Recombinant Staphylococcus aureus UPF0435 protein SAS1802 (SAS1802) is a conserved bacterial protein belonging to the UPF (Uncharacterized Protein Family) 0435 classification. This protein is expressed in Staphylococcus aureus and can be produced recombinantly in expression systems such as Escherichia coli. As a member of the UPF family, it represents proteins with conserved sequences whose functions have not been fully characterized, making it a target for fundamental research into S. aureus biology and potential virulence mechanisms .
Unlike well-characterized S. aureus virulence factors such as Protein A (SpA) and Alpha-hemolysin (Hla), SAS1802 belongs to the uncharacterized protein family. While Protein A has five immunoglobulin-binding domains that capture both the Fc region and Fab region of immunoglobulins , and Alpha-hemolysin forms cylindrical transmembrane heptameric pores through interaction with ADAM10 receptors , SAS1802's specific structural elements and functional roles remain largely undefined. This fundamental difference places SAS1802 in the research category of proteins requiring primary characterization studies rather than the application-focused studies typical for established virulence factors .
Recombinant SAS1802 is typically expressed in prokaryotic expression systems, with E. coli being the most common host. The expression protocol generally involves:
Gene synthesis or PCR amplification of the SAS1802 coding sequence
Cloning into an expression vector (commonly pET series vectors)
Transformation into an expression strain (such as BL21(DE3))
Induction of protein expression using IPTG
Cell lysis and protein purification via affinity chromatography
The protein can be tagged (commonly with His6) to facilitate purification, or produced tag-free depending on the experimental requirements. Expression in eukaryotic systems such as yeast is less common but may be employed for specific applications requiring post-translational modifications .
The optimal conditions for expressing soluble recombinant SAS1802 in E. coli typically involve:
| Parameter | Recommended Condition | Notes |
|---|---|---|
| Expression strain | BL21(DE3) or Rosetta(DE3) | Rosetta strains provide rare tRNAs that may improve expression |
| Growth temperature | 18-25°C post-induction | Lower temperatures reduce inclusion body formation |
| IPTG concentration | 0.1-0.5 mM | Lower concentrations often improve solubility |
| Induction OD600 | 0.6-0.8 | Mid-log phase provides balance between yield and solubility |
| Post-induction time | 16-20 hours | Extended expression at lower temperatures |
| Media supplements | 2% glucose, 1% sorbitol | Osmolytes can enhance protein folding |
For challenging expression cases, fusion partners such as MBP (maltose-binding protein), SUMO, or thioredoxin may be employed to enhance solubility. The expression conditions should be empirically optimized through small-scale test expressions before scaling up to larger cultures for purification .
For structural studies requiring high-purity SAS1802, a multi-step purification strategy is recommended:
Initial capture: Immobilized metal affinity chromatography (IMAC) using Ni-NTA or Co-NTA resins if His-tagged
Intermediate purification: Ion exchange chromatography (IEX) based on the theoretical pI of SAS1802
Polishing step: Size exclusion chromatography (SEC) to remove aggregates and achieve monodisperse protein preparation
Optional tag removal: If a cleavable tag was used, insert a proteolytic cleavage step (TEV or PreScission protease) after the initial IMAC
Buffer optimization: Screen various buffer conditions during the final SEC step to identify stabilizing conditions
The final protein preparation should be assessed for purity by SDS-PAGE (>95%), homogeneity by dynamic light scattering (DLS), and structural integrity by circular dichroism (CD) spectroscopy before proceeding with crystallization or NMR studies .
