Phosphorylation regulates P protein localization and function. Key findings include:
Phosphorylation sites:
Functional impact:
The P CTD directly antagonizes host antiviral responses:
STAT1 inhibition: Binds STAT1 via residues Ile201–Phe209, blocking interferon-activated gene transcription .
IRF3 suppression: Prevents phosphorylation of IRF3 at Ser386, inhibiting interferon-β production .
Microtubule bundling: Sequesters viral replication complexes away from immune sensors .
Recombinant P protein studies have enabled:
Drug targeting: Identification of the NPYNE motif (residues 1929–1933) in L protein as a site for inhibiting L-P interaction .
Structural mapping: NMR and mutagenesis reveal P CTD interaction interfaces with N protein (Leu381, Asp383/384, pSer389) and STAT1 .
| Protein | Critical Residues | Interaction Role | Reference |
|---|---|---|---|
| P CTD | Lys211, Lys214, Arg260 | Electrostatic binding to N protein loop | |
| N CTD | pSer389, Asp383, Asp384 | Enhances binding affinity via phosphorylation |
While RABV P protein is well-characterized, Khujand virus P remains unstudied. Priorities include:
Comparative genomics: Identify conserved regions between Khujand virus and RABV P proteins.
Functional assays: Test cross-reactivity of RABV-targeted inhibitors (e.g., NPYNE blockers) on Khujand virus.
KEGG: vg:21011768
Khujand virus (KHUV) is a bat lyssavirus isolated from the whiskered bat (Myotis mystacinus) in Tajikistan in 2001. It belongs to the Lyssavirus genus within the Rhabdoviridae family . Phylogenetically, KHUV is most closely related to European Bat Lyssavirus-2 (EBLV-2), with 79.0% nucleotide identity in the nucleoprotein (N) gene .
The KHUV phosphoprotein (P) consists of 297 amino acids with a molecular weight of 24,373 Da . Like other lyssavirus P proteins, it functions as a multifunctional protein with roles in:
Sequence analysis of KHUV P protein reveals the characteristic amino acid sequence "MSKIFVNPSAIRAGLADLEMAEETVDLINRNVEDNQAHLQGEPIEVEALPEDRRLHISEQKHSQLDSACGKEEGSDDDFY..." . Comparing this sequence with other lyssavirus P proteins indicates conserved functional domains that are essential for viral replication and immune evasion.
Based on comparative analysis with other lyssaviruses, particularly rabies virus (RABV), the KHUV phosphoprotein contains several functional domains:
N-terminal domain: Likely contains regions involved in IFN antagonism. In RABV, amino acids 176-186 are essential for inhibiting IFN induction . Similar regions may exist in KHUV P.
Central domain: Contains regions for interaction with nucleoprotein (N) to form the viral nucleocapsid complex.
C-terminal domain: Likely contains the L protein binding site. In RABV, the sequence NPYNE at positions 1929-1933 in the L protein is critical for binding to P protein .
These domains make the P protein valuable for research applications including:
Studying virus-host interactions
Developing antiviral strategies targeting P-L interactions
Understanding mechanisms of immune evasion by lyssaviruses
Comparative studies of lyssavirus evolution and host adaptation
The recombinant KHUV P protein can be expressed in several systems, each with advantages for different research applications:
Most commonly used for basic structural studies and antibody production
The commercially available recombinant KHUV P protein is produced in E. coli with ≥85% purity
Protocol: The full-length P gene (encoding amino acids 1-297) is cloned into an expression vector (e.g., pET series), transformed into BL21(DE3) cells, induced with IPTG, and purified using affinity chromatography (typically His-tag purification)
Advantages: High yield, cost-effective, suitable for structural studies
Limitations: May lack post-translational modifications present in mammalian systems
Mammalian expression systems (HEK293, BHK-21):
Optimal for functional studies where proper folding and post-translational modifications are critical
Protocol: The P gene is cloned into mammalian expression vectors (e.g., pcDNA3.1), transfected into mammalian cells, and the protein is purified using affinity tags
Advantages: Proper protein folding and modifications, suitable for interaction studies
Similar approaches have been successful for expressing and studying lyssavirus proteins, as demonstrated with ABLV glycoprotein expressed in 293F cells
Provides a balance between yield and proper folding
Advantages: Higher yield than mammalian systems while maintaining proper folding
Useful for large-scale production for structural studies
For functional studies investigating P protein interactions with host factors or other viral proteins, mammalian expression systems are recommended despite lower yields.
