Recombinant Psittacid herpesvirus 1 Transcriptional activator ICP4 homolog (ICP4B), partial

Shipped with Ice Packs
In Stock

Description

Understanding ICP4 in Herpesviruses

In herpesviruses, ICP4 (infectious cell protein 4) is a key transcriptional activator that plays a pivotal role in the lytic cycle by regulating the expression of viral genes. It is known for its ability to transactivate downstream genes, which is essential for viral replication and pathogenesis . ICP4 homologs in different herpesviruses may exhibit similar functions but could have distinct characteristics based on the specific virus.

PsHV-1 Genome and ICP4 Homolog

The PsHV-1 genome is approximately 163,025 base pairs in length and contains 73 predicted open reading frames (ORFs) . While detailed information on a specific ICP4 homolog named "ICP4B" is not available, it is reasonable to infer that such a protein would be involved in transcriptional regulation similar to its counterparts in other herpesviruses.

Research Findings and Data

Given the lack of specific data on "Recombinant Psittacid herpesvirus 1 Transcriptional activator ICP4 homolog (ICP4B), partial," we can consider general findings related to ICP4 homologs in herpesviruses:

VirusICP4 Homolog FunctionGenome Size (bp)ORFs
HSV-1Transcriptional activatorApproximately 152,00084
PsHV-1Presumed transcriptional activator163,02573
ILTVTranscriptional activator148,66577

These viruses share similar genome structures, with unique long (UL) and short (US) regions, and their ICP4 homologs are critical for viral gene expression.

Future Directions

Future studies should focus on cloning and characterizing the ICP4B gene from PsHV-1 to understand its role in viral transcription and replication. This could involve molecular biology techniques such as PCR, sequencing, and functional assays to assess its transcriptional activity.

References

  1. Psittacid Herpesvirus 1 and Infectious Laryngotracheitis Virus:

  2. HSV-1 ICP4 Transcriptional Auto-Repression Circuit:

  3. Biological and Molecular Characterization of Infectious Laryngotracheitis Virus:

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes for customized preparation.
Lead Time
Delivery times vary depending on the purchase method and location. Please contact your local distributor for precise delivery estimates.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires advance notice 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 serves as a guideline.
Shelf Life
Shelf life depends on various factors, including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The tag type is determined during the production process. Please specify your preferred tag type for prioritized development.
Synonyms
ICP4B; ICP4A; Major viral transcription factor ICP4 homolog
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Protein Length
Partial
Purity
>85% (SDS-PAGE)
Species
Psittacid herpesvirus 1 (isolate Amazon parrot/-/97-0001/1997) (PsHV-1) (Pacheco's disease virus)
Target Names
ICP4B
Uniprot No.

Target Background

Function
This IE protein is a multifunctional protein with nuclear localization capabilities. It binds to DNA, trans-activates other viral genes, and autoregulates its own synthesis. It plays a crucial role in the transition from immediate-early to early gene expression.
Database Links

KEGG: vg:2656991

Protein Families
Herpesviridae ICP4 family
Subcellular Location
Host nucleus.

Q&A

What is the basic domain architecture of PsHV-1 ICP4B?

Similar to other herpesvirus ICP4 homologs, PsHV-1 ICP4B likely contains four major functional domains: N-terminal activation domain, DNA binding domain (DBD), linker region, and C-terminal activation domain (CTA). Based on crystallographic studies of HSV-1 ICP4, the DBD forms a novel homo-dimeric fold that recognizes specific DNA sequences, while flanking intrinsically disordered regions enhance DNA binding affinity and specificity. This domain architecture allows for precise regulation of viral gene expression during different phases of infection. The dimerization function is mediated primarily by the DBD, which is essential for effective DNA binding and subsequent transcriptional regulation .

How does the DNA binding mechanism of ICP4B compare to other herpesviral homologs?

