Recombinant Enterobacter sp. Protein AaeX (aaeX)

Shipped with Ice Packs
In Stock

Description

Recombinant Proteins in Enterobacter spp.: Contextual Insights

Enterobacter species are known for their diverse enzymatic and structural proteins, often studied in biotechnological or pathogenic contexts. Below are key recombinant proteins from Enterobacter spp. documented in the literature, which may provide indirect insights into aaeX’s potential functions.

ProteinFunction/RoleSourceKey Findings
ENT638_2183Hypothetical protein (OPCA94095) Recombinant protein tested in Western blot (WB) and ELISA applications.
Alkanesulfonate monooxygenase (ssuD)Catalyzes sulfonate metabolism Recombinant variants expressed in E. coli, yeast, and mammalian systems.
Tae4Type VI secretion system effector Forms complexes with Tai4 to inhibit amidase activity; structural insights from cryo-EM.
Enterobacter sp. 638 genome-encoded proteinsPlant growth promotion, phytohormone synthesis Includes genes for acetoin/2,3-butanediol production linked to sucrose metabolism.

Critical Analysis of AaeX-Specific Data Gaps

Despite extensive documentation of Enterobacter-derived proteins, no peer-reviewed studies explicitly mention "aaeX". This absence suggests several possibilities:

  1. Nomenclature Confusion: AaeX may be a novel or misidentified protein. For example, Enterobacter spp. 638 encodes over 4,000 genes, many with uncharacterized functions .

  2. Limited Research Focus: AaeX might belong to a less-studied class of enzymes (e.g., sulfonatases, efflux pumps) or regulatory proteins.

  3. Commercial or Proprietary Data: Recombinant proteins like ENT638_2183 or ssuD are often developed for diagnostic or industrial applications, which may not be publicly disclosed.

Mechanistic Hypotheses for AaeX

While speculative, aaeX could plausibly belong to one of the following functional categories based on Enterobacter’s established biochemical pathways:

Hypothesized FunctionSupporting Evidence
Antibiotic ResistanceEnterobacter spp. are notorious for multidrug resistance via efflux pumps (e.g., AcrAB) , β-lactamases , or porin mutations . AaeX could modulate efflux systems or β-lactamase activity.
Metabolic RegulationEnterobacter sp. 638 produces plant growth-promoting compounds (e.g., acetoin) linked to sucrose metabolism . AaeX might regulate similar pathways.
Pathogenicity FactorsE. cloacae secretes cytotoxins (e.g., hemolytic/leukotoxic proteins) . AaeX could act as a virulence factor.

Methodological Recommendations for Future Research

To address the knowledge gap, the following steps are proposed:

  1. Bioinformatic Mining: Cross-reference "aaeX" with Enterobacter genomic databases (e.g., Enterobacter sp. 638 ) to identify homologs.

  2. Functional Assays:

    • Enzymatic Activity: Screen for sulfonatase, β-lactamase, or amidase activity using substrates (e.g., cephalosporins, sulfonates).

    • Protein Interactions: Co-immunoprecipitation (Co-IP) to identify binding partners (e.g., efflux pumps, regulatory proteins).

  3. Structural Studies: Cryo-EM or X-ray crystallography to determine aaeX’s tertiary structure, as demonstrated for Tae4-Tai4 complexes .

Product Specs

Form
Lyophilized powder
Note: We will prioritize shipping the format that we have in stock. However, if you have specific requirements for the format, please indicate them when placing the order. We will prepare the product according to your request.
Lead Time
Delivery time may vary depending on the purchasing method or location. For specific delivery timeframes, please consult your local distributors.
Note: All of our proteins are shipped with standard blue ice packs. If you require dry ice shipment, please contact us in advance, as additional fees will apply.
Notes
Repeated freeze-thaw cycles are not recommended. Store working aliquots at 4°C for up to one week.
Reconstitution
We recommend centrifuging the vial briefly prior to opening to ensure the contents settle at the bottom. Reconstitute the protein in deionized sterile water to a concentration of 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 may use this as a reference.
Shelf Life
The shelf life is influenced by various factors, including storage conditions, buffer ingredients, storage temperature, and the inherent stability of the protein.
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 the development of your specified tag.
Synonyms
aaeX; Ent638_3679; 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
Enterobacter sp. (strain 638)
Target Names
aaeX
Target Protein Sequence
MSLFPVFVVFGLSFPPIFFELILSLAIFWLVRKLLAPTGIYDFVWHPALFNTALYCCLFY LISRMFV
Uniprot No.

Target Background

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

Q&A

What is AaeX protein and what is its role in Enterobacter species?