Successful purification of functional recombinant KHUV P protein requires careful attention to several factors:
His-tag (6×His): Most commonly used, enables purification using Ni-NTA agarose
GST-tag: Alternative approach useful for solubility enhancement and pull-down assays
The tag position (N or C-terminal) should be selected based on known functional domains to minimize interference
Phosphate buffers (pH 7.4-8.0) with 150-300 mM NaCl are typically suitable
Addition of 5-10% glycerol and 1-5 mM DTT or 2-ME helps maintain stability
For proteins prone to aggregation, low concentrations (0.05-0.1%) of non-ionic detergents (Triton X-100, NP-40) may be included
Cell lysis: Sonication or French press in buffer containing protease inhibitors
Clarification: Centrifugation at 15,000-20,000×g for 30 minutes
Affinity chromatography: Binding to Ni-NTA resin for His-tagged proteins
Washing: Multiple washes with increasing imidazole concentrations (10-40 mM)
Elution: With 250-300 mM imidazole
Dialysis: Against storage buffer to remove imidazole
Quality control: SDS-PAGE and Western blotting to confirm purity
Circular dichroism to assess secondary structure
Thermal shift assays to evaluate stability
Binding assays with known interaction partners (e.g., L protein fragments)
For studying interactions with host proteins, additional purification steps such as size exclusion chromatography may be necessary to ensure high purity (>95%).
Studying KHUV P protein interactions with host immune factors requires specialized experimental approaches:
Express tagged recombinant KHUV P protein in mammalian cells (e.g., HEK293T)
Lyse cells in buffer containing 50 mM Tris-HCl (pH 7.4), 150 mM NaCl, 1% NP-40, and protease inhibitors
Incubate lysates with antibodies against the tag or suspected interaction partners
Precipitate using protein A/G beads and analyze by Western blotting
This approach has successfully identified interactions between RABV P and pattern recognition receptors like RIG-I, MDA5, and Lgp2
Useful for discovering novel interaction partners
Clone KHUV P into bait vector and screen against human cDNA libraries
Validate hits using co-IP or GST pull-down assays
Transfect cells with IFN-β promoter-luciferase reporter construct
Co-transfect with KHUV P expression plasmid
Stimulate cells with poly(I:C) or other IFN inducers
Measure luciferase activity to quantify IFN inhibition
Similar approaches have demonstrated that RABV P protein inhibits poly(I:C)-induced IFN-β promoter activity
Express fluorescently tagged KHUV P in mammalian cells
Co-stain for cellular compartments or co-express fluorescently tagged host factors
Analyze colocalization using confocal microscopy
This approach can reveal interactions with IRF3, STAT1, or other immune signaling proteins
These methods can be used to compare KHUV P protein with P proteins from other lyssaviruses to understand species-specific differences in immune evasion strategies.
Developing high-quality monoclonal antibodies against KHUV P protein requires systematic approaches:
Prepare 100 μg of purified recombinant KHUV P protein emulsified with Freund's complete adjuvant for initial immunization
Boost with the same amount of protein mixed with Freund's incomplete adjuvant every 2 weeks (twice)
Final boost with purified protein before hybridoma fusion
This approach has been successful for generating mAbs against lyssavirus L protein
Isolate splenocytes from immunized mice (BALB/c preferred)
Fuse with SP2/0 myeloma cells using polyethylene glycol 2000
Culture in RPMI 1640 medium containing HAT selection medium
Screen hybridomas by indirect ELISA using purified KHUV P protein (2 μg/mL in carbonate buffer, pH 9.6)
Subclone positive hybridomas three times by limiting dilution
Determine isotypes using isotyping kits
Test reactivity by Western blot and immunofluorescence assay
Assess cross-reactivity with P proteins from other lyssaviruses
Map epitopes using truncated protein fragments
Generate overlapping fragments of KHUV P protein
Express fragments as fusion proteins with His or GST tags
Test reactivity of mAbs with each fragment by Western blot
Progressively narrow down the epitope by expressing smaller fragments
Confirm the minimal epitope using alanine scanning mutagenesis
This approach can identify conserved epitopes across lyssaviruses
For optimal results, consider developing antibodies against both linear and conformational epitopes by using both denatured and native protein for screening.
