Recombinant Erwinia carotovora subsp. atroseptica Protein AaeX (aaeX) is a bioengineered protein derived from the bacterium Erwinia carotovora subsp. atroseptica (also known as Pectobacterium atrosepticum). This pathogen is a Gram-negative enterobacterium responsible for soft-rot diseases in plants. While aaeX is not extensively studied in the literature, commercial recombinant versions are available for research purposes, primarily expressed in Escherichia coli with N-terminal His-tags for purification .
Attribute | Details |
---|---|
Gene Name | aaeX |
Locus | ECA0277 (based on E. carotovora subsp. atroseptica genome annotation) |
Uniprot ID | Q6DAH6 |
Protein Length | Full-length (1–67 amino acids) |
Sequence | MSSLPVMVLFGLSFPPVFFVLMVSLTLFFVVNRLLQPTGIYDFVWHPALFNSALFCCLFYLLFRYGL |
Expression Host | E. coli |
Tag | N-terminal His-tag |
The protein is annotated as a "hypothetical" or "putative" protein in genomic databases, with no defined functional role reported in peer-reviewed studies .
AaeX shares structural similarity with proteins involved in membrane transport or enzymatic processes in other bacterial species, but direct evidence is lacking .
While no experimental data on AaeX’s biological role exists in the provided sources, its genomic context and sequence features suggest potential involvement in:
Membrane-Associated Processes: The presence of hydrophobic regions (e.g., leucine-rich motifs) may indicate membrane localization .
Regulatory Networks: Homology to proteins in quorum-sensing or virulence-related pathways (e.g., ExpR, RsmA) in Erwinia species hints at a regulatory role .
The absence of peer-reviewed studies on AaeX limits its characterization. Key gaps include:
Functional Studies: No assays or knockout models are described to validate its activity.
Interactions: Binding partners, substrates, or regulatory targets remain undefined.
Expression Context: Conditions (e.g., environmental cues) triggering aaeX expression in Erwinia are unknown.
To advance understanding of AaeX, future studies should:
Conduct Biochemical Assays: Test enzymatic activity (e.g., hydrolase, kinase) or ligand-binding capacity.
Perform Comparative Genomics: Identify orthologs in other Erwinia species to infer conserved functions.
Generate Mutants: Use CRISPR-Cas9 to disrupt aaeX in Erwinia and assess phenotypic changes (e.g., virulence, enzyme production).
KEGG: eca:ECA0277
STRING: 218491.ECA0277
Erwinia carotovora subsp. atroseptica (now reclassified as Pectobacterium atrosepticum) is an enterobacterial phytopathogen that causes soft rot and blackleg diseases in potatoes. The significance of studying its proteins lies in understanding plant pathogenicity mechanisms within the Enterobacteriaceae family. According to genome sequencing studies, approximately 33% of its genes are not shared with sequenced enterobacterial human pathogens, suggesting unique metabolic traits such as nitrogen fixation and opine catabolism . Proteins from this organism provide insights into novel pathogenicity determinants and bacterial adaptation to plant hosts, making them valuable targets for agricultural research aimed at disease control and prevention.
Recombinant proteins like the ATP synthase subunit c (atpE) from E. carotovora subsp. atroseptica are typically expressed with fusion tags (such as His-tags) that facilitate purification while maintaining functional properties. The recombinant atpE protein (79 amino acids in length) retains its primary sequence integrity as evidenced by the amino acid sequence: MENLSVDLLYMAAALMMGLAAIGAAIGIGILGGKFLEGAARQPDLIPLLRTQFFIVMGLVDAIPMIAVGLGLYVMFAVA . When expressing such proteins, researchers should consider that N-terminal tags may affect protein folding or activity in some cases. For structural studies, it's advisable to compare both tagged and untagged versions of the protein to ensure the tag doesn't interfere with native conformation or function.