AaeX is a membrane protein associated with p-hydroxybenzoic acid efflux and acid resistance mechanisms in Enterobacter species. The protein functions as part of a membrane transport system that helps the bacterium respond to environmental stresses. In pathogenic species like Enterobacter bugandensis, membrane proteins often play crucial roles in virulence and survival within host environments. Studies have shown that Enterobacter species can survive in high concentrations of human serum, indicating potential roles of membrane transport proteins in mediating resistance to host defense mechanisms .

How is the aaeX gene regulated in Enterobacter species?

The aaeX gene expression is typically regulated in response to environmental stresses, particularly acidic conditions and the presence of aromatic compounds. Whole genome-based transcriptome analysis of Enterobacter bugandensis revealed that approximately 7% of the genome is mobilized during growth in human serum, with significant upregulation of genes involved in iron uptake, storage, and metabolism . While specific regulatory mechanisms for aaeX were not directly addressed in the studies, related membrane transport systems are often controlled through complex regulatory networks involving stress response pathways. Research has shown that Enterobacter can rapidly adapt to environmental changes by modulating expression of membrane transport proteins.

What methods are recommended for identifying aaeX homologs across Enterobacter species?

For identifying aaeX homologs across different Enterobacter species, a multi-faceted approach is recommended:

  • Sequence-based analysis: Use BLAST and HMMER algorithms with the known aaeX sequences as queries against Enterobacter genome databases

  • Phylogenetic profiling: Construct phylogenetic trees based on aligned sequences to identify evolutionary relationships

  • Structural prediction: Analyze predicted transmembrane domains and protein topology using programs like TMHMM and TOPCONS

  • Genomic context analysis: Examine the genomic neighborhood of putative aaeX genes to identify conserved synteny

What expression systems yield the best results for recombinant AaeX protein?

Optimal expression of recombinant AaeX requires careful selection of expression systems, with several approaches showing success:

For membrane proteins like AaeX, E. coli C43(DE3) often provides the best balance between yield and proper folding. Codon optimization is essential when expressing Enterobacter proteins in heterologous systems, and fusion tags (such as MBP or SUMO) can significantly improve solubility. Expression protocols should include careful temperature control (typically 16-25°C after induction) to minimize inclusion body formation.

How can researchers optimize purification protocols for AaeX to maintain structural integrity?

Purification of membrane proteins like AaeX requires specialized approaches to maintain native structure:

  • Membrane extraction: Use mild detergents like n-dodecyl-β-D-maltoside (DDM) or lauryl maltose neopentyl glycol (LMNG) for initial solubilization

  • Affinity chromatography: His-tag purification under optimized detergent conditions

  • Size exclusion chromatography: Critical for removing aggregates and ensuring homogeneity

  • Detergent exchange: Consider switching to more stabilizing detergents during purification

  • Stability assessment: Monitor protein stability using techniques like thermostability assays

Maintaining the proper pH (typically 7.0-8.0) and including stabilizing agents like glycerol (10-15%) can significantly improve protein stability during purification. For functional studies, reconstitution into nanodiscs or liposomes may better preserve native activity. Similar approaches have been used successfully for membrane proteins from related pathogenic bacteria, including those that contribute to serum resistance and iron acquisition .

What are the optimal buffer conditions for maintaining AaeX stability during purification?

Based on studies of similar membrane transport proteins, the following buffer conditions are recommended for AaeX purification:

Buffer ComponentRecommended RangeRationale
pH7.2-7.6Mimics physiological conditions of bacterial periplasm
NaCl150-300 mMProvides ionic strength while preventing aggregation
Glycerol10-15%Acts as stabilizing agent
Detergent1-2× CMCMaintains micelle integrity without excess detergent
Reducing agent1-5 mM DTT or TCEPPrevents oxidation of cysteine residues
EDTA0.5-1 mMChelates metal ions that may promote degradation

For long-term storage, inclusion of protease inhibitors and maintaining temperatures of 4°C or below is essential. Buffer optimization should be empirically determined for each recombinant construct, as minor variations in the protein sequence can significantly impact stability profiles.

What methodologies are most effective for studying AaeX interactions with other bacterial proteins?

Multiple complementary approaches are recommended for investigating AaeX protein interactions:

  • Co-immunoprecipitation: Using antibodies against AaeX or epitope tags to pull down interaction partners

  • Bacterial two-hybrid assays: Modified for membrane proteins by using split-ubiquitin systems

  • Cross-linking coupled with mass spectrometry: To capture transient interactions

  • FRET/BRET analysis: For studying interactions in living bacterial cells

  • Proteomic analysis: Comparing wild-type and aaeX knockout strains to identify affected pathways

When analyzing interaction networks, it's important to consider that approximately 7% of the Enterobacter genome can be mobilized during adaptive responses, as observed in Enterobacter bugandensis during growth in serum . Focus on interactions with proteins involved in stress response pathways, particularly those related to iron acquisition and utilization, as these systems are critical for pathogenesis.