Investigating KHUV P protein's role in viral RNA synthesis requires specialized techniques:
Construct a KHUV-specific minigenome containing a reporter gene (luciferase or GFP) flanked by KHUV leader and trailer sequences
Co-transfect cells with the minigenome and plasmids expressing KHUV N, P, and L proteins
Measure reporter gene expression as an indicator of viral RNA synthesis
Create P protein mutants to identify functional domains
This approach has been used successfully with rabies virus to evaluate P protein function as a cofactor of viral RNA polymerase
Express and purify recombinant KHUV P and L proteins
Perform in vitro binding assays to map interaction domains
Generate truncated or mutated versions of P to identify essential regions
In RABV, the sequence NPYNE at positions 1929-1933 in the L protein is critical for binding P protein , suggesting similar critical regions may exist in KHUV
Generate P-deficient KHUV or use P-deficient RABV as a surrogate system
Complement with wild-type or mutant KHUV P protein
Measure viral replication efficiency
This approach can identify functionally important regions of P protein
Use X-ray crystallography or cryo-EM to determine the structure of KHUV P protein alone or in complex with N or L protein fragments
Perform molecular dynamics simulations to understand structural flexibility
Use hydrogen-deuterium exchange mass spectrometry to identify regions involved in protein-protein interactions
These approaches together provide comprehensive insights into the roles of KHUV P protein in viral RNA synthesis and replication.
Comparing interferon antagonism activities of KHUV P with other lyssavirus P proteins requires systematic approaches:
Transfect cells with reporters containing IFN-β promoter, ISRE (Interferon-Stimulated Response Element), or GAS (Gamma-Activated Sequence) controlling luciferase expression
Co-transfect with expression plasmids for P proteins from different lyssaviruses (KHUV, RABV, EBLV-1, EBLV-2, etc.)
Stimulate with appropriate inducers (poly(I:C) for IFN-β, IFN-α/β for ISRE, IFN-γ for GAS)
Measure luciferase activity to quantify and compare inhibition levels
Similar approaches have shown that RABV P protein isoforms inhibit poly(I:C)-induced IFN-β promoter activity
Express KHUV P and other lyssavirus P proteins in cells
Stimulate with poly(I:C) or RIG-I ligands
Analyze IRF3 phosphorylation by Western blot using phospho-specific antibodies
Examine IRF3 nuclear translocation by immunofluorescence or nuclear/cytoplasmic fractionation
RABV P has been shown to bind to an activation-intermediate form of IRF3, preventing its full activation
Express P proteins in appropriate cell lines (e.g., human or bat cells)
Stimulate with IFN inducers
Measure mRNA levels of IFN-β, Mx1, OAS1, and other ISGs by qRT-PCR
This approach has revealed that RABV P protein mutants lacking amino acids 176-186 cannot inhibit IFN induction
Express tagged P proteins from different lyssaviruses
Immunoprecipitate and identify bound host factors by Western blot or mass spectrometry
Compare binding profiles to identify conserved and species-specific interactions
RABV P has been shown to interact with RIG-I-like receptors (RLRs) and IRF3
A comprehensive table comparing the interferon antagonism mechanisms of different lyssavirus P proteins would help identify conserved strategies and virus-specific adaptations.
Generating and utilizing truncated forms of KHUV P protein involves several methodological considerations:
Analyze sequence alignments of lyssavirus P proteins to identify conserved domains
Design truncation mutants targeting specific functional regions:
N-terminal truncations (similar to natural P2, P3, P4, P5 isoforms in RABV)
C-terminal truncations
Internal deletions of specific domains
Clone into expression vectors with appropriate tags (His, FLAG, HA) for detection and purification
Express in E. coli or mammalian cells depending on the experimental goals
Similar approaches with RABV have identified that truncated P proteins (P2, P3) function as IFN antagonists
| Truncation | Expected Function | Experimental Approach |
|---|---|---|
| N-terminal | IFN antagonism | IFN reporter assays, IRF3 phosphorylation analysis |
| Central region | N protein binding | Co-IP with N protein, minigenome assays |
| C-terminal | L protein binding | Co-IP with L protein, in vitro RNA synthesis |
Identify potential alternative start codons in KHUV P gene (similar to positions 20, 53, 69, and 83 in RABV P gene )
Generate constructs expressing each potential isoform (P1, P2, P3, etc.)
Compare their functions in:
IFN antagonism (reporter assays)
Subcellular localization (immunofluorescence)
Host protein interactions (co-IP)
Research with RABV has shown that these truncated isoforms contribute to pathogenesis through IFN antagonism
Generate chimeric proteins swapping domains between KHUV P and other lyssavirus P proteins
Compare functions to identify species-specific adaptations
Use in minigenome systems to assess polymerase cofactor functions
Test in IFN antagonism assays to map domains responsible for host immune evasion
These approaches provide comprehensive insights into domain-specific functions of KHUV P protein and their conservation across the lyssavirus genus.
