Recombinant Human cytomegalovirus Protein UL41A (UL41A) is a gene product of Human cytomegalovirus (HCMV), a member of the Herpesviridae family. While UL41A’s natural expression and function remain poorly characterized, its recombinant form (expressed in E. coli) has been studied for potential roles in viral pathogenesis and immune modulation. The protein is 78 amino acids long (full-length) and is annotated as a putative viral membrane (VM) protein, though its biological significance is still under investigation .
UL41A is synthesized as a recombinant protein with a His-tag for purification. Key physicochemical properties include:
Property | Value | Source |
---|---|---|
Molecular Weight | 17.9 kDa | |
Theoretical pI | 9.82 | |
Charged Residues (Asp+Glu) | 8 | |
Positively Charged Residues (Arg+Lys) | 24 | |
Total Atoms | 2,542 | |
Aliphatic Index | 72.18 | |
Hydropathicity (GRAVY) | 0.122 |
The protein lacks documented post-translational modifications in recombinant forms, which may differ from its native state .
While UL41A’s endogenous function is uncharacterized, its inclusion in vaccine designs suggests potential immunogenicity:
Epitope Mapping: UL41A contains cytotoxic T lymphocyte (CTL) epitopes (e.g., RTANSTAGY, FCSTLKAFY) and B cell epitopes (e.g., ETTVWEKRRMESDTDF), indicating immune recognition during infection .
Vaccine Targeting: A multi-epitope vaccine construct (MEVC-UL41A) was designed to elicit broad immune responses, though its efficacy remains untested in vivo .
Immune Evasion: Similar to other HCMV proteins (e.g., US3, UL40), UL41A may modulate host immune responses to favor viral persistence .
Viral Replication: Though speculative, UL41A’s transmembrane domain (VM classification) suggests possible involvement in viral entry or membrane remodeling .
A recombinant UL41A-based vaccine (MEVC-UL41A) was engineered to combine CTL, B cell, and helper T lymphocyte (HTL) epitopes. Key features include:
Component | Sequence Example | Function |
---|---|---|
CTL Epitope | RTANSTAGY | T cell activation |
B Cell Epitope | ETTVWEKRRMESDTDF | Antibody binding |
HTL Epitope | ITKIMLARRKARAMV | T helper cell activation |
The construct’s antigenicity score (0.71) indicates moderate immunogenic potential, though lower than other HCMV targets like US3 (0.83) .
UL41A’s recombinant form was docked with human Toll-like receptor 4 (TLR4) to assess potential immune modulation:
Interaction Type | Count | Docking Score |
---|---|---|
Salt Bridges | 2 | −336.44 |
Hydrogen Bonds | 7 | |
Non-Bonded Contacts | 225 |
These results suggest weak binding affinity to TLR4, limiting its direct role in innate immune activation .
Property | Value |
---|---|
Molecular Weight | 17.9 kDa |
Theoretical pI | 9.82 |
Hydropathicity (GRAVY) | 0.122 |
Aliphatic Index | 72.18 |
Interaction Type | Count | Docking Score |
---|---|---|
Salt Bridges | 2 | −336.44 |
Hydrogen Bonds | 7 | |
Non-Bonded Contacts | 225 |
KEGG: vg:3077475
UL41A is a protein encoded by the Human cytomegalovirus genome. Like other HCMV proteins, it may contain transmembrane domains that can be predicted using bioinformatic tools such as Phobius, PureseqTM, and TMHMM . These computational methods identify potential membrane-spanning regions that could indicate association with viral envelopes or cellular membranes. Structural characterization typically begins with these in silico predictions before proceeding to experimental approaches like X-ray crystallography or cryo-electron microscopy. Researchers should apply multiple prediction algorithms and compare results across different HCMV strains to identify conserved structural features.