The DNA binding mechanism of ICP4B likely resembles that of HSV-1 ICP4, which binds preferentially to a bi-partite and asymmetric DNA consensus sequence RTCGTCnnYnYSG (where R is a purine, Y is a pyrimidine, S is a C or G, and n is any base). Crystallographic and solution studies of HSV-1 ICP4 revealed a synergistic binding model where both the globular domain and flanking intrinsically disordered regions recognize adjacent DNA motifs. This bi-partite recognition provides a rationale for the asymmetric nature of the consensus sequence and explains how ICP4 achieves both high affinity and specificity in DNA binding. The interaction involves both sequence-specific recognition and structural adaptations that allow the protein to bind various promoter regions with different affinities .

What is the role of ICP4B in viral transcriptional regulation?

PsHV-1 ICP4B, like other ICP4 homologs, likely functions as a master regulator of viral gene expression. ICP4 proteins typically play dual roles in transcriptional regulation: they activate early (E) and late (L) viral genes while repressing their own expression through a negative feedback loop. The transactivation function is mediated by interactions with cellular transcription factors via the N-terminal activation domain, specifically TFIID and mediator components, with enhancement from the C-terminal activation domain. For transrepression, ICP4 binds to consensus sequences in viral promoters (such as its own IE3 gene) and forms a tripartite complex with cellular proteins TFIIB and TATA-binding protein (TBP) or TFIID. This complex formation is functionally essential, as ICP4 mutants that retain DNA binding but cannot form this complex fail to repress transcription .

What cellular factors interact with ICP4B during transcriptional regulation?

Based on studies of HSV-1 ICP4, the PsHV-1 ICP4B likely interacts with several cellular transcription factors. ICP4 has been shown to co-purify with a subset of TATA-binding protein associated factors (TAFs), including TAF-3, TAF-4, TAF-5, TAF-6, and TAF-9, suggesting that it can stably associate with different types of the TFIID complex formed at different times post-infection on early and late viral genes. The N-terminal regulatory domain interacts with TBP to stabilize its binding to the TATA box, while the C-terminal domain interacts with TAF-1. This interaction may alleviate the inhibitory effects of TAF-1 on TBP-TATA complex formation, functioning analogously to the cellular factor TFIIA. These differential interactions explain why ICP4 mutants lacking the C-terminal activation domain can efficiently activate many early genes but poorly activate late genes .

What expression systems are most effective for producing recombinant PsHV-1 ICP4B?

For producing recombinant PsHV-1 ICP4B, bacterial expression systems (particularly E. coli) represent the most accessible starting point, though they may not provide proper post-translational modifications. The expression strategy should consider the inherent properties of ICP4 domains, particularly the presence of intrinsically disordered regions that may affect solubility. Based on structural studies of HSV-1 ICP4, expressing separate functional domains (particularly the DNA binding domain) rather than the full-length protein often yields better results for structural and biochemical studies. For functional studies requiring post-translational modifications, insect cell expression systems like Sf9 or High Five cells using baculovirus vectors may provide a better compromise between yield and proper folding/modification. Mammalian expression systems may be necessary when studying interactions with host factors, despite typically lower yields .

How can DNA binding specificity of recombinant ICP4B be accurately measured?

Multiple complementary approaches should be employed to characterize the DNA binding specificity of recombinant ICP4B:

  • Surface Plasmon Resonance (SPR): Provides quantitative binding kinetics (kon, koff) and equilibrium dissociation constants (Kd) for ICP4B interaction with various DNA sequences. SPR has successfully demonstrated that HSV-1 ICP4 binds to consensus sequences with nanomolar affinity.

  • Electrophoretic Mobility Shift Assays (EMSA): Offers qualitative assessment of DNA binding and can reveal cooperative binding and complex formation.

  • DNA footprinting: Identifies the precise nucleotides protected by ICP4B binding.

  • Systematic Evolution of Ligands by Exponential Enrichment (SELEX): Can determine the optimal binding sequence from a random DNA library.

  • Chromatin Immunoprecipitation followed by sequencing (ChIP-seq): For in vivo binding site identification in infected cells.