How can researchers assess the role of AaeX in acid resistance mechanisms?

To investigate AaeX's contribution to acid resistance, consider the following methodological approach:

  • Generate gene knockouts or knockdowns: Create aaeX deletion mutants using CRISPR-Cas9 or traditional homologous recombination

  • pH survival assays: Expose wild-type and mutant strains to varying pH conditions (typically pH 2.5-5.5) and measure survival rates

  • Gene expression analysis: Use qRT-PCR or RNA-seq to measure changes in gene expression during acid stress

  • Membrane potential measurements: Assess proton gradient maintenance using fluorescent probes

  • Complementation studies: Reintroduce wild-type and mutant aaeX to confirm phenotypes

  • In vivo infection models: Similar to the Galleria mellonella and mouse models used to study Enterobacter bugandensis virulence

For effective acid resistance studies, standardize experimental conditions including growth phase (typically late log phase), media composition, and exposure times. The ability to survive in acidic environments often correlates with virulence potential, as demonstrated in pathogenic Enterobacter strains.

What approaches can determine the membrane topology and structural characteristics of AaeX?

Multiple experimental and computational methods should be combined to elucidate AaeX structure:

For membrane proteins from pathogenic bacteria like Enterobacter bugandensis, relating structural features to functional roles in virulence is particularly valuable. Understanding the structural basis for interactions with antimicrobial compounds could inform development of novel inhibitors, similar to how the lasso peptide microcin J25 was found to inhibit Enterobacter bugandensis growth by targeting iron uptake systems .

How does AaeX contribute to Enterobacter virulence and pathogenesis mechanisms?

The contribution of AaeX to Enterobacter virulence likely involves multiple mechanisms:

  • Acid resistance: Enhancing survival during passage through the acidic environment of the stomach or within phagolysosomes

  • Efflux of toxic compounds: Removal of host-derived antimicrobial molecules

  • Maintenance of membrane integrity: Contributing to serum resistance, which is critical for pathogenesis

Studies on Enterobacter bugandensis demonstrated its high virulence in both Galleria mellonella and mouse models of infection, with the ability to induce systemic infection and proinflammatory cytokine release comparable to Salmonella Typhimurium . The bacterium showed remarkable serum resistance, which is often mediated by membrane proteins. Transcriptome analysis revealed extensive mobilization of genes related to iron uptake and metabolism during growth in serum, suggesting that membrane transport systems play crucial roles in adaptation to the host environment .

To investigate AaeX's specific contributions to virulence, researchers should compare wild-type and aaeX knockout strains in infection models, focusing on colonization efficiency, dissemination to organs, and induction of inflammatory responses.

What techniques can resolve contradictory data regarding AaeX function across different studies?

When faced with contradictory findings about AaeX function, implement the following systematic approach:

  • Standardize experimental conditions: Ensure all laboratories use identical strains, growth conditions, and assay parameters

  • Cross-laboratory validation: Exchange materials (strains, plasmids) between research groups for independent verification

  • Sequential deletion analysis: Create truncation variants to identify critical functional domains

  • Multi-omics approach: Combine transcriptomics, proteomics, and metabolomics to build comprehensive functional models

  • Environmental context consideration: Test function under varying conditions (pH, osmolarity, nutrient availability)

  • Meta-analysis: Systematically analyze all available data using statistical approaches like forest plots

Contradictions in functional data often arise from subtle differences in experimental conditions or strain backgrounds. When analyzing the role of AaeX in processes like iron acquisition, consider that Enterobacter species can upregulate multiple pathways simultaneously, as observed in transcriptome studies of Enterobacter bugandensis .

How can AaeX be targeted for potential therapeutic applications against pathogenic Enterobacter?

Therapeutic targeting of AaeX might follow several strategic approaches:

  • Small molecule inhibitors: Design molecules that block substrate binding sites or inhibit conformational changes

  • Peptide-based inhibitors: Develop peptides that interfere with protein-protein interactions, similar to how microcin J25 inhibits iron uptake in Enterobacter bugandensis

  • Antibody-based approaches: Generate antibodies against extracellular epitopes

  • Antisense technologies: Target aaeX mRNA to reduce protein expression

  • CRISPR-based antimicrobials: Develop sequence-specific nucleases targeting the aaeX gene

For therapeutic development, focus on features that distinguish bacterial proteins from human homologs. Studies on Enterobacter bugandensis demonstrated that the lasso peptide microcin J25, which inhibits iron uptake and RNA polymerase activity, effectively inhibited bacterial growth . Similar approaches targeting membrane transport systems might prove effective against multi-drug resistant Enterobacter strains.