E. coli expression systems are widely used for the recombinant production of E. carotovora proteins due to their genetic compatibility as fellow members of the Enterobacteriaceae family. Commercial recombinant proteins such as the ATP synthase subunit c (atpE) are successfully expressed in E. coli with high purity (>90% as determined by SDS-PAGE) . For optimal expression, researchers should consider codon optimization based on the expression host and use vectors with appropriate promoters. While E. coli is the most common host, alternative expression systems may be required for proteins that form inclusion bodies or require post-translational modifications. The choice of expression system should be guided by the specific properties of the target protein and the intended downstream applications.
For efficient transformation of E. carotovora subsp. atroseptica, researchers should employ a modified version of the Hanahan method. This approach yields transformation frequencies ranging from 1 × 10² to 4 × 10⁴ colonies per microgram of plasmid DNA, as demonstrated with plasmids pBR322, pBR325, and pAT153 . The methodology involves preparing competent cells in the presence of divalent cations, followed by a heat shock procedure.
For optimal results, researchers should:
Use fresh overnight cultures diluted 1:100 and grown to early log phase (OD₆₀₀ 0.4-0.6)
Harvest cells by gentle centrifugation and maintain at 4°C throughout the procedure
Resuspend cells in ice-cold transformation buffer containing Ca²⁺ and Mg²⁺ ions
Introduce plasmid DNA (50-500 ng) to the competent cells
Apply heat shock at 42°C for 90 seconds followed by immediate cooling
Allow for expression of antibiotic resistance genes before plating on selective media
Confirmation of successful transformants should be performed through plasmid isolation and restriction analysis to verify the integrity of the introduced construct .
Optimizing the expression and purification of membrane proteins such as ATP synthase subunits requires specialized approaches due to their hydrophobic nature. For the ATP synthase subunit c (atpE), which contains multiple transmembrane domains, researchers should consider the following protocol:
Expression optimization:
Test multiple expression strains (BL21(DE3), C41(DE3), C43(DE3)) specifically designed for membrane proteins
Employ lower induction temperatures (16-25°C) to slow protein synthesis and improve folding
Use lower IPTG concentrations (0.1-0.5 mM) for induction
Include membrane-stabilizing agents such as glycerol (5-10%) in growth media
Purification strategy:
For highest purity (>90%), researchers should implement a multi-step purification process and analyze results using SDS-PAGE. The reconstitution of lyophilized protein should be performed in deionized sterile water to a concentration of 0.1-1.0 mg/mL, with addition of 5-50% glycerol for long-term storage .
When characterizing structural features of E. carotovora proteins, researchers should employ a multi-technique approach:
Primary structure analysis:
Mass spectrometry (MS) for molecular weight confirmation and post-translational modifications
N-terminal sequencing to verify protein identity and signal peptide cleavage
Secondary structure determination:
Circular dichroism (CD) spectroscopy to quantify α-helix, β-sheet, and random coil content
FTIR spectroscopy as a complementary technique for secondary structure analysis
Tertiary structure characterization:
X-ray crystallography for high-resolution structural information (resolution <2.5Å)
NMR spectroscopy for solution structure and dynamics (for proteins <30 kDa)
Cryo-EM for larger protein complexes or membrane proteins
Quaternary structure assessment:
Size exclusion chromatography combined with multi-angle light scattering (SEC-MALS)
Analytical ultracentrifugation to determine oligomerization state
For membrane proteins like the ATP synthase subunit c, which has a distinctive amino acid sequence with hydrophobic regions (MENLSVDLLYMAAALMMGLAAIGAAIGIGILGGKFLEGAARQPDLIPLLRTQFFIVMGLVDAIPMIAVGLGLYVMFAVA), structural predictions using computational tools can provide preliminary insights before experimental verification .