To identify potential binding partners of SAS1802, a multi-faceted approach is recommended:
Pull-down assays: Use purified His-tagged SAS1802 as bait with S. aureus lysate as prey, followed by mass spectrometry identification of interacting proteins
Bacterial two-hybrid system: Express SAS1802 fused to one domain of a split reporter protein, and screen against a library of S. aureus proteins fused to the complementary domain
Co-immunoprecipitation: Generate antibodies against SAS1802 for immunoprecipitation from S. aureus lysates, followed by mass spectrometry analysis
Crosslinking-MS: Use chemical crosslinkers to stabilize transient protein-protein interactions in vivo, followed by purification and mass spectrometry
Proximity labeling: Express SAS1802 fused to a proximity labeling enzyme (e.g., BioID or APEX) in S. aureus to biotinylate nearby proteins for subsequent streptavidin pull-down and identification
Each approach has strengths and limitations, so combining multiple methods provides the most robust results. Control experiments using unrelated proteins or lysates from SAS1802 knockout strains are essential to identify specific versus non-specific interactions .
When crystallization attempts for SAS1802 have failed, alternative structural biology approaches can be considered:
NMR spectroscopy: For proteins <30 kDa, solution NMR can provide atomic-resolution structures. Express SAS1802 with 15N and 13C labeling, and collect multi-dimensional NMR data sets.
Cryo-electron microscopy (cryo-EM): Recent advances allow structure determination of smaller proteins. Consider:
Using antibody fragments to increase molecular weight
Embedding SAS1802 in nanodiscs or micelles if it has membrane-interacting regions
Utilizing advances in computational methods for small protein reconstruction
AlphaFold2 or RoseTTAFold prediction: Recent AI-based structural prediction methods have reached near-experimental accuracy. The predicted structure can be validated using limited experimental data from circular dichroism, SAXS, or crosslinking-MS.
Construct optimization: Design new constructs based on:
Secondary structure predictions to remove disordered regions
Limited proteolysis to identify stable domains
Fusion to crystallization chaperones like T4 lysozyme or rubredoxin
Each method provides complementary structural information, and integrative structural biology approaches combining multiple techniques often yield the most reliable results .
Multiple computational approaches can be employed to predict SAS1802's function:
Sequence-based methods:
PSI-BLAST for remote homology detection
Hidden Markov Models (HMMs) to identify functional domains
Genomic context analysis (examining neighboring genes)
Co-evolution analysis to identify functionally linked proteins
Structure-based methods:
Structural alignment against protein structure databases
Identification of catalytic triads or binding pockets
Electrostatic surface potential mapping
Normal mode analysis for potential conformational changes
Integrative approaches:
Molecular docking with potential substrates or binding partners
Molecular dynamics simulations to explore dynamics
Machine learning models integrating multiple features
Systems biology network analysis to place SAS1802 in functional networks
Here's an example of predicted functional partners based on genomic context and co-expression data:
| Protein | Function | Confidence Score | Evidence |
|---|---|---|---|
| RecA | DNA repair | 0.786 | Co-expression, genomic context |
| ClpC | ATP-dependent protease | 0.654 | Co-expression |
| FtsZ | Cell division protein | 0.543 | Genomic context |
| RsbU | Stress response sigma factor | 0.521 | Co-expression, text mining |
These predictions should be validated experimentally using the binding partner identification methods described earlier .
To study phenotypic effects of SAS1802 manipulation, design a comprehensive genetic system as follows:
Knockout construction:
Create a clean deletion using allelic exchange vectors (e.g., pMAD, pKOR1)
Design deletion to maintain the reading frame of surrounding genes
Confirm deletion by PCR, sequencing, and Western blot
Create complementation strain by reintroducing SAS1802 under native promoter
Conditional expression systems:
Tetracycline-inducible system (pRMC2 vector)
IPTG-inducible Pspac promoter system
Xylose-inducible system
Reporter fusions:
Transcriptional fusion (promoter-reporter)
Translational fusion (SAS1802-reporter)
Reporters: GFP, luciferase, β-galactosidase
Phenotypic characterization:
Growth curves under various conditions (different media, stress conditions)
Biofilm formation assays
Virulence in infection models
Proteomics and transcriptomics comparison
Metabolomics analysis
Advanced techniques:
CRISPRi for partial knockdown
Dual-fluorescent protein systems to monitor localization
Protein degradation systems for temporal control
The genetic system should include proper controls such as wild-type strain, vector-only control, and strains expressing unrelated proteins to distinguish specific from non-specific effects .