Expression patterns of HCMV proteins vary significantly between latent and lytic phases. To determine UL41A's expression profile, researchers should employ quantitative proteomics approaches similar to the iTRAQ-based methods used for other HCMV proteins . These techniques have successfully identified differentially expressed proteins between latent and reactivated virus. For UL41A analysis, collecting samples at various timepoints and using both transcriptomic (qRT-PCR) and proteomic (Western blotting) validation methods would reveal its temporal expression pattern. This approach has confirmed that other HCMV proteins show consistent patterns between mRNA and protein levels during different infection phases .
Analyzing conservation patterns requires comparison of UL41A sequences across multiple HCMV strains, including both laboratory-adapted and clinical isolates. Researchers have developed comprehensive approaches examining nine different CMV strains for transmembrane protein identification . For UL41A analysis, sequence alignment should identify regions with high conservation (likely functional domains) versus variable regions (potentially adaptable to different environments). Conservation analysis across the 335 available CMV genomes in GenBank could provide substantial insight into essential versus dispensable regions of UL41A .
Production of recombinant UL41A should consider potential transmembrane domains identified through bioinformatic analysis . If transmembrane regions are present, eukaryotic expression systems are preferable to bacterial systems for maintaining proper protein folding and post-translational modifications. Expression constructs should include appropriate epitope tags for purification and detection, similar to the FLAG-tagged constructs used in VPS4A studies . Researchers should optimize expression conditions by testing different host systems, induction parameters, and detergents for membrane protein solubilization. Purification typically involves affinity chromatography followed by size exclusion chromatography to ensure homogeneity.
Generating UL41A-deficient viral strains requires markerless BAC mutagenesis, similar to approaches used for creating pUL71 mutants . This method allows precise gene deletion or introduction of specific mutations without leaving selection markers in the viral genome. Researchers should design mutations that either completely ablate UL41A expression or target specific functional domains. When introducing mutations, appropriate revertant viruses must be generated as controls to ensure phenotypic changes result specifically from the intended mutations. Successful mutagenesis should be confirmed by sequencing and protein expression analysis before proceeding to functional studies .
For investigating UL41A protein interactions, researchers should employ multiple complementary approaches. Co-immunoprecipitation experiments followed by mass spectrometry can identify potential binding partners, as demonstrated with other HCMV proteins . For visualization of protein-protein interactions in living cells, bimolecular fluorescence complementation (BiFC) has proven effective, as shown in studies of pUL71-VPS4A interactions during HCMV infection . This technique involves splitting a fluorescent protein (e.g., Citrine) into two non-fluorescent fragments and fusing them to potentially interacting proteins. If interaction occurs, fluorescence is reconstituted, providing spatial information about where in the cell these interactions take place .
Investigating UL41A's potential role in the cVAC requires immunofluorescence microscopy to determine its localization during infection. Similar studies with pUL71 have shown that it localizes to the cVAC and recruits cellular proteins essential for viral assembly . To study UL41A's function in this context, researchers should perform co-localization studies with known cVAC markers and examine how UL41A-deficient viruses affect cVAC formation. Electron microscopy of cells infected with UL41A-mutant viruses would reveal ultrastructural changes in viral particle formation. If UL41A contributes to assembly, researchers would expect phenotypes similar to those observed with pUL71 mutations, including defects in secondary envelopment and reduced viral titers .
HCMV proteins frequently use molecular mimicry to recruit and repurpose host cell proteins. This has been elegantly demonstrated with pUL71, which contains a MIM2-like motif that mimics cellular ESCRT-III proteins to recruit VPS4A to viral assembly sites . For UL41A, researchers should examine its sequence for short linear motifs that resemble host protein interaction domains. Potential mimicry can be validated through structure-informed mutagenesis of key residues, followed by interaction studies and functional assays. Identifying such mechanisms would reveal how UL41A potentially contributes to viral evasion of host defenses or recruitment of cellular machinery for viral replication .