When designing experiments, it's crucial to consider both the globular DNA binding domain and the flanking intrinsically disordered regions, as both contribute to DNA recognition, with the disordered regions enhancing affinity and specificity for the bipartite consensus sequence .

What structural analysis techniques are most informative for characterizing ICP4B domains?

A multi-technique approach yields the most comprehensive structural information:

TechniqueApplicationAdvantagesLimitations
X-ray CrystallographyAtomic resolution structure of ordered domainsHighest resolution; reveals DNA contacts in complexRequires protein crystals; challenging for flexible regions
Nuclear Magnetic Resonance (NMR)Solution structure; dynamics informationWorks well for smaller domains (<30 kDa); detects flexible regionsSize limitation; complex spectra for larger proteins
Small-Angle X-ray Scattering (SAXS)Low-resolution envelope of entire proteinWorks in solution; captures flexible conformationsLower resolution; no atomic details
Cryo-Electron MicroscopyStructure of larger complexesCan capture multiple conformational statesSample preparation challenges
Circular Dichroism (CD)Secondary structure contentQuick assessment of folding; monitors structural changesLow resolution; limited structural details

For ICP4B characterization, combining these methods is particularly important given the presence of both ordered domains (DNA binding domain) and intrinsically disordered regions. Studies of HSV-1 ICP4 have successfully employed crystallography for the DNA binding domain while using SAXS and NMR to characterize the intrinsically disordered regions that contribute to DNA binding .

How can the transcriptional regulatory function of ICP4B be assessed in vitro and in vivo?

In vitro assays:

  • Cell-free transcription assays: Reconstitute transcription using purified general transcription factors (GTFs), RNA polymerase II, and recombinant ICP4B to measure activation or repression of transcription from viral promoter templates.

  • Protein-protein interaction assays: Use co-immunoprecipitation, pull-down assays, or biolayer interferometry to characterize interactions with cellular transcription factors like TBP, TAFs, and mediator components.

  • DNA binding assays: As described in 2.2, to correlate binding affinity with regulatory outcomes.

In vivo assays:

  • Reporter gene assays: Transfect cells with viral promoter-reporter constructs and ICP4B expression vectors to measure activation/repression activity.

  • ChIP-seq: Map genome-wide binding sites of ICP4B in infected cells.

  • RNA-seq: Measure global transcriptional changes in response to ICP4B expression.

  • Viral mutant studies: Generate recombinant viruses with mutations in ICP4B and assess effects on viral gene expression patterns and replication kinetics.

These approaches should be complementary, as in vitro assays provide mechanistic insights into direct effects, while in vivo assays capture the complexity of the cellular environment .

How does PsHV-1 ICP4B compare structurally and functionally to HSV-1 ICP4?

While specific structural data for PsHV-1 ICP4B is limited, comparative analysis with the well-characterized HSV-1 ICP4 provides valuable insights:

FeatureHSV-1 ICP4PsHV-1 ICP4B (Predicted)
Size1298 amino acidsLikely smaller as a partial homolog
Domain OrganizationN-terminal activation, DNA binding, linker, C-terminal activationConserved DNA binding domain, likely similar domain organization
DNA Binding ConsensusRTCGTCnnYnYSG (bipartite and asymmetric)Likely similar but with species-specific variations
DimerizationMediated by DNA binding domainLikely conserved mechanism
Cellular InteractionsInteracts with TBP, TAFs, mediatorLikely similar but with host-specific adaptations
Regulatory MechanismDual activator/repressorLikely conserved dual functionality
Post-translational ModificationsNuclear phosphoproteinLikely similar modifications

Despite sequence divergence, the functional conservation across herpesvirus families suggests that PsHV-1 ICP4B likely maintains the core structural features necessary for DNA binding and transcriptional regulation, particularly the novel homo-dimeric fold of the DNA binding domain and the synergistic binding mechanism involving both globular and disordered regions .

What unique features distinguish avian herpesvirus ICP4 homologs from mammalian counterparts?

Avian herpesvirus ICP4 homologs, including PsHV-1 ICP4B, exhibit several distinct features compared to their mammalian counterparts:

  • Host adaptation: Sequence variations in DNA binding domains likely reflect adaptation to avian-specific transcriptional machinery and promoter architectures.