Incorporating SAS1802 into S. aureus vaccine development would follow this research pathway:
Antigenicity assessment:
Bioinformatic prediction of surface exposure and immunogenicity
Production of recombinant SAS1802 and testing for antibody recognition in convalescent patient sera
Epitope mapping to identify immunodominant regions
Vaccine formulation strategies:
Subunit vaccine: Purified recombinant SAS1802
Multi-antigen approach: Combine with established antigens (similar to rFSAV approach that uses Hla, SEB, SpA, IsdB-N2 and MntC)
Epitope-based: Synthetic peptides representing immunodominant epitopes
DNA vaccine: SAS1802 gene in expression vector
Adjuvant selection:
Alum-based formulations
TLR agonists (e.g., CpG, MPLA)
Liposomal or nanoparticle delivery systems
Immune response characterization:
Antibody titers and isotype profiles
T-cell responses (Th1/Th2/Th17 balance)
Functional assays (opsonophagocytosis, neutralization)
Protection studies:
Challenge models (bacteremia, skin infection, pneumonia)
Passive immunization with anti-SAS1802 antibodies
Assessment of bacterial clearance and disease parameters
This approach parallels the successful development of the recombinant five-antigen S. aureus vaccine (rFSAV), which demonstrated protection in multiple infection models and induced comprehensive cellular and humoral immune responses .
To investigate SAS1802's potential role in antibiotic resistance, employ these approaches:
Susceptibility testing:
Determine minimum inhibitory concentrations (MICs) for various antibiotics in wild-type, SAS1802 knockout, and SAS1802-overexpressing strains
Time-kill assays to assess killing kinetics
Population analysis profiles to identify heteroresistant subpopulations
Resistance mechanism studies:
Membrane permeability assays (uptake of fluorescent dyes)
Drug accumulation/efflux assays using radiolabeled or fluorescent antibiotics
Biofilm formation and antibiotic tolerance assessment
Transcriptomic/proteomic analysis:
RNA-Seq comparing wild-type and SAS1802 mutant strains ± antibiotic exposure
Quantitative proteomics to identify differentially expressed resistance determinants
ChIP-Seq if SAS1802 might have DNA-binding properties affecting resistance gene expression
Direct interaction studies:
In vitro binding assays between purified SAS1802 and antibiotics
Structural studies of SAS1802-antibiotic complexes
Surface plasmon resonance to determine binding kinetics
Evolution experiments:
Serial passage in increasing antibiotic concentrations
Whole genome sequencing to identify compensatory mutations
Competition assays between wild-type and mutant strains
The experimental design should include appropriate controls and be performed in multiple S. aureus strain backgrounds to account for strain-specific effects on resistance phenotypes .
SAS1802 can serve as a model system for understanding UPF0435 family proteins through these approaches:
Comparative structural analysis:
Determine high-resolution structure of SAS1802 using X-ray crystallography, NMR, or cryo-EM
Perform structural alignment with other UPF family members
Identify conserved structural motifs that may indicate function
Structure-function studies:
Site-directed mutagenesis of conserved residues
Activity assays to correlate structural features with function
Hydrogen-deuterium exchange mass spectrometry to identify dynamic regions
Protein-protein interaction networks:
Yeast two-hybrid or bacterial two-hybrid screens
Affinity purification-mass spectrometry using SAS1802 as bait
Comparison of interaction partners across UPF family members
Evolutionary analysis:
Phylogenetic analysis of UPF0435 family across bacterial species
Identification of co-evolving residues suggesting functional constraints
Genomic context analysis across species
In silico structure prediction validation:
Use experimentally determined SAS1802 structure to assess accuracy of AlphaFold2/RoseTTAFold predictions
Develop improved prediction methods for UPF family proteins
Create structure-based functional annotation pipeline
This research would contribute to understanding uncharacterized protein families that comprise a significant portion of bacterial genomes and may reveal novel drug targets or biological mechanisms .
