interactome of UL31P during viral infection

  • Map temporal changes in UL31P interactions throughout the replication cycle

  • Develop live-cell imaging approaches to visualize UL31P dynamics during infection

  • Create comprehensive models of the viral replication complex

3. Functional Genomics:

  • Generate and characterize a comprehensive library of UL31P mutants

  • Use deep mutational scanning to identify critical residues for various functions

  • Develop conditional systems to study essential functions

  • Apply CRISPR interference/activation to modulate UL31P expression levels

4. Comparative Virology:

  • Compare UL31P function across different herpesviruses

  • Identify conserved and divergent mechanisms

  • Develop systems for functional complementation studies

  • Explore evolution of DNA-binding specificity

5. Host-Pathogen Interactions:

  • Characterize how UL31P interfaces with host DNA replication and repair machinery

  • Investigate potential roles in immune evasion

  • Determine how UL31P contributes to cell type-specific replication efficiency

  • Explore roles in latency establishment or reactivation

6. Translational Research:

  • Develop UL31P-targeted antivirals

  • Evaluate UL31P modifications for attenuated vaccine development

  • Create diagnostic tools based on UL31P detection

  • Establish correlates between UL31P function and disease outcomes

7. Advanced Technologies:

  • Apply single-molecule approaches to study UL31P-DNA interactions

  • Develop organoid models to study UL31P function in complex tissues

  • Use super-resolution microscopy to visualize UL31P within replication compartments

  • Implement computational approaches to predict DNA-binding properties

These research directions would address critical knowledge gaps regarding UL31P function and potentially lead to practical applications in disease control. Particularly promising are approaches that combine structural insights with functional genomics to develop a comprehensive understanding of UL31P's multiple roles in the viral life cycle .

How can recombinant UL31P be used as a tool to study EHV-1 pathogenesis and host immune responses?

Recombinant UL31P offers versatile applications as a research tool for investigating EHV-1 pathogenesis and host immune responses:

As a Probe for Viral Replication Mechanisms:

  • DNA Replication Studies:

    • Use fluorescently labeled recombinant UL31P to visualize viral DNA replication sites

    • Track the formation and dynamics of replication compartments in infected cells

    • Identify cellular factors recruited to viral replication sites

  • Functional Inhibition:

    • Introduce dominant-negative UL31P mutants to disrupt viral replication

    • Use cell-penetrating UL31P fragments to compete with endogenous protein

    • Assess impact on different stages of the viral life cycle

  • Interaction Mapping:

    • Employ recombinant UL31P to capture and identify interaction partners

    • Characterize the viral replication complex assembly

    • Map binding sites for both viral and cellular proteins

For Immunological Applications:

  • Antigen for Antibody Production:

    • Generate specific antibodies against UL31P for detection in infected tissues

    • Develop tools to differentiate between active infection and vaccination

    • Create reagents for studying UL31P localization and expression kinetics

  • T-Cell Response Analysis:

    • Identify UL31P epitopes recognized by T cells

    • Evaluate cell-mediated immune responses to this conserved viral protein

    • Assess patterns of immunodominance during EHV-1 infection

  • Vaccine Development:

    • Test UL31P as a component in subunit vaccine formulations

    • Evaluate protection conferred by immune responses to UL31P

    • Develop UL31P-based immunogens designed to elicit neutralizing antibodies

For Diagnostic Applications:

  • Serological Assays:

    • Develop ELISA or other immunoassays using recombinant UL31P

    • Distinguish between infected and vaccinated animals (DIVA strategy)

    • Monitor herd immunity in equine populations

  • Molecular Diagnostic Tools:

    • Create standards for quantitative PCR assays targeting UL31

    • Develop protein-based detection methods for active infection

    • Design point-of-care diagnostic systems

Experimental Models:

  • Ex Vivo Systems:

    • Track UL31P expression in respiratory mucosal explants

    • Study viral replication in tissue-specific contexts

    • Evaluate antiviral compounds targeting UL31P function

  • In Vivo Applications:

    • Use recombinant UL31P to track immune responses in experimental infections

    • Develop imaging agents based on UL31P for in vivo visualization

    • Create reporter systems to monitor viral replication

Recombinant UL31P thus serves as a versatile molecular tool for studying multiple aspects of EHV-1 biology, from basic mechanisms of replication to applied aspects of diagnosis and prevention. Its high conservation across viral strains makes it particularly valuable for developing broadly applicable research tools and potential countermeasures .

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.