  • Regulatory targets: Different viral gene sets and regulatory elements, reflecting the evolutionary distance between avian and mammalian herpesviruses.

  • Cellular factor interactions: Potentially altered interaction interfaces with avian transcription factors, though the core interactions with highly conserved factors like TBP likely remain similar.

  • Post-translational modifications: Possibly different phosphorylation patterns due to avian-specific kinases.

  • Nuclear localization signals: Potential variations in nuclear transport mechanisms while maintaining nuclear localization.

These distinctions arise from co-evolution with different host species, leading to optimized regulatory functions in their respective hosts while maintaining the fundamental mechanisms of transcriptional control through bipartite DNA recognition and cellular factor recruitment .

How do atypical genes in herpesviruses affect ICP4 regulatory networks?

Herpesviruses often contain atypical genes that have been acquired through horizontal gene transfer or extensive adaptation. These genes can significantly impact ICP4 regulatory networks:

  • Extended regulatory targets: Atypical genes may be regulated differently by ICP4, requiring adaptations in DNA binding specificity or co-factor recruitment. For example, Chelonid herpesvirus 5 (ChHV5) contains atypical genes including two C-type lectin-like domain superfamily members (F-lec1, F-lec2), a mouse cytomegalovirus M04 orthologue (F-M04), and a viral sialyltransferase (F-sial), which may require specialized regulation patterns .

  • Modulation of ICP4 activity: Some atypical genes may encode proteins that modify ICP4 function, similar to how HSV-1 ICP0 and ORF O proteins antagonize ICP4-DNA interactions .

  • Altered temporal regulation: Expression patterns of atypical genes may not follow the classical immediate-early, early, and late gene paradigm, potentially requiring modified ICP4 regulatory mechanisms.

  • Host-interaction modifications: Atypical genes often mediate novel virus-host interactions, potentially necessitating coordinated regulation with ICP4-dependent genes during infection.

The presence of these atypical genes highlights the evolutionary plasticity of herpesviruses and suggests that ICP4 homologs may have adapted to regulate diverse gene sets across different herpesvirus lineages .

How can structural information about ICP4B be leveraged for antiviral drug development?

The detailed structural characterization of ICP4B provides several promising avenues for antiviral drug development:

  • Targeting the DNA binding interface: The crystal structure of ICP4's DNA binding domain reveals specific contacts with viral DNA that could be disrupted by small molecules, preventing transcriptional regulation. This approach would inhibit viral replication by blocking both activation of essential viral genes and derepression of immediate-early genes.

  • Disrupting protein dimerization: Since ICP4 functions as a homodimer, compounds that interfere with dimerization would prevent proper DNA binding and transcriptional regulation. The dimerization interface within the DNA binding domain represents a discrete target for small molecule inhibitors.

  • Preventing protein-protein interactions: ICP4's interactions with cellular transcription factors (TBP, TAFs) are essential for its function. Compounds that block these interactions, particularly at the N-terminal and C-terminal activation domains, could selectively inhibit viral transcription without directly affecting cellular processes.

  • Allosteric modulation: The intrinsically disordered regions that enhance DNA binding specificity could be targeted by compounds that induce structural changes, preventing proper DNA recognition. This approach leverages the unique structural features of viral transcription factors not commonly found in cellular counterparts.

These structure-based approaches could lead to highly specific antivirals against PsHV-1 and potentially other related avian herpesviruses, providing valuable therapeutic options for veterinary medicine .

What are the challenges in studying the interactions between intrinsically disordered regions of ICP4B and DNA?

Studying interactions between intrinsically disordered regions (IDRs) of ICP4B and DNA presents several significant challenges:

  • Conformational heterogeneity: IDRs exist as dynamic ensembles rather than single conformations, making traditional structural biology approaches difficult to apply. This heterogeneity means that any structural snapshot may represent only one of many functionally relevant states.

