In HSV-1, the UL11 protein is a 96-amino-acid tegument protein found on the cytoplasmic faces of nuclear, Golgi apparatus-derived, and plasma membranes of infected cells . It plays a role in virus budding, although its specific function remains unknown. The protein is abundant in virions, with approximately 700 copies per particle . The packaging of UL11 is selective for underphosphorylated forms, similar to other tegument proteins .
While specific research on the recombinant PsHV-1 UL11 protein is limited, studies on similar proteins in other herpesviruses provide valuable insights. For instance, UL21 in bovine herpesvirus 1 (BoHV-1) plays critical roles in viral secondary envelopment and cell-to-cell spreading, highlighting the importance of tegument proteins in viral replication . Similarly, UL11's role in HSV-1 suggests that tegument proteins are crucial for efficient viral budding and replication.
| Virus | UL11 Characteristics | Function | Packaging Determinants |
|---|---|---|---|
| HSV-1 | Myristylated, palmitylated, phosphorylated; 96 amino acids | Virus budding | Myristylation, palmitylation, acidic cluster motif |
| PsHV-1 | Limited data available | Presumed similar to HSV-1 | Unknown, potentially similar to HSV-1 |
UL11 in Psittacid herpesvirus 1 belongs to the conserved herpesvirus tegument protein family. Studies have revealed that UL11 is an intrinsically disordered protein (IDP) that undergoes liquid-liquid phase separation (LLPS) in vitro . Through techniques including circular dichroism, limited proteolysis, small-angle X-ray scattering (SAXS), and light microscopy, researchers have characterized its structural properties .
The protein contains:
A C-terminal region with a disordered structure
Fatty acyl modification sites for myristate and palmitate that mediate membrane binding
An acidic cluster and dileucine motif important for recycling from plasma membrane to Golgi apparatus
Methodological approach: To characterize UL11 structure, employ a combination of:
Expression and purification of recombinant PsHV-1 UL11 involves several methodological considerations:
Expression Systems:
Bacterial expression using E. coli (BL21 or similar strains) with vectors like pET or pGEX systems
For studying acylation and post-translational modifications, mammalian expression systems using HEK293T or chicken embryo fibroblasts (CEF) are preferred
Purification Protocol:
For bacterial expression: Use affinity tags (His, GST) followed by size exclusion chromatography
For mammalian expression: Concentrate virions from infected cell monolayers showing 90-95% cytopathic effect using PEG 8000 precipitation and pelleting through 30% sucrose cushion
Extract proteins using 10% sodium dodecyl sulfate and proteinase K treatment
For interaction studies: Use GST-pull down assays or co-immunoprecipitation methods to study binding partners
For optimal expression, culture conditions must be optimized to account for UL11's disordered nature, potentially using specialized media formulations like DMEM/F-12 supplemented with penicillin (50 μg/ml), streptomycin (50 μg/ml), and 10% fetal bovine serum .
UL11 demonstrates dynamic localization during the viral replication cycle:
Primarily accumulates on the cytoplasmic face of internal membranes
Cycles between the plasma membrane and Golgi apparatus
Trafficking to the Golgi apparatus is mediated by an acidic cluster and dileucine motif in UL11
Co-localizes with UL16 in a Golgi-like compartment when co-expressed
Methodological approach for studying localization:
Construct fluorescently tagged UL11 (GFP, mCherry) or use epitope tags (HA, FLAG)
Perform immunofluorescence microscopy with markers for cellular compartments
Use time-lapse microscopy to track trafficking patterns
Employ subcellular fractionation with Western blotting to confirm localization
Perform co-localization studies with Golgi markers and other viral proteins
The localization pattern is consistent across different alpha-herpesviruses, suggesting evolutionary conservation of UL11 trafficking mechanisms .
