KEGG: efa:EF1184
STRING: 226185.EF1184
Dihydrodipicolinate synthase (DapA) catalyzes the first committed step in the lysine biosynthesis pathway of bacteria. In E. faecalis, as in other bacteria, this enzyme is essential due to the requirement for lysine or meso-diaminopimelate in the cross-linking of cell-wall peptidoglycan . The reaction involves Schiff-base formation between pyruvate and the ε-nitrogen of lysine, followed by nucleophilic attack by the enamine form of the Schiff base on aspartate β-semialdehyde, ultimately leading to the production of 4-hydroxy-2,3,4,5-tetrahydroxydipicolinate . This pathway is critical for bacterial cell wall integrity, making DapA an essential enzyme for E. faecalis survival.
E. faecalis DapA represents a promising antibiotic target for several reasons:
It catalyzes an essential step in the lysine biosynthetic pathway, which is crucial for bacterial cell wall formation
This pathway is absent in humans, allowing for selective targeting
E. faecalis is increasingly problematic due to its propensity for intrinsic and acquired multiple drug resistance (MDR)
Targeting novel pathways helps address resistance to traditional antibiotics like ampicillin and vancomycin, to which E. faecalis has developed varying levels of resistance
The ability to inhibit an essential bacterial pathway not present in humans makes DapA an attractive target for developing novel antimicrobials against resistant E. faecalis strains.
While the search results don't provide comprehensive structural data specific to E. faecalis DapA, comparative analysis with other bacterial DapA enzymes reveals important structural features. Most bacterial DapA enzymes share a conserved (β/α)8-barrel fold with an active site containing key catalytic residues . A notable structural feature involves regions that undergo conformational shifts upon substrate binding.
Of particular interest is the variation in key residues between species. For example, E. coli DapA contains Arg138, which is proposed to bind to the carboxyl group of aspartate β-semialdehyde, while some other bacterial species naturally have histidine in this position, potentially resulting in reduced catalytic efficiency . Structural analysis of various bacterial DapA enzymes has helped identify determinants that define lysine-mediated allosteric inhibition, with E. faecalis DapA being among the enzymes studied for prediction of lysine sensitivity .
Based on published protocols for similar bacterial DapA enzymes, the following methodology is recommended:
Cloning and Expression Protocol:
PCR amplification of the dapA gene from E. faecalis genomic DNA
Cloning into an expression vector such as pET11a, pET28a, or pRSET A
Transformation into E. coli BL21-DE3 expression host
Induction with 1 mM isopropyl β-D-1-thiogalactopyranoside (IPTG) in Luria broth
Culture growth at either 37°C (standard) or 16°C (for potentially improved folding)
Cell harvesting and lysis using appropriate buffer systems
This protocol is based on successful expression strategies for related bacterial DHDPS enzymes and should yield functional recombinant E. faecalis DapA with high purity (>98% homogeneity) .
E. faecalis presents significant challenges for genetic manipulation that researchers must address:
Physical Barriers:
Enzymatic Barriers:
Type I, II, and IV restriction modification systems that cleave foreign DNA
CRISPR-Cas systems that target and degrade foreign genetic elements
To overcome these barriers when studying DapA, researchers should consider:
Using DNA from the same strain to avoid restriction
Employing DNA methylation to protect against restriction enzymes
Optimizing electroporation conditions specifically for E. faecalis
Using specialized vectors designed to bypass restriction systems
Considering alternative approaches when direct manipulation of native E. faecalis is challenging, such as expressing the gene in heterologous systems
For accurate kinetic characterization of E. faecalis DapA:
Recommended Kinetic Assay Approach:
Spectrophotometric coupled assay: Monitor NADPH oxidation at 340 nm when coupling the DapA reaction with dihydrodipicolinate reductase
Direct detection assay: Measure the formation of dihydrodipicolinate at 270 nm
Critical Parameters to Control:
Enzyme concentration (typically 1-10 nM final concentration)
Substrate concentrations (varied systematically for Km determination)
Buffer composition and pH (typically pH 7.5-8.5)
Temperature (commonly 25°C or 37°C)
Presence of potential inhibitors (e.g., lysine at various concentrations)
Data Analysis:
Fit initial velocity data to Michaelis-Menten equation
For potential allosteric effects, analyze data using Hill equation or other appropriate models for cooperative binding
This methodological approach ensures reliable kinetic data that can be compared with other bacterial DapA enzymes to understand evolutionary and structural relationships.
