Recombinant Salmonella Paratyphi B Protein AaeX(aaeX) is a 67-amino-acid polypeptide (UniProt ID: A9N864) fused to an N-terminal His tag for purification. It is produced in Escherichia coli systems and stored as a lyophilized powder .
The protein sequence is:
MSLFPVIVVFGLSFPPIFFELLLSLAIFWLVRRMLVPTGIYDFVWHPALFNTALYCCLFYLISRLFV
This sequence corresponds to the full-length aaeX gene product (1-67aa), which is annotated as a putative membrane-associated protein .
While AaeX itself is not directly linked to vaccine candidates, studies on related Salmonella proteins (e.g., Vi capsular polysaccharide in S. Paratyphi A) highlight the utility of recombinant proteins in designing conjugate vaccines . AaeX could serve as an antigen for antibody production or epitope mapping.
Metabolomic profiling of Salmonella infections has identified serovar-specific biomarkers . AaeX could contribute to developing targeted assays for distinguishing Paratyphi B from other serovars.
Immunogenicity: No direct data on AaeX’s immunogenicity exist, but homologous proteins in Salmonella Typhi and Paratyphi A elicit partial immune protection .
Pathogenic Role: aaeX is implicated in membrane integrity and virulence, though mechanistic studies are lacking .
Cross-Reactivity: Prior exposure to Salmonella Typhi or Paratyphi A does not confer cross-protection against Paratyphi B, underscoring the need for serovar-specific research .
Functional Studies: Elucidate AaeX’s role in bacterial adhesion, biofilm formation, or host-cell invasion.
Therapeutic Exploration: Evaluate its potential as a drug target or vaccine component using animal models.
KEGG: spq:SPAB_04194
AaeX is a small membrane protein consisting of 67 amino acids found in Salmonella paratyphi B. The protein has the amino acid sequence: MSLFPVIVVFGLSFPPIFFELLLSLAIFWLVRRMLVPTGIYDFVWHPALFNTALYCCLFY LISRLFV . This protein belongs to a family of membrane-associated proteins thought to be involved in bacterial response to environmental stresses. While its exact function remains under investigation, research suggests it may play a role in membrane integrity, potentially contributing to the bacterium's survival under adverse conditions. Understanding AaeX is significant because Salmonella paratyphi B is associated with enteric fever, particularly in travelers to certain regions such as the Andean region of South America .
Recombinant AaeX protein is typically produced using Escherichia coli expression systems, with the addition of a histidine tag (His-tag) at the N-terminus to facilitate purification . The methodological approach involves:
Cloning the full-length aaeX gene (encoding amino acids 1-67) into an expression vector
Transforming the recombinant plasmid into a compatible E. coli strain
Inducing protein expression under optimized conditions
Cell lysis and protein extraction
Purification using affinity chromatography (utilizing the His-tag)
Quality assessment through SDS-PAGE (typically achieving >90% purity)
Lyophilization for storage stability
The resulting lyophilized protein can be reconstituted in deionized sterile water to concentrations of 0.1-1.0 mg/mL, with recommended addition of 5-50% glycerol for long-term storage at -20°C or -80°C .
For optimal stability and activity of recombinant AaeX protein, the following handling protocols are recommended:
Storage Parameter | Recommended Protocol |
---|---|
Storage temperature | -20°C to -80°C |
Storage buffer | Tris/PBS-based buffer, 6% Trehalose, pH 8.0 |
Reconstitution | Deionized sterile water to 0.1-1.0 mg/mL |
Long-term preservation | Add glycerol to 5-50% final concentration |
Working aliquots | Store at 4°C for up to one week |
Freeze-thaw cycles | Minimize; repeated cycles not recommended |
Pre-use preparation | Brief centrifugation to bring contents to bottom |
Researchers should note that proper aliquoting after initial reconstitution is crucial to prevent protein degradation from repeated freeze-thaw cycles . For experimental reproducibility, it is advisable to document the specific storage conditions used when reporting research findings involving this protein.
Using recombinant AaeX protein for immunogenicity studies requires careful experimental design that addresses several methodological considerations:
Given that paratyphoid fever is a severe illness often acquired during international travel, particularly to regions such as South Asia and the Andean region of South America , these immunogenicity studies have significant translational implications.