The impact of viral proteins on host cell secretion pathways represents an important aspect of virus-host interaction. Studies have shown that during latent HCMV infection, the virus modifies the cellular secretome to first recruit CD4+ T cells by increasing CCL8 expression and then inhibits their antiviral functions . To investigate UL41A's influence on the secretome, researchers should compare media from cells infected with wild-type versus UL41A-deficient viruses using mass spectrometry. Any differentially secreted proteins should be validated using ELISA and functional assays to determine how these changes might affect immune response or cellular function during infection.
Membrane-associated viral proteins often present solubility challenges during expression and purification. If UL41A contains predicted transmembrane domains , researchers should test various detergents (DDM, LMNG, or digitonin) for solubilization. Alternatively, truncated constructs that remove transmembrane regions while preserving functional domains may yield soluble protein. Fusion partners like maltose-binding protein can enhance solubility, though researchers must verify that such modifications don't disrupt protein function. Temperature optimization during expression (typically lowering to 16-18°C) and inclusion of stabilizing agents in purification buffers can further improve yields of correctly folded protein.
Validating antibody specificity is crucial for reliable immunodetection of UL41A. Multiple controls should be employed, including: (1) cells infected with UL41A-knockout virus to confirm absence of signal; (2) overexpression of tagged UL41A to verify co-localization with antibody signal; and (3) pre-absorption of antibody with recombinant UL41A to demonstrate signal reduction. Western blotting should show a band of appropriate molecular weight that disappears in knockout samples. For immunofluorescence studies, signal specificity can be confirmed using approaches similar to those employed for pUL71 detection, where antibody staining patterns were compared between wild-type and mutant virus infections .
When analyzing UL41A's impact on viral replication, appropriate controls are critical for accurate interpretation. These should include: (1) wild-type parental virus; (2) UL41A-deficient virus; and (3) a revertant virus where the wild-type sequence has been restored, similar to the control strategy used in pUL71 studies . The revertant control ensures that any observed phenotypes result specifically from UL41A modification rather than unintended mutations elsewhere in the viral genome. Growth curves should include multiple timepoints to capture potential differences in replication kinetics, and experiments should be performed in relevant cell types that support the full viral life cycle. Viral titers should be determined using multiple independent methods, such as plaque assays and qPCR quantification of viral genomes.
Recent research has shown that HCMV protein pUL71 contains a viral MIM2 (vMIM2) motif that directly interacts with the MIT domain of cellular VPS4A, an essential component of the ESCRT machinery involved in membrane remodeling . This interaction recruits VPS4A to the cytoplasmic virion assembly compartment during infection. Researchers investigating UL41A should examine whether it contains similar motifs or interacts with other ESCRT components. Techniques such as co-immunoprecipitation, BiFC, and immunofluorescence microscopy can determine if UL41A co-localizes with ESCRT machinery. Creating recombinant viruses with mutations in potential interaction motifs would reveal the functional significance of such interactions for viral replication .
Developing inhibitors targeting UL41A function requires establishing robust high-throughput screening assays. If UL41A has enzymatic activity, biochemical assays using purified recombinant protein can screen for direct inhibitors. Alternatively, cell-based assays using reporter systems linked to UL41A function can identify compounds that disrupt its activity in a cellular context. Researchers should develop counter-screens to eliminate compounds with non-specific activity or cytotoxicity. Hit compounds should be validated using orthogonal assays and tested against UL41A mutants to confirm target specificity. Structure-activity relationship studies would guide optimization of lead compounds toward higher potency and selectivity.
HCMV employs multiple strategies to evade host immune responses, including modulation of the cellular secretome to inhibit T cell function . To investigate UL41A's potential role in immune evasion, quantitative proteomics approaches like iTRAQ can compare protein expression profiles between cells infected with wild-type and UL41A-deficient viruses . Researchers should focus on changes in immune-related proteins, particularly those involved in antigen presentation, cytokine signaling, or pattern recognition receptor pathways. Findings from proteomics should be validated using qRT-PCR and Western blotting, followed by functional immunological assays to determine how observed protein changes affect immune cell recognition and activation .