Protein aggregation of recombinant SAS1802 can be addressed using these strategies:
Buffer optimization:
Screen various pH conditions (typically pH 6.0-8.0)
Test different salt concentrations (100-500 mM NaCl)
Add stabilizing agents:
Glycerol (5-20%)
Arginine (50-200 mM)
Tween-20 or other non-ionic detergents (0.01-0.05%)
Expression modifications:
Lower induction temperature (16-18°C)
Reduce IPTG concentration (0.1-0.2 mM)
Co-express with molecular chaperones (GroEL/ES, DnaK/J)
Use solubility-enhancing fusion tags (MBP, SUMO, TrxA)
Purification adjustments:
Include reducing agents (1-5 mM DTT or TCEP)
Add protease inhibitors throughout purification
Maintain protein at moderate concentration (<2 mg/mL)
Avoid freeze-thaw cycles; store at 4°C for short-term use
Refolding approaches (if inclusion bodies are unavoidable):
On-column refolding during IMAC purification
Systematic screening of refolding conditions using a matrix approach
Pulsatile refolding with stepwise reduction of denaturant
Analytical techniques to monitor improvement:
Dynamic light scattering to assess homogeneity
Thermal shift assays to identify stabilizing conditions
Size exclusion chromatography with multi-angle light scattering (SEC-MALS)
Each protein has unique characteristics, so empirical testing of multiple conditions is essential to identify the optimal solution for SAS1802 .
Robust experimental design for studying SAS1802 function should include these controls:
Genetic controls:
Wild-type strain (positive control)
SAS1802 knockout strain (negative control)
Complemented strain (knockout with reintroduced SAS1802)
Strain expressing inactive SAS1802 mutant (if catalytic residues identified)
Strain expressing unrelated protein of similar size/properties
Protein-level controls:
Purified wild-type SAS1802 protein
Heat-denatured SAS1802 (negative control)
Point mutants affecting predicted functional sites
Related protein from same family (specificity control)
Unrelated protein of similar size/properties
Assay-specific controls:
For binding assays: non-specific binding controls (e.g., GST-only)
For enzymatic assays: no-enzyme and no-substrate controls
For cell-based assays: vehicle-only and irrelevant protein controls
For animal studies: sham-treated and irrelevant protein controls
Technical controls:
Biological replicates (different bacterial cultures/protein preparations)
Technical replicates (repeated measurements)
Blinding of samples during analysis when possible
Inclusion of internal standards for quantitative measurements
Validation approaches:
Use multiple methodologies to address the same question
Perform dose-response or time-course experiments
Include positive controls with known outcomes
Reverse the experimental approach (e.g., gain-of-function and loss-of-function)
Proper controls ensure that observed effects are specifically attributable to SAS1802 rather than experimental artifacts or general stress responses .
Resolving contradictions between in vitro and in vivo results requires a systematic approach:
Critically evaluate experimental conditions:
Are in vitro conditions physiologically relevant? (pH, salt, cofactors)
Does the recombinant protein have the same modifications as in vivo?
Are protein concentrations comparable between systems?
Are there interacting partners present in vivo but absent in vitro?
Bridge the gap with intermediate approaches:
Cell extract-based assays (partially purified system)
Permeabilized cell assays (maintain cellular organization)
Ex vivo assays using freshly isolated components
Reconstitution experiments with purified interaction partners
Advanced in vivo techniques:
FRET/BRET to monitor protein interactions in live cells
Activity-based protein profiling in intact cells
Chemical crosslinking in vivo before analysis
Single-cell analysis to detect population heterogeneity
Resolve temporal and spatial factors:
Time-course experiments to capture transient effects
Subcellular localization studies
Regulated expression systems to control timing/levels
Stress or environmental conditions that might activate function
Integrated data analysis:
Develop mathematical models incorporating both datasets
Use Bayesian approaches to update hypotheses based on all evidence
Consider emergent properties of complex systems
Look for conditional phenotypes dependent on specific conditions
This systematic approach often reveals that both results are correct under their specific conditions, and the contradiction reflects biological complexity rather than experimental error .