What are common pitfalls in recombinant AaeX expression and how can they be overcome?

Researchers frequently encounter these challenges when working with recombinant AaeX:

ChallengePotential CausesSolutions
Low expression levelsToxicity to host cells, rare codonsUse specialized strains (C43), lower induction temperature, codon optimization
Inclusion body formationRapid expression, improper foldingReduce induction temperature to 16-18°C, use fusion tags (MBP, SUMO)
Proteolytic degradationInstability in expression hostAdd protease inhibitors, use protease-deficient strains
Poor solubilizationInadequate detergent selectionScreen multiple detergents (DDM, LMNG, GDN) for extraction efficiency
Loss of activityDenaturation during purificationInclude stabilizing agents, minimize purification steps
Aggregation during concentrationDetergent concentration issuesMaintain detergent above CMC, use centrifugal concentrators with appropriate MWCO

When expressing membrane proteins from pathogenic bacteria like Enterobacter, cellular toxicity can be a significant challenge. Similar issues were likely addressed when studying membrane proteins involved in iron acquisition in Enterobacter bugandensis, which showed upregulation during growth in human serum .

How should researchers approach experimental design when studying AaeX interactions with iron acquisition systems?

Given the importance of iron acquisition for Enterobacter pathogenesis , studying AaeX interactions with these systems requires careful experimental design:

  • Growth conditions: Compare iron-replete and iron-depleted conditions to identify differential interactions

  • Model system selection: Use both in vitro reconstituted systems and intact bacterial cells

  • Mutational analysis: Create point mutations in potential interaction interfaces

  • Competition assays: Use purified protein domains to competitively inhibit interactions

  • Comparative analysis: Study the same interactions in non-pathogenic Enterobacter strains as controls

Transcriptome analysis of Enterobacter bugandensis revealed that genes involved in iron uptake and storage were significantly upregulated during growth in serum, suggesting a critical role in pathogenesis . When designing experiments, include positive controls with known iron acquisition proteins and negative controls with unrelated membrane proteins to establish specificity of interactions.

What statistical approaches are most appropriate for analyzing variability in AaeX functional assays?

To address variability in AaeX functional data:

When analyzing functional data related to virulence or pathogenesis, consider using similar statistical approaches to those employed in studies of Enterobacter bugandensis infection models, which demonstrated significant differences in survival rates and cytokine production compared to control organisms .

How might single-cell techniques advance our understanding of AaeX expression heterogeneity?

Single-cell technologies offer promising approaches to understand AaeX expression dynamics:

  • Single-cell RNA-seq: Reveals expression heterogeneity across bacterial populations during infection or stress

  • Time-lapse fluorescence microscopy: With AaeX-fluorescent protein fusions to track expression and localization in real-time

  • Mass cytometry (CyTOF): Using metal-conjugated antibodies against AaeX to quantify protein levels at single-cell resolution

  • Microfluidics-based assays: To study expression dynamics under changing environmental conditions

  • Spatial transcriptomics: To examine aaeX expression in the context of host-pathogen interactions

What emerging technologies might improve structural characterization of AaeX and similar membrane proteins?

Several cutting-edge technologies are transforming membrane protein structural biology:

  • CryoEM advances: New direct electron detectors and processing algorithms allow structure determination at near-atomic resolution

  • Integrative structural biology: Combining multiple data sources (SAXS, NMR, crosslinking-MS) for comprehensive structural models

  • AlphaFold2 and RoseTTAFold: AI-based structure prediction showing remarkable accuracy even for membrane proteins

  • Serial femtosecond crystallography: Using X-ray free electron lasers for structure determination from microcrystals

  • Native mass spectrometry: For analyzing membrane protein complexes in near-native conditions

  • Hydrogen-deuterium exchange with mass spectrometry: For probing dynamics and conformational changes

Structural insights into AaeX could reveal mechanisms similar to those involved in the serum resistance of Enterobacter bugandensis, potentially identifying targets for therapeutic intervention . Comparing structures across different functional states would be particularly valuable for understanding transport mechanisms.

How might systems biology approaches integrate AaeX function into broader pathogenicity networks?

Systems biology offers powerful frameworks to contextualize AaeX within cellular networks:

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.