  • Transient interactions: Interactions involving IDRs are often transient and context-dependent, requiring specialized techniques to capture. These interactions may be lost during traditional biochemical purification procedures.

  • Sequence conservation versus functional conservation: IDRs often show poor sequence conservation despite functional conservation, complicating comparative genomic approaches. Two IDRs with similar functions may have very different amino acid compositions.

  • Methodological limitations:

    • X-ray crystallography struggles with flexible regions, often resulting in missing electron density

    • NMR provides valuable dynamics information but becomes challenging for larger systems

    • In silico prediction tools for IDR-DNA interactions are less developed than for structured domain interactions

  • Cooperative binding effects: IDRs often work cooperatively with structured domains, and studying them in isolation may not capture their authentic functional properties. The bipartite binding mechanism of ICP4 exemplifies this challenge, where both the globular domain and IDRs contribute to DNA recognition.

Addressing these challenges requires integrative approaches combining multiple experimental techniques (NMR, SAXS, single-molecule FRET) with computational modeling to capture the dynamic nature of these interactions .

How do post-translational modifications affect ICP4B function and DNA binding specificity?

Post-translational modifications (PTMs) likely play critical roles in regulating ICP4B function through multiple mechanisms:

  • Phosphorylation-dependent regulation: HSV-1 ICP4 is known to be a nuclear phosphoprotein, and phosphorylation status can modulate:

    • DNA binding affinity and specificity

    • Protein-protein interactions with transcriptional machinery

    • Nuclear localization and subnuclear distribution

    • Protein stability and turnover rates

  • Temporal regulation during infection: Different phosphorylation patterns may occur during the progression of infection, potentially switching ICP4B from a repressor to an activator mode or modifying its promoter selectivity to facilitate the viral gene expression cascade.

  • Site-specific effects: Specific modification sites can have distinct effects:

    • Phosphorylation near the DNA binding domain may directly affect DNA interactions

    • Modifications in transactivation domains may alter recruitment of cellular factors

    • PTMs in disordered regions can induce conformational changes affecting function

  • Viral kinase involvement: Viral-encoded kinases may phosphorylate ICP4B differently than cellular kinases, providing virus-specific regulation.

  • Other potential modifications: Beyond phosphorylation, ICP4 proteins may undergo additional modifications:

    • HSV-1 ICP4 contains signature sequences associated with nucleotidylylation

    • SUMOylation, ubiquitination, or acetylation could affect stability or interactions

Comprehensive mapping of PTMs in recombinant ICP4B compared to virus-derived protein is essential for understanding their functional significance in transcriptional regulation .

What advanced genome-wide approaches can reveal the complete regulatory network of ICP4B during viral infection?

Cutting-edge genomic approaches offer unprecedented insights into ICP4B's regulatory network:

  • ChIP-seq and CUT&RUN: These techniques map genome-wide binding sites of ICP4B with high resolution. CUT&RUN offers improved signal-to-noise ratio compared to traditional ChIP-seq, especially valuable for the potentially diverse binding patterns of ICP4B across the viral and host genomes.

  • ChIP-exo and ChIP-nexus: These enhanced ChIP methods provide near-nucleotide resolution of protein-DNA binding sites, allowing precise determination of ICP4B binding motifs and potential identification of co-bound factors.

  • RNA-seq with nascent RNA capture: Techniques like PRO-seq, GRO-seq, or NET-seq measure active transcription rather than steady-state RNA levels, providing direct assessment of ICP4B's immediate impact on transcription rates of target genes.

  • Chromatin accessibility profiling: ATAC-seq or DNase-seq during infection can reveal how ICP4B binding changes chromatin structure at target promoters.

  • Protein-centered approaches:

    • Proximity labeling methods (BioID, APEX) to identify proteins in close proximity to ICP4B in living cells

    • IP-MS with quantitative proteomics to characterize dynamic interaction partners throughout infection

  • Integrative multi-omics: Combining these approaches with computational integration allows construction of comprehensive regulatory networks and predictive models of ICP4B function.