When designing experiments to study E. carotovora protein function in plant-pathogen interactions, researchers should implement a comprehensive multi-level approach:
In vitro functional assays:
Enzymatic activity assays for relevant functions (e.g., pectate lyase, cellulase activity)
Protein-protein interaction studies using pull-down assays, yeast two-hybrid, or surface plasmon resonance
Substrate specificity determination through enzyme kinetics
Cell-based assays:
Plant-pathogen interaction studies:
Plant bioassays with wild-type and mutant bacterial strains
Microscopy to visualize infection process
Quantification of virulence factors in planta
Transcriptome analysis during infection
Controls and validation:
Include positive and negative controls in all assays
Validate protein expression using Western blotting or mass spectrometry
Perform statistical analysis of replicated experiments
This experimental framework has proven effective in investigating novel pathogenicity determinants in E. carotovora subsp. atroseptica, as demonstrated by studies using insertional mutants that showed reduced virulence in plant bioassays .
For optimal stability of recombinant E. carotovora proteins, researchers should follow these evidence-based storage and handling procedures:
Short-term storage (up to one week):
Long-term storage:
Reconstitution protocol:
Stability assessment:
Monitor protein integrity using SDS-PAGE before experiments
Check activity using functional assays periodically
Document storage duration and conditions for reproducibility
These protocols are based on established guidelines for recombinant proteins from E. carotovora, such as the ATP synthase subunit c, which maintains >90% purity when properly stored and handled .
To accurately measure and interpret the effects of protein modifications on E. carotovora protein function, researchers should implement the following methodological approach:
Systematic protein modification strategy:
Site-directed mutagenesis of conserved or putative functional residues
Truncation analysis to identify functional domains
Fusion protein construction to assess the impact of tags
Post-translational modification analysis using mass spectrometry
Comparative functional assessment:
Protein Variant | Activity Assay | Binding Affinity | Structural Stability | In Vivo Function |
---|---|---|---|---|
Wild-type | Baseline | Baseline | Baseline | Baseline |
Modified | % of wild-type | Kd value | Tm value | % of wild-type |
Structural impact analysis:
Circular dichroism to assess secondary structure changes
Thermal shift assays to determine protein stability
Molecular dynamics simulations to predict conformational changes
Biological significance verification:
Complementation studies in bacterial mutants
Plant infection assays with modified protein variants
Expression analysis in native conditions vs. experimental conditions
This approach has been successfully applied to regulatory proteins in E. carotovora, such as the activator of extracellular protein production (aepA) and aepH, which control the production of pectolytic enzymes, cellulase, and protease that are critical for virulence .
Researchers working with E. carotovora proteins frequently encounter several challenges that can be addressed with specific strategies:
Low expression levels:
Optimize codon usage for the expression host
Test different promoter systems (T7, tac, araBAD)
Vary induction conditions (temperature, inducer concentration, induction time)
Screen multiple E. coli strains (BL21, Rosetta, Arctic Express)
Inclusion body formation:
Protein instability:
Purification interference:
These strategies have been successfully applied to the expression and purification of E. carotovora proteins, resulting in high-purity preparations (>90% as determined by SDS-PAGE) suitable for functional and structural studies .
When troubleshooting transformation efficiency issues with E. carotovora strains, researchers should implement a systematic approach:
Competent cell preparation optimization:
Use early log phase cultures (OD₆₀₀ 0.4-0.6)
Ensure all solutions and equipment are pre-chilled
Minimize exposure to room temperature
Test different buffer compositions with varying concentrations of divalent cations
DNA quality and quantity assessment:
Use highly purified plasmid DNA (A₂₆₀/A₂₈₀ ratio >1.8)
Optimize DNA concentration (50-500 ng per transformation)
Verify plasmid integrity by gel electrophoresis
Consider plasmid size (smaller plasmids generally transform more efficiently)
Transformation parameter optimization:
Parameter | Standard Condition | Optimization Range | Effect on Efficiency |
---|---|---|---|
Heat shock duration | 90 seconds | 30-120 seconds | Strain-dependent |
Heat shock temperature | 42°C | 37-45°C | Critical for membrane permeability |
Recovery medium | SOC | LB, 2XYT, SOB | Nutrient availability affects recovery |
Recovery time | 1 hour | 0.5-3 hours | Extended time for slow-growing strains |
Strain-specific considerations:
This systematic approach has been successful in achieving transformation frequencies ranging from 1 × 10² to 4 × 10⁴ colonies per microgram of plasmid DNA with E. carotovora subsp. carotovora and E. carotovora subsp. atroseptica .