Allosteric regulation is a critical consideration for understanding E. faecalis DapA function and its targetability:
Allosteric Regulation Characteristics:
Lysine acts as an allosteric inhibitor for many bacterial DHDPS enzymes
E. faecalis DHDPS has been studied to predict its sensitivity to lysine inhibition based on identified structural determinants
The presence or absence of lysine-mediated inhibition can be predicted reliably based on specific structural features
Implications for Drug Development:
Allosteric sites may offer alternative targeting strategies beyond active site inhibition
Understanding species-specific differences in allosteric regulation may allow for selective inhibition
Enzymes with different allosteric profiles may require different drug design approaches
These considerations are vital for rational drug design targeting E. faecalis DapA, as allosteric mechanisms could be exploited to develop species-specific inhibitors with reduced likelihood of resistance development.
Site-directed mutagenesis provides valuable insights into structure-function relationships:
Key Mutational Findings:
The EfDHDPS-E56K mutant has been synthesized and studied , suggesting the importance of this residue
Mutations of key catalytic or allosteric site residues can drastically affect enzyme function
Comparative analysis with other bacterial DapA enzymes reveals that mutations such as arginine-to-histidine substitutions (as observed in E. coli DapA) can result in drastically reduced catalytic efficiency
Research Applications:
Identification of residues essential for catalysis
Understanding substrate specificity determinants
Mapping allosteric regulation pathways
Informing rational drug design by highlighting critical residues for inhibitor interaction
Site-directed mutagenesis studies thus serve as a foundation for understanding the molecular basis of E. faecalis DapA function and developing targeted inhibitors.
Selective inhibitor design relies on exploiting structural differences:
Key Structural Considerations:
The lysine biosynthesis pathway is absent in humans, making DapA inherently selective as a target
Substrate binding pockets and catalytic residues unique to bacterial DapA can be exploited
Species-specific variations in allosteric sites provide opportunities for selective targeting
Design Strategy Framework:
Identify unique structural features of E. faecalis DapA through crystallography or homology modeling
Focus on regions with low sequence/structural conservation with human proteins
Design compounds that interact with E. faecalis-specific residues
Incorporate features that enhance penetration through the bacterial cell wall
Consider the impact of potential resistance mutations based on structural analysis
This approach leverages the evolutionary distance between bacterial and human enzymes to design inhibitors with minimal off-target effects on human proteins.
Proper interpretation of kinetic data is essential for understanding inhibition mechanisms:
Types of Inhibition Patterns and Their Interpretation:
| Inhibition Pattern | Lineweaver-Burk Plot | Km Effect | Vmax Effect | Mechanistic Implication |
|---|---|---|---|---|
| Competitive | Lines intersect on y-axis | Increases | No change | Inhibitor binds active site |
| Non-competitive | Lines intersect on x-axis | No change | Decreases | Inhibitor binds enzyme-substrate complex |
| Uncompetitive | Parallel lines | Decreases | Decreases | Inhibitor binds only to enzyme-substrate complex |
| Mixed | Lines intersect in quadrant | Changes | Decreases | Inhibitor binds both free enzyme and enzyme-substrate complex |
| Allosteric | Non-linear effects | Variable | Variable | Inhibitor binds at site distinct from active site |
Key Analysis Considerations:
Use global fitting approaches rather than transformed plots when possible
Consider enzyme oligomeric state when interpreting allosteric effects
Examine temperature and pH dependence to understand thermodynamic and ionization effects
Compare inhibition patterns with structural data to build comprehensive inhibition models
This framework enables researchers to accurately characterize inhibition mechanisms and develop more effective inhibitors against E. faecalis DapA.