While the search results don't directly link AaeX to antimicrobial resistance in Salmonella paratyphi B, research into this question would be valuable given the rising concerns about antimicrobial resistance in Salmonella species. Studies have shown that Salmonella Paratyphi A isolates frequently display nalidixic acid resistance and decreased susceptibility to ciprofloxacin, particularly in isolates from South Asia . A methodological approach to investigate potential roles of AaeX in antimicrobial resistance could include:
Gene knockout studies:
Create aaeX deletion mutants in Salmonella paratyphi B
Compare minimum inhibitory concentrations (MICs) of various antibiotics between wild-type and ΔaaeX strains
Complement the mutant with functional aaeX to confirm phenotype restoration
Expression correlation analysis:
Analyze aaeX expression levels under antibiotic stress using qRT-PCR
Compare expression between susceptible strains and resistant isolates
Perform RNA-seq to identify co-regulated genes in response to antibiotic exposure
Protein interaction studies:
Use pull-down assays with His-tagged AaeX to identify interaction partners
Investigate if AaeX interacts with known efflux pumps or regulatory proteins
Perform bacterial two-hybrid assays to validate protein-protein interactions
Membrane permeability assessment:
Evaluate if AaeX affects membrane permeability using fluorescent dyes
Compare antibiotic accumulation in wild-type vs. ΔaaeX strains
Analyze membrane lipid composition changes associated with AaeX expression
This investigation would be particularly relevant given that 90% of Salmonella Paratyphi B isolates in certain studies were susceptible to all antimicrobial agents tested, while resistance patterns differ significantly in Salmonella Paratyphi A .
The interaction between AaeX and virulence factors encoded by Salmonella pathogenicity islands (SPIs) represents an important research direction. While the search results primarily discuss SPI-related genes in Salmonella Paratyphi A rather than B , the methodological approaches can be adapted for AaeX studies:
Transcriptional analysis:
RNA-seq comparing wild-type and aaeX mutant strains under SPI-inducing conditions
qRT-PCR validation of differentially expressed SPI genes
Promoter-reporter fusions to monitor SPI gene expression in various genetic backgrounds
Secretion system functionality assays:
Western blot analysis of secreted effector proteins in culture supernatants
β-lactamase fusion assays to quantify translocation efficiency of SPI effectors
Microscopy-based approaches to visualize T3SS needle complex formation
In vitro infection models:
Cell invasion assays using epithelial cell lines (e.g., Caco-2, HeLa)
Intracellular survival assays in macrophage-like cells (e.g., THP-1, RAW264.7)
Analysis of SPI-dependent phenotypes such as:
Actin cytoskeleton rearrangements
Inflammatory response modulation
Intracellular trafficking alterations
Co-immunoprecipitation studies:
Use anti-His antibodies to pull down AaeX-His and identify co-precipitating SPI proteins
Perform reverse co-IP with antibodies against key SPI proteins
Mass spectrometry analysis of protein complexes
These approaches would be particularly valuable since studies of Salmonella Paratyphi A have identified genetic differences in several SPI-1 and SPI-2 genes that encode essential virulence mechanisms for bacterial entry and survival within host cells .
When designing experiments using recombinant AaeX protein, the following controls and validation steps are essential to ensure reliable and reproducible results:
Additionally, researchers should perform endotoxin testing of the final recombinant protein preparation, especially for immunological studies, to prevent lipopolysaccharide contamination from the E. coli expression system from confounding experimental results.
To investigate AaeX's potential roles in host-pathogen interactions, a comprehensive experimental approach should include:
Expression analysis during infection:
Quantify aaeX expression during different stages of infection
Compare expression across multiple infection models (cell lines, animal models)
Analyze expression in response to host defense mechanisms
Genetic manipulation approaches:
Generate clean aaeX deletion mutants using lambda Red recombinase system
Create complemented strains with wild-type aaeX under native or inducible promoters
Develop point mutants to identify critical amino acid residues
Cell culture infection models:
Epithelial cell adhesion and invasion assays
Macrophage survival and replication studies
Dendritic cell activation and cytokine production analysis
Ex vivo tissue models:
Intestinal organoid infection studies
Polarized epithelial cell monolayer translocation assays
Human blood infection models to assess survival in serum
In vivo infection models:
Mouse infection studies comparing wild-type and ΔaaeX strains
Colonization assessment across different tissues
Competitive index assays between wild-type and mutant strains
Immune response characterization (antibody production, T-cell responses)
Host response evaluation:
Transcriptomics of infected host cells
Cytokine/chemokine profiling
Inflammasome activation assessment
Analysis of autophagy and other cellular defense mechanisms
These approaches would be particularly relevant given that paratyphoid fever is a severe illness with a high hospitalization rate (62% in one study), and understanding host-pathogen interactions could inform treatment and prevention strategies .
Investigating protein-protein interactions involving membrane proteins like AaeX presents several methodological challenges that require specific analytical approaches:
Membrane protein solubility issues:
Detection sensitivity limitations:
Challenge: Potentially weak or transient interactions may be missed
Solution: Implement crosslinking strategies prior to co-immunoprecipitation
Validation: Use proximity labeling techniques (BioID, APEX) as complementary approaches
False positives in pull-down assays:
Challenge: His-tagged proteins may non-specifically bind bacterial proteins
Solution: Include stringent washing steps and appropriate negative controls
Validation: Confirm interactions using reverse pull-downs and alternative tags
Structural context loss in vitro:
Challenge: Membrane environment is crucial for native protein conformation
Solution: Reconstitute AaeX in nanodiscs or liposomes before interaction studies
Validation: Compare results from detergent-solubilized vs. membrane-reconstituted systems
Data analysis complexity:
Challenge: Distinguishing significant interactions from background
Solution: Apply statistical filters and enrichment thresholds to mass spectrometry data
Validation: Implement computational network analysis to identify functionally related interactors
Physiological relevance verification:
Challenge: In vitro interactions may not occur in vivo
Solution: Verify key interactions using in vivo techniques like FRET or split-protein complementation
Validation: Correlate interaction data with phenotypic outcomes in genetic studies
By systematically addressing these challenges, researchers can generate more reliable data on AaeX's interaction partners, potentially revealing connections to virulence mechanisms similar to those observed in studies of Salmonella Paratyphi A pathogenicity islands .