UL11 plays a critical role in secondary envelopment, as demonstrated by ultrastructural analyses of UL11-deleted mutants:
Mechanistic contributions:
Forms a critical bridge between nucleocapsids and envelopment membranes
Interacts with multiple viral proteins, particularly UL16, forming part of a protein network essential for envelopment
May facilitate the recruitment of cellular ESCRT machinery to envelopment sites
Deletion of UL11 homologs results in unenveloped capsid accumulation in the cytoplasm
Experimental evidence from comparative studies:
In Bovine herpesvirus 1 (BoHV-1), deletion of UL21 (which interacts with UL16, a UL11 binding partner) resulted in:
1,000-fold lower replication
85% smaller plaque size
Methodological approach for investigating envelopment:
Generate UL11-deletion mutants using BAC mutagenesis
Perform transmission electron microscopy to visualize envelopment defects
Quantify cytoplasmic versus enveloped virions
Use correlative light and electron microscopy to track UL11-tagged particles during envelopment
UL11 establishes a complex interaction network essential for viral assembly:
Key interaction partners:
UL16: Forms a stable complex with UL11; this interaction is conserved across alpha-herpesviruses
Capsid-associated proteins: Suggested interactions to bridge capsid and envelope
Experimental validation methods:
Co-immunoprecipitation (Co-IP):
Express tagged versions of UL11 (HA-tag) and potential partners
Pull down with appropriate antibodies
Analyze by Western blotting
GST pull-down assays:
Yeast two-hybrid screening:
Use UL11 as bait to screen for viral and cellular interactors
Validate hits with orthogonal methods
Proximity labeling techniques:
BioID or APEX2 fusions to identify proteins in close proximity
Mass spectrometry identification of labeled proteins
Förster Resonance Energy Transfer (FRET):
Tag UL11 and interacting partners with appropriate fluorophores
Measure energy transfer to confirm direct interactions
The UL11-UL16 interaction is particularly significant as UL11 homologs from pseudorabies and Marek's disease herpesviruses can also bind to HSV-1 UL16, suggesting evolutionary conservation of this interaction .
Recent research has revealed that UL11 belongs to a class of intrinsically disordered proteins (IDPs) with unique biophysical properties:
Characteristics and evidence:
UL11 undergoes liquid-liquid phase separation (LLPS) in vitro
The C-terminus has a disordered structure that fails to bind ribosomal RNA
This disorder is found in multiple tegument proteins, suggesting a general assembly mechanism
Functional implications:
LLPS may facilitate the concentration of viral components during assembly
Disordered regions provide conformational flexibility for multiple interactions
May contribute to the formation of viral assembly compartments as biomolecular condensates
Methodological approaches for studying LLPS and disorder:
In vitro phase separation assays:
Observe condensate formation under various conditions
Test effects of salt, pH, and RNA on phase behavior
Disorder prediction and validation:
Use computational tools (PONDR, IUPred) to predict disorder
Validate with CD spectroscopy and NMR
Mutagenesis of disordered regions:
Generate variants with altered disorder propensity
Assess impact on viral assembly and replication
Live-cell imaging of condensate formation:
Express fluorescently tagged UL11
Track formation of puncta during infection
| Tegument Protein | Disorder Prediction Score | LLPS Propensity | Key Interaction Partners |
|---|---|---|---|
| UL11 (HSV-1) | High | High | UL16, UL21 |
| UL11 (PsHV-1) | High | Predicted high | UL16 |
| pp28 (HCMV) | Moderate | Unknown | Viral capsid proteins |
Table: Comparative analysis of disorder characteristics across herpesvirus tegument proteins
The acidic cluster and dileucine motif of UL11 serve as critical functional domains:
Effects of mutations:
Disruption of the acidic cluster and dileucine motif prevents association with the 40 kDa protein (UL16)
These motifs are essential for recycling of UL11 from the plasma membrane to the Golgi apparatus
Mutations affect trafficking but not necessarily membrane binding, which is primarily mediated by fatty acylation
Experimental design for mutational analysis:
Generate a panel of UL11 mutants with:
Deletions or substitutions in the acidic cluster
Alterations in the dileucine motif
Combinations of both modifications
Assess each mutant for:
Ability to bind UL16 via co-immunoprecipitation
Subcellular localization using immunofluorescence
Complementation of UL11-null virus replication
Impact on secondary envelopment via electron microscopy
Quantitative measurements:
Binding affinities using surface plasmon resonance
Trafficking kinetics using live-cell imaging
Viral growth curves and plaque size analyses
Research findings:
Mutational studies revealed that while the entire second half of UL11 is not required for UL16 binding, the acidic cluster and dileucine motif are essential for this interaction . These motifs likely form a recognition surface that mediates both protein trafficking and establishment of the viral tegument protein network.