Systematic structure-activity relationship (SAR) analysis requires:
Recommended SAR Analysis Methodology:
Scaffold Identification:
Screen diverse compound libraries against purified E. faecalis DapA
Identify chemical scaffolds with inhibitory activity
Systematic Modification:
Synthesize analogs with variations at key positions
Test each analog for inhibitory potency (IC50, Ki)
Data Analysis:
Create SAR tables correlating structural features with activity
Develop quantitative structure-activity relationship (QSAR) models
Use computational approaches (docking, molecular dynamics) to predict binding modes
Biological Validation:
Test most promising compounds against E. faecalis cultures
Assess specificity by testing against human cell lines
Evaluate pharmacokinetic properties for lead compounds
This methodical approach enables researchers to optimize inhibitors against E. faecalis DapA while maintaining selectivity and favorable drug-like properties.
Researchers commonly encounter several challenges when working with recombinant E. faecalis DapA:
Expression Challenges:
Low solubility: Recombinant DapA may form inclusion bodies
Low expression levels:
Solution: Optimize codon usage, test different promoters, adjust media composition
Enzymatic barriers in native expression:
Purification Challenges:
Protein instability:
Solution: Include protease inhibitors, maintain cold temperatures, minimize freeze-thaw cycles
Loss of activity:
Solution: Ensure proper buffer composition, include stabilizing agents, verify cofactor requirements
Oligomerization issues:
Solution: Use size exclusion chromatography to isolate properly formed tetramers (if E. faecalis DapA forms tetramers like other bacterial DHDPS enzymes)
These troubleshooting approaches address the most common technical challenges researchers face when working with recombinant E. faecalis DapA.
Inconsistent inhibition results may stem from several sources:
Common Sources of Variability and Solutions:
Enzyme heterogeneity:
Assay conditions:
Standardize buffer composition, pH, and temperature
Use consistent substrate preparation methods
Control for potential interfering compounds
Inhibitor properties:
Check inhibitor solubility in assay buffer
Verify inhibitor stability under assay conditions
Consider potential aggregation of inhibitor compounds
Statistical considerations:
Perform experiments in triplicate at minimum
Use appropriate controls in each experiment
Apply robust statistical analysis to determine significance
By systematically addressing these potential sources of variability, researchers can obtain more consistent and reliable inhibition data for E. faecalis DapA.
Several innovative approaches show promise for advancing E. faecalis DapA research:
Emerging Methodologies:
CRISPR-based approaches: Despite barriers to genetic manipulation in E. faecalis , adapted CRISPR systems may enable more precise genetic studies
Fragment-based drug discovery: Screening small molecular fragments against E. faecalis DapA may identify novel chemical scaffolds
Cryo-EM analysis: High-resolution structural studies may reveal dynamic aspects of E. faecalis DapA function not visible in crystal structures
Systems biology approaches: Understanding the network effects of DapA inhibition may identify synergistic drug combinations
Targeted drug delivery: Developing E. faecalis-specific delivery systems may enhance antibiotic efficacy while reducing off-target effects
These approaches represent the cutting edge of research in this field and may accelerate the development of novel therapeutics targeting E. faecalis DapA.
Comparative analysis between Enterococcus species provides valuable insights:
Key Comparative Considerations:
E. faecium infections are typically more difficult to treat than E. faecalis due to higher levels of ampicillin and vancomycin resistance
Analysis of clinical isolates found that 13.4% of E. faecium strains compared to only 1.4% of E. faecalis strains were resistant to linezolid
Understanding species-specific differences in DapA structure and function may reveal:
Differences in inhibitor susceptibility
Varying potential for resistance development
Species-specific regulatory mechanisms
Research Strategy Implications:
Develop broad-spectrum inhibitors targeting conserved features in both species
Design species-selective inhibitors when warranted by resistance profiles
Create inhibitor combinations that address species-specific variations
Leverage comparative genomics to predict potential resistance mechanisms
This comparative approach ensures that DapA-targeted antibiotics can address the full spectrum of Enterococcus infections, including highly resistant E. faecium strains.