Reconciling discrepancies between in vitro and in vivo findings is a common challenge in bacterial protein research. For AaeX studies, researchers should implement the following methodological approaches:
Systematic comparison of experimental conditions:
Document all parameters that differ between in vitro and in vivo systems
Create intermediate models that bridge the complexity gap (e.g., ex vivo tissue systems)
Gradually increase complexity to identify where discrepancies emerge
Contextual expression analysis:
Compare aaeX expression levels between laboratory media and infection models
Identify environmental triggers that may activate or repress aaeX in vivo
Adjust in vitro conditions to better mimic the in vivo expression context
Functional redundancy assessment:
Identify potential compensatory mechanisms active in vivo but absent in vitro
Create multiple knockout strains to address redundant pathways
Perform transcriptome analysis to identify differentially regulated genes in vivo
Host factor incorporation:
Supplement in vitro systems with specific host factors identified in vivo
Develop co-culture systems with relevant host cells
Use host-derived environmental cues (e.g., bile acids, antimicrobial peptides)
Temporal dynamics consideration:
Implement time-course analyses for both in vitro and in vivo experiments
Identify critical time points where phenotypes diverge
Develop mathematical models to explain temporal discrepancies
Strain background effects:
Test aaeX mutations in multiple clinical isolates, not just laboratory strains
Consider genomic context differences between strains used in different experiments
Analyze if virulence factor variations contribute to discrepant results, similar to how genetic differences in SPI genes affected Salmonella Paratyphi A virulence
By systematically addressing these areas, researchers can develop more comprehensive models of AaeX function that account for the complex environments encountered during actual infection processes.
Several cutting-edge technologies offer promising approaches to elucidate AaeX structure-function relationships:
Cryo-electron microscopy (Cryo-EM):
AlphaFold and deep learning predictions:
Application: Generate computational models of AaeX structure and protein-protein interactions
Advantage: Provides structural hypotheses when experimental structures are unavailable
Implementation: Validate predictions through targeted mutagenesis of predicted functional domains
Single-molecule tracking in live bacteria:
Application: Visualize AaeX localization and dynamics during infection
Advantage: Reveals spatial and temporal aspects of protein function
Implementation: Create fluorescent protein fusions with AaeX while preserving membrane topology
CRISPR interference (CRISPRi) with inducible systems:
Application: Precisely control aaeX expression at different infection stages
Advantage: Allows temporal dissection of AaeX function
Implementation: Design guide RNAs targeting aaeX promoter regions with inducible dCas9
High-throughput mutagenesis and phenotyping:
Integrative multi-omics approaches:
Application: Correlate AaeX expression with global cellular responses
Advantage: Places AaeX within broader molecular networks
Implementation: Combine transcriptomics, proteomics, and metabolomics data from wild-type and aaeX mutant strains
These technologies would be particularly valuable given the importance of membrane proteins in bacterial adaptation to environmental stresses and host interactions, potentially revealing how AaeX contributes to Salmonella paratyphi B pathogenesis similar to the way virulence determinant variations affected Salmonella Paratyphi A clades .
Research on AaeX protein can provide valuable insights into paratyphoid fever epidemiology and transmission through several methodological approaches:
Geographically diverse strain analysis:
Compare aaeX gene sequences across clinical isolates from different geographical regions
Correlate sequence variations with regional epidemiological patterns
Analyze if aaeX variations contribute to the association of Salmonella Paratyphi B with specific regions like the Andean area of South America
Molecular typing implementation:
Environmental persistence studies:
Investigate if AaeX contributes to survival in water, food, or environmental surfaces
Compare persistence between wild-type and aaeX mutant strains under various conditions
Correlate findings with environmental transmission routes
Host specificity analysis:
Determine if AaeX variants correlate with adaptation to different hosts
Compare human isolates with potential animal or environmental reservoirs
Assess if variations in aaeX contribute to zoonotic transmission potential
Transmission model development:
Incorporate AaeX functional data into mathematical models of pathogen transmission
Predict how alterations in AaeX function might affect transmission dynamics
Validate models using epidemiological data from different geographical regions
One Health approach integration:
By connecting molecular insights about AaeX with epidemiological data, researchers can potentially identify factors contributing to the global distribution of paratyphoid fever, which has shown distinct geographical patterns such as Salmonella Paratyphi B association with South America and Salmonella Paratyphi A with South Asia .