Cell-to-cell transmission is a critical aspect of herpesvirus pathogenesis and UL11 plays an essential role in this process:
Experimental approaches:
Co-culture systems:
Neutralizing antibody assays:
Live-cell imaging:
Express fluorescently tagged viral proteins
Track virological synapse formation between cells
Monitor virus movement across cell junctions
Virological synapse characterization:
Immunostain for UL11 at cell-cell contacts
Perform super-resolution microscopy to visualize synapse architecture
Use electron microscopy to examine ultrastructural features
Genome delivery quantification:
Data from related research:
In studies of HCMV (which has the UL99 homolog of UL11), cell-to-cell transmission was shown to deliver larger numbers of viral genomes compared to cell-free infection, and this process occurred despite the presence of neutralizing antibodies .
UL11 is conserved across herpesvirus subfamilies but exhibits important structural and functional variations:
Comparative features:
| Virus Family | UL11 Homolog | Size (aa) | Key Features | Special Functions |
|---|---|---|---|---|
| Alphaherpesviruses (HSV-1) | UL11 | 96 | Myristoylation, palmitoylation | Secondary envelopment |
| Betaherpesviruses (HCMV) | UL99 (pp28) | Larger | Leader sequences important for gene expression | Antiviral therapy effectiveness marker |
| Gammaherpesviruses | ORF38 | Variable | Less characterized | Unknown |
| Psittacid herpesvirus 1 | UL11 | Similar to HSV-1 | Conserved structure | Cell-to-cell transmission |
Research approaches for comparative studies:
Sequence alignment and phylogenetic analysis:
Identify conserved versus divergent regions
Map functional domains across viral species
Use computational methods to predict structural conservation
Complementation assays:
Express UL11 homologs from different viruses in UL11-null mutants
Assess functional complementation across species
Determine which features are universally conserved
Domain swapping experiments:
Create chimeric UL11 proteins with domains from different viral species
Test functionality in various assays
Identify minimal functional domains
Interactome comparison:
Characterize protein interaction networks for each UL11 homolog
Identify conserved versus species-specific interactions
Use proteomics approaches to map comprehensive interactomes
Studies have shown that UL11 homologs from pseudorabies and Marek's disease herpesviruses can interact with UL16 from HSV-1, indicating functional conservation despite sequence divergence . In beta-herpesviruses, the UL99 homolog has been determined to be an important factor in antiviral therapy research .
CRISPR-Cas9 technology offers powerful approaches for studying UL11 function in herpesvirus biology:
Methodological strategies:
Bacterial Artificial Chromosome (BAC) editing:
In situ viral genome editing:
Deliver CRISPR-Cas9 components to virus-infected cells
Target UL11 in replicating viral genomes
Use HDR templates to insert reporters or tags
Conditional knockout strategies:
Create destabilization domain-tagged UL11 variants
Enable conditional protein degradation to study timing effects
Use inducible promoters to control UL11 expression levels
High-throughput screening:
Design CRISPR library targeting multiple regions of UL11
Screen for phenotypes related to viral assembly and spread
Identify critical residues through deep mutational scanning
Experimental design considerations:
Include proper controls (wild-type and revertant viruses)
Verify editing by sequencing and protein expression analysis
Perform complementation assays with ectopically expressed UL11
Utilize multiple cell types to assess cell-type specific functions
Expected outcomes:
UL11 deletion would likely result in significant replication defects, particularly in cell-to-cell spread and secondary envelopment, as observed with the UL21 deletion in BoHV-1 which showed 1,000-fold lower replication and 85% smaller plaque size .