Researchers working with recombinant KHUV P protein often encounter several technical challenges:
Challenge: P proteins can form aggregates during expression and purification
Solutions:
Reduce induction temperature to 16-18°C
Use solubility-enhancing tags (MBP, SUMO, or GST)
Add 5-10% glycerol to all buffers
Include low concentrations (0.05-0.1%) of non-ionic detergents
Consider refolding approaches if inclusion bodies form
Challenge: P protein may degrade during purification or storage
Solutions:
Include protease inhibitors in all buffers
Add reducing agents (1-5 mM DTT or 2-ME)
Store at -80°C in small aliquots with 10-15% glycerol
Avoid repeated freeze-thaw cycles
Challenge: P proteins can exhibit non-specific interactions with cellular proteins
Solutions:
Increase salt concentration (300-500 mM NaCl) in binding buffers
Include 0.1-0.5% BSA as a blocking agent
Use more stringent washing conditions
Include appropriate negative controls (unrelated proteins with similar tags)
Challenge: Variable results in functional assays
Solutions:
Verify protein folding by circular dichroism
Confirm activity in well-established assays before proceeding to more complex experiments
Use freshly purified protein for critical experiments
Include positive controls (e.g., RABV P protein) in parallel experiments
Challenge: Antibodies may cross-react with P proteins from other lyssaviruses
Solutions:
Carefully validate antibody specificity using multiple lyssavirus P proteins
Consider epitope mapping to identify unique regions for antibody development
Use tagged proteins and tag-specific antibodies for detection when possible
Implementing these strategies can significantly improve the success rate of experiments involving recombinant KHUV P protein.
Analysis of codon usage in the KHUV P gene reveals important patterns with implications for recombinant expression:
The KHUV P gene, like other lyssavirus P genes, shows distinct codon usage bias
The GC content of KHUV P gene is approximately 50.50%, with GC1>GC3>GC2 pattern typical for lyssaviruses
At the third codon position, T3% is highest, followed by A3%, C3%, and G3%
Effective Number of Codons (ENC) value for KHUV P gene is around 50.50-55.80, indicating moderate codon bias
| Virus | P Gene GC Content (%) | ENC Value | Mutational Responsive Index |
|---|---|---|---|
| Khujand virus | 50.50 | 0.10 | 0.61 |
| Irkut virus | 54.70 | 0.09 | 0.44 |
| European bat Lyssavirus 1 | ~54-56 | 0.09-0.10 | 0.45-0.50 |
| Aravan virus | ~51-53 | 0.09-0.11 | 0.50-0.55 |
| Rabies virus | ~54-56 | 0.08-0.10 | 0.45-0.50 |
Data adapted from codon usage analysis of Rhabdoviridae genomes
Expression system selection: Codon usage differences between KHUV and expression hosts (E. coli, yeast, insect cells) may affect expression efficiency
Codon optimization strategies:
For E. coli expression: Optimize rare codons (particularly those encoding Arg, Leu, Ile, and Pro)
For mammalian expression: Less optimization may be needed as mammalian cells generally accommodate lyssavirus codon usage
Expression construct design:
Consider incorporating a strong Kozak sequence for mammalian expression
For bacterial expression, reduce secondary structure in the 5' region of the mRNA
Generate both native and codon-optimized constructs
Compare expression levels in different systems
Assess protein functionality to ensure optimization doesn't affect folding or activity
For structural or functional studies, verify that codon optimization doesn't introduce artifacts
Understanding these patterns helps researchers optimize expression strategies for obtaining high yields of functional recombinant KHUV P protein.
Recent advancements offer new opportunities for studying KHUV P protein structure and interactions:
Allows visualization of P protein complexes with N or L proteins without crystallization
Can resolve dynamic regions that are challenging for X-ray crystallography
Single-particle analysis enables determination of structures at near-atomic resolution
Sample preparation: Purify protein complexes to high homogeneity and optimize buffer conditions to prevent aggregation
Maps protein-protein interaction surfaces by detecting changes in hydrogen-deuterium exchange rates
Identifies flexible regions and conformational changes upon binding
Particularly valuable for studying P protein interactions with host factors
Protocol optimization: Test various deuterium labeling times (10 seconds to 24 hours) to capture fast and slow exchanging regions
Provides accurate structural models without experimental structure determination
Can predict structures of P protein domains and their complexes
Useful for generating hypotheses about functional domains and guiding mutagenesis
Validation approach: Compare predictions with experimental data from limited proteolysis or HDX-MS
Combining multiple techniques (SAXS, NMR, cross-linking/mass spectrometry)
Creates comprehensive structural models of P protein complexes
Particularly useful for intrinsically disordered regions in P protein
Data integration: Use specialized software (e.g., IMP, HADDOCK) to combine various structural constraints
BioID or TurboID fused to P protein identifies proximal proteins in living cells
APEX2 fusion allows electron microscopy visualization of P protein localization
Maps the dynamic interactome of P protein during infection
Optimization: Compare different tagging positions to minimize functional disruption