Recombinant Salmonella paratyphi B Protein AaeX (aaeX)

Shipped with Ice Packs
In Stock

Description

Definition and Basic Properties

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 .

PropertyDetails
Source OrganismSalmonella paratyphi B (strain ATCC BAA-1250/SPB7)
Expression SystemE. coli
Molecular Weight~8.3 kDa (calculated)
Purity>90% (SDS-PAGE verified)
Storage-20°C/-80°C in Tris/PBS buffer with 6% trehalose; avoid freeze-thaw cycles

Amino Acid Sequence

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 .

Vaccine Development

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.

Diagnostic Tools

Metabolomic profiling of Salmonella infections has identified serovar-specific biomarkers . AaeX could contribute to developing targeted assays for distinguishing Paratyphi B from other serovars.

Research Findings and Gaps

  • 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 .

Future Directions

  • 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.

Product Specs

Form
Lyophilized powder
Note: We prioritize shipping the format currently in stock. However, if you have specific format requirements, please indicate them during order placement, and we will fulfill your request.
Lead Time
Delivery time may vary depending on the purchase method and location. Please contact your local distributors for specific delivery timelines.
Note: All our proteins are shipped with standard blue ice packs by default. If you require dry ice shipping, please inform us in advance, and additional fees will apply.
Notes
Repeated freezing and thawing is not recommended. Store working aliquots at 4°C for up to one week.
Reconstitution
We recommend briefly centrifuging the vial before opening to ensure the contents settle at the bottom. Reconstitute the protein in deionized sterile water to a concentration between 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our default final glycerol concentration is 50%. Customers can use this as a reference.
Shelf Life
Shelf life is influenced by various factors including storage conditions, buffer ingredients, storage temperature, and the protein's inherent stability.
Generally, the shelf life of liquid form is 6 months at -20°C/-80°C. The shelf life of lyophilized form is 12 months at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is necessary for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type will be determined during the manufacturing process.
The tag type will be determined during the production process. If you have a specific tag type in mind, please inform us, and we will prioritize developing the specified tag.
Synonyms
aaeX; SPAB_04194; Protein AaeX
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-67
Protein Length
full length protein
Species
Salmonella paratyphi B (strain ATCC BAA-1250 / SPB7)
Target Names
aaeX
Target Protein Sequence
MSLFPVIVVFGLSFPPIFFELLLSLAIFWLVRRMLVPTGIYDFVWHPALFNTALYCCLFY LISRLFV
Uniprot No.

Target Background

Database Links
Protein Families
AaeX family
Subcellular Location
Cell membrane; Multi-pass membrane protein.

Q&A

What is the AaeX protein and what is its significance in Salmonella paratyphi B?

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 .

How is recombinant AaeX protein typically produced for research applications?

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 .

What are the standard storage and handling protocols for recombinant AaeX protein?

For optimal stability and activity of recombinant AaeX protein, the following handling protocols are recommended:

Storage ParameterRecommended Protocol
Storage temperature-20°C to -80°C
Storage bufferTris/PBS-based buffer, 6% Trehalose, pH 8.0
ReconstitutionDeionized sterile water to 0.1-1.0 mg/mL
Long-term preservationAdd glycerol to 5-50% final concentration
Working aliquotsStore at 4°C for up to one week
Freeze-thaw cyclesMinimize; repeated cycles not recommended
Pre-use preparationBrief 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.

How can AaeX protein be used in immunogenicity studies, and what are the key experimental considerations?

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.

What role might AaeX play in antimicrobial resistance of Salmonella paratyphi B and how can this be investigated?

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 .

How might AaeX interact with virulence factors in Salmonella pathogenicity islands, and what experimental approaches can elucidate these relationships?

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 .

What controls and validation steps are essential when working with recombinant AaeX protein in functional studies?

When designing experiments using recombinant AaeX protein, the following controls and validation steps are essential to ensure reliable and reproducible results:

Control/Validation TypeImplementation MethodPurpose
Protein quality controlSDS-PAGE analysisConfirm protein integrity and purity (>90%)
Western blot with anti-His antibodyVerify identity of the recombinant protein
Mass spectrometryConfirm accurate amino acid sequence
Negative controlsEmpty vector-expressed proteinControl for E. coli host protein contamination
Denatured AaeX proteinControl for non-specific protein effects
Buffer-only conditionsControl for buffer component effects
Positive controlsKnown membrane protein of similar sizeBenchmark for expected behavior
Native AaeX (if available)Compare recombinant vs. native function
Activity validationSecondary structure analysisConfirm proper protein folding using CD spectroscopy
Membrane integration assayVerify protein insertion into lipid bilayers
Functional complementationTest if recombinant protein rescues aaeX knockout phenotypes
Dose-response assessmentTitration experimentsEstablish optimal concentration ranges
Technical replicatesMinimum 3 independent preparationsAccount for batch-to-batch variation
Biological replicatesMultiple experimental runsEnsure reproducibility of observations

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.

How should researchers design experiments to investigate potential roles of AaeX in host-pathogen interactions?

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 .

What are the key challenges in analyzing protein-protein interactions involving AaeX, and how can these be addressed?

Investigating protein-protein interactions involving membrane proteins like AaeX presents several methodological challenges that require specific analytical approaches:

  • Membrane protein solubility issues:

    • Challenge: AaeX's hydrophobic nature (evident in its amino acid sequence) complicates traditional interaction assays

    • Solution: Use mild detergents optimized for membrane proteins (e.g., DDM, LMNG)

    • Validation: Test multiple detergent conditions to ensure interaction preservation

  • 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 .

How can researchers reconcile potential discrepancies between in vitro and in vivo findings when studying AaeX function?

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.

What emerging technologies could advance our understanding of AaeX structure-function relationships?

Several cutting-edge technologies offer promising approaches to elucidate AaeX structure-function relationships:

  • Cryo-electron microscopy (Cryo-EM):

    • Application: Determine high-resolution structure of AaeX in membrane environments

    • Advantage: Preserves native protein conformation without crystallization

    • Implementation: Reconstitute purified His-tagged AaeX in nanodiscs for single-particle analysis

  • 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:

    • Application: Systematic analysis of amino acid contributions to AaeX function

    • Advantage: Identifies essential residues and domains

    • Implementation: Create a saturating mutagenesis library of the 67-amino acid sequence followed by functional screening

  • 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 .

How can research on AaeX contribute to our understanding of paratyphoid fever epidemiology and transmission?

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:

    • Incorporate aaeX sequence analysis into molecular epidemiology tools

    • Develop aaeX-based markers for strain tracking in outbreak investigations

    • Combine with existing typing methods like PFGE for enhanced discrimination, similar to how molecular subtyping helped identify travel-associated strains

  • 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:

    • Connect AaeX research with environmental, animal, and human health data

    • Analyze if AaeX variants correlate with antimicrobial resistance patterns observed in different regions

    • Develop integrated surveillance approaches that include molecular characterization of aaeX

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 .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.