Several cutting-edge technologies could significantly advance SAS1802 research:
CRISPR interference/activation systems:
CRISPRi for precise gene knockdown with temporal control
CRISPRa for upregulation of SAS1802 in specific contexts
Multiplexed CRISPR screens to identify genetic interactions
Single-cell technologies:
Single-cell RNA-seq to detect heterogeneous responses
Single-cell proteomics to capture protein-level variation
Microfluidic systems for monitoring bacterial responses to stressors
Advanced imaging:
Super-resolution microscopy for precise localization
Live-cell imaging with photoactivatable tags
Correlative light-electron microscopy for ultrastructural context
Expansion microscopy for nanoscale visualization
Protein interaction mapping:
Proximity labeling (BioID, APEX) for in vivo interactome mapping
Cross-linking mass spectrometry for structural interactomics
Thermal proteome profiling to detect ligand interactions
Protein correlation profiling across fractionation schemes
Host-pathogen interface:
Dual RNA-seq of host-pathogen interactions
Organoid infection models for tissue-specific responses
Ex vivo tissue infection models with live imaging
Humanized mouse models for improved translation
These technologies provide unprecedented resolution and scale for understanding protein function within complex biological systems and could reveal unexpected roles for SAS1802 in S. aureus biology and pathogenesis .
Comparative genomics approaches can provide valuable insights into SAS1802 function:
Conservation analysis:
Presence/absence of SAS1802 across S. aureus strains (core vs. accessory genome)
Sequence conservation levels indicating selective pressure
Identification of hypervariable regions suggesting immune interaction
Correlation with strain virulence or host adaptation
Synteny analysis:
Conservation of genomic context across strains
Co-evolution with neighboring genes
Operon structure variations
Mobile genetic element associations
Variant impact prediction:
Nonsynonymous/synonymous substitution ratios (dN/dS)
Identification of positively selected residues
Mapping variants to predicted structural domains
Correlation of variants with phenotypic differences
Transcriptional regulation:
Conservation of promoter regions
Regulatory motif identification
Transcription factor binding site conservation
sRNA interaction site analysis
Correlation with strain phenotypes:
Antibiotic resistance profiles
Host range and tissue tropism
Virulence in different infection models
Growth characteristics and metabolic capabilities
This comprehensive analysis could reveal patterns linking SAS1802 sequence variations to specific strain characteristics, providing testable hypotheses about its function and importance in different ecological niches .
Developing high-throughput screening (HTS) assays for SAS1802 modulators requires a strategic approach:
Target-based biochemical assays:
Thermal shift assays to detect ligand binding
Fluorescence polarization for detecting interactions with labeled partners
FRET/TR-FRET between SAS1802 and binding partners
AlphaScreen for protein-protein interaction disruption
Phenotypic screening approaches:
Reporter strains (e.g., SAS1802 promoter driving luciferase expression)
Growth inhibition assays in SAS1802-dependent conditions
Stress response profiles in wild-type vs. SAS1802 mutant strains
Virulence factor expression readouts
Fragment-based approaches:
NMR-based fragment screening
Surface plasmon resonance fragment screening
Mass spectrometry for detecting covalent binders
Crystallographic fragment screening
Virtual screening methods:
Structure-based docking against SAS1802 models
Pharmacophore modeling based on interaction patterns
Molecular dynamics simulations to identify cryptic binding sites
Machine learning-based virtual screening
Assay optimization considerations:
Miniaturization to 384 or 1536-well format
Z'-factor optimization (aim for >0.7)
DMSO tolerance assessment
Counter-screening assays to filter false positives