Determining protein-protein interactions involving E. carotovora virulence factors presents unique challenges that can be addressed through complementary methodologies:
In vitro interaction studies:
Pull-down assays using recombinant His-tagged proteins
Surface plasmon resonance (SPR) for quantitative binding parameters
Isothermal titration calorimetry (ITC) for thermodynamic characterization
Microscale thermophoresis (MST) for interactions in complex solutions
Cell-based interaction methods:
Bacterial two-hybrid systems adapted for E. carotovora proteins
Förster resonance energy transfer (FRET) with fluorescent protein fusions
Bimolecular fluorescence complementation (BiFC) for visualizing interactions in situ
Co-immunoprecipitation from bacterial lysates with specific antibodies
Proximity-based interaction mapping:
Crosslinking mass spectrometry (XL-MS) to capture transient interactions
BioID or APEX2 proximity labeling to identify interaction neighborhoods
Hydrogen-deuterium exchange mass spectrometry (HDX-MS) to map interaction interfaces
Validation strategies:
Mutagenesis of predicted interaction sites
Competitive binding assays with peptide mimics
Functional assays measuring virulence factor activity
In planta confirmation of interactions during infection
These approaches are particularly valuable for studying interactions between virulence regulators like aepA and aepH, which control the production of extracellular enzymes that are essential for E. carotovora pathogenicity .
Several promising research avenues exist for utilizing E. carotovora proteins to enhance our understanding of bacterial pathogenicity mechanisms:
Comparative genomics and proteomics:
Secretion system characterization:
Regulatory network analysis:
Host-pathogen protein interactions:
Identifying plant proteins targeted by E. carotovora virulence factors
Characterizing molecular mechanisms of plant immunity suppression
Developing protein-based strategies for disease resistance
These research directions leverage the significant genomic and molecular tools available for E. carotovora subsp. atroseptica, including the complete genome sequence of strain SCRI1043 and established transformation protocols . The insights gained could lead to novel approaches for controlling bacterial plant diseases and provide broader understanding of pathogenicity mechanisms in the Enterobacteriaceae family.
Emerging technologies are poised to revolutionize structural and functional studies of E. carotovora proteins:
Advanced structural biology approaches:
Cryo-electron microscopy for near-atomic resolution of membrane proteins without crystallization
Integrative structural biology combining multiple data sources (SAXS, XL-MS, cryo-EM)
Microcrystal electron diffraction (MicroED) for proteins resistant to traditional crystallization
AlphaFold2 and related AI methods for accurate structure prediction
High-throughput functional genomics:
CRISPR-Cas9 genome engineering for rapid mutant generation
Transposon sequencing (Tn-Seq) to identify essential genes under infection conditions
RNA-Seq to map transcriptional responses to environmental stimuli
Ribosome profiling to monitor translation efficiency of virulence factors
Advanced imaging technologies:
Super-resolution microscopy to visualize protein localization during infection
Correlative light and electron microscopy (CLEM) for structural context
Label-free imaging techniques to study native protein distributions
Multiplexed imaging to track multiple proteins simultaneously
Systems biology integration:
Multi-omics data integration (genomics, transcriptomics, proteomics, metabolomics)
Network analysis to identify critical nodes in virulence pathways
Machine learning approaches to predict protein function from sequence and structure
Mathematical modeling of host-pathogen interactions
These technologies will provide unprecedented insights into the function of proteins like ATP synthase subunits and regulatory proteins such as aepA and aepH , advancing our understanding of E. carotovora pathogenicity mechanisms and potentially leading to novel disease control strategies.