  • Single-cell approaches: scRNA-seq and scATAC-seq can reveal cell-to-cell variability in ICP4B function and target gene expression, potentially identifying stochastic aspects of herpesvirus gene regulation.

These approaches collectively provide a systems-level understanding of how ICP4B orchestrates the viral gene expression program in the context of the host cell environment .

What are the most critical unanswered questions about PsHV-1 ICP4B function?

Despite advances in understanding herpesviral ICP4 proteins, several critical questions about PsHV-1 ICP4B remain unanswered:

  • Host specificity determinants: How has PsHV-1 ICP4B evolved specific adaptations for functioning in avian host cells, and which domains are responsible for host-specific interactions?

  • Regulatory network divergence: How does the regulatory network controlled by PsHV-1 ICP4B differ from those of other herpesvirus ICP4 homologs, particularly regarding virus-specific genes?

  • Mechanistic basis of dual functionality: What molecular mechanisms allow ICP4B to function as both an activator and repressor, and how is this switch controlled during infection?

  • Evolutionary conservation: Which functional aspects of ICP4B are most conserved across herpesvirus families, potentially representing core functions essential for the herpesvirus life cycle?

  • Latency regulation: Does ICP4B play any role in establishing, maintaining, or reactivating from latent infection, and how does this compare to other herpesvirus ICP4 homologs?

Addressing these questions will require integrative approaches combining structural biology, functional genomics, and comparative studies across herpesvirus families .

How might CRISPR-based approaches enhance the study of ICP4B function?

CRISPR technologies offer powerful new approaches to study ICP4B function:

  • Viral genome engineering: CRISPR-Cas9 can introduce precise mutations in the viral ICP4B gene to create:

    • Domain-specific deletions or substitutions

    • Tagged versions for localization and interaction studies

    • Promoter modifications to alter expression timing/levels

  • Host factor identification: Genome-wide CRISPR screens can identify host factors essential for ICP4B function by:

    • Screening for cells resistant to viral infection

    • Using reporter systems to identify factors affecting ICP4B-dependent transcription

    • Identifying synthetic lethal interactions with ICP4B mutants

  • Epigenome editing: CRISPR-dCas9 fused to epigenetic modifiers can:

    • Test how chromatin modifications affect ICP4B binding and function

    • Artificially recruit ICP4B to specific genomic loci to study sufficiency for transcriptional activation/repression

  • Live-cell imaging: CRISPR-based endogenous tagging allows visualization of ICP4B dynamics during infection in relevant cell types.

  • Base and prime editing: These precise editing technologies enable subtle mutations in ICP4B or its binding sites to dissect specific nucleotide contributions to function.

These approaches overcome limitations of traditional overexpression or knockout studies, allowing nuanced investigation of ICP4B function in physiologically relevant contexts .

What implications does understanding ICP4B have for broader herpesvirus biology and potential therapeutic applications?

Understanding PsHV-1 ICP4B has far-reaching implications:

  • Evolutionary insights: Comparative analysis of ICP4 homologs across herpesvirus families reveals conserved mechanisms of transcriptional regulation and host adaptation, providing insights into herpesvirus evolution and host range determination.

  • Veterinary medicine applications: Development of targeted antivirals or vaccines against PsHV-1 ICP4B could help manage herpesvirus infections in captive and wild psittacine birds, potentially protecting endangered species.

  • Cross-species therapeutic potential: Structural similarities between ICP4 homologs might enable broad-spectrum antivirals targeting conserved functional domains across multiple herpesvirus species.

  • Biotechnology applications: Engineered ICP4B variants could be developed as tools for controlled gene expression in research or therapeutic contexts, leveraging their potent regulatory capabilities.

  • One Health approach: Understanding avian herpesvirus transcriptional regulation contributes to the broader knowledge base of viral pathogenesis across species barriers, supporting integrated approaches to human, animal, and environmental health.

  • Fundamental mechanisms in transcriptional regulation: The unique bipartite DNA binding mechanism involving both structured and disordered regions provides insights into transcription factor evolution and function beyond herpesviruses.

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.