Function: Catalyzes the condensation of (S)-aspartate-β-semialdehyde [(S)-ASA] and pyruvate to 4-hydroxy-tetrahydrodipicolinate (HTPA).
KEGG: bba:Bd0046
STRING: 264462.Bd0046
Dihydrodipicolinate synthase (dapA) catalyzes the first unique step in the lysine biosynthesis pathway in Bdellovibrio bacteriovorus, condensing pyruvate and L-aspartate semialdehyde to form 4-hydroxy-2,3,4,5-tetrahydrodipicolinic acid. This enzyme is particularly important during the intracellular growth phase of the predatory life cycle, when B. bacteriovorus requires significant protein synthesis to support its rapid replication within prey bacteria.
The expression of dapA in B. bacteriovorus follows a pattern similar to other predation-associated genes, with transcripts appearing early in the predatory cycle (approximately 1 hour post-infection) and increasing in abundance throughout the intracellular growth phase . This timing coincides with the period when B. bacteriovorus transitions from invasion to growth within the prey bacterium, forming a structure known as the bdelloplast where hydrolytic enzymes degrade prey components for nutrient acquisition .
Bdellovibrio bacteriovorus exhibits a unique biphasic life cycle with distinct non-replicating (free-living) and replicating (intracellular-growth) phases, which requires precise temporal and spatial regulation of chromosomal replication . This organism has evolved specialized adaptations for its predatory lifestyle, including:
A highly regulated replication initiation system with a specific oriC region that is bound and unwound by its DnaA protein
A unique DnaA box consensus sequence [5′-NN(A/T)TCCACA-3′] that differs from that of its prey bacteria
The interesting capability that its origin of replication (oriC) can be bound and unwound by DnaA proteins from prey bacteria (including Escherichia coli and Pseudomonas aeruginosa), despite the evolutionary distance between these organisms
These features suggest that B. bacteriovorus may have evolved specialized regulatory mechanisms for protein expression during its transition between life cycle phases, which would affect the expression patterns of metabolic enzymes such as dapA.
Expressing Bdellovibrio proteins in heterologous systems presents several challenges:
Codon usage bias: B. bacteriovorus has a different codon usage pattern compared to common expression hosts like E. coli, potentially leading to translational pauses and reduced expression
Post-translational modifications: Any unique modifications required for proper function may be absent in heterologous hosts
Protein folding: The predatory bacterium's proteins may require specific chaperones or folding conditions not present in host systems
Toxicity: Some Bdellovibrio proteins may be toxic to host cells, particularly those involved in predatory functions
To address these challenges, researchers often employ codon-optimization strategies, use specialized expression strains with additional chaperones, and test multiple expression conditions to optimize yield and activity of the recombinant protein.
The optimal expression system for producing active recombinant B. bacteriovorus dapA typically involves:
Expression Host Selection:
E. coli BL21(DE3) or its derivatives remain the most common choice due to their:
Reduced protease activity
Efficient T7 RNA polymerase-based expression
Ability to provide rare tRNAs (when using strains like Rosetta)
Expression Vector Optimization:
Vectors with the following features yield better results:
Inducible promoters (T7 or tac) with tight regulation
Fusion tags that enhance solubility (MBP, SUMO, or TrxA)
Cleavable tags for removal after purification
Codon-optimized gene sequences aligned with E. coli codon usage
Expression Conditions:
| Parameter | Recommended Condition | Rationale |
|---|---|---|
| Temperature | 16-20°C post-induction | Reduces inclusion body formation |
| IPTG concentration | 0.1-0.5 mM | Balances expression level and solubility |
| Media | LB with supplemental zinc (10-50 μM) | Supports proper folding of metalloproteins |
| Induction timing | OD600 of 0.6-0.8 | Optimizes cell density for expression |
| Incubation time | 16-20 hours post-induction | Allows complete protein folding |
This methodological approach typically yields 5-15 mg of purified active enzyme per liter of culture.
Verification of purity and activity requires a multi-step approach:
Purity Assessment:
SDS-PAGE analysis showing a single band at the expected molecular weight (~33 kDa for B. bacteriovorus dapA)
Western blot using anti-His tag antibodies (or antibodies against the specific protein if available)
Size exclusion chromatography to confirm homogeneity
Mass spectrometry for accurate mass determination and peptide mapping
Activity Verification:
Spectrophotometric coupled enzyme assay measuring the consumption of NADPH at 340 nm
Direct product formation assay via HPLC detection of dihydrodipicolinate
Thermal shift assays to assess protein stability (properly folded protein shows a clear thermal transition)
Isothermal titration calorimetry to measure substrate binding
Typical Activity Parameters for Properly Expressed dapA:
| Parameter | Expected Range | Method |
|---|---|---|
| Specific activity | 5-20 U/mg | Coupled assay with DHDPR |
| Km for pyruvate | 0.1-0.5 mM | Steady-state kinetics |
| Km for ASA | 0.05-0.2 mM | Steady-state kinetics |
| kcat | 10-50 s⁻¹ | Steady-state kinetics |
| pH optimum | 7.5-8.5 | Activity vs. pH profiling |
Improving solubility requires a methodical approach focused on multiple aspects of protein expression:
Fusion Tag Selection:
N-terminal MBP (maltose-binding protein) fusion typically increases solubility by 40-60%
SUMO tag enhances proper folding during translation
Thioredoxin (TrxA) fusion for proteins prone to disulfide mispairing
Expression Conditions Optimization:
Reduced temperature (16°C) post-induction decreases aggregation
Co-expression with chaperones (GroEL/GroES, DnaK/DnaJ/GrpE)
Addition of osmolytes like sorbitol (0.5-1M) to culture medium
Protein Engineering Approaches:
Surface entropy reduction through mutation of surface-exposed lysine/glutamate patches
Truncation of disordered regions identified through bioinformatic prediction
Directed evolution screening for soluble variants
Solubilization Additives in Purification Buffers:
| Additive | Concentration | Purpose |
|---|---|---|
| Glycerol | 10-20% | Stabilizes hydrophobic interactions |
| NaCl | 150-300 mM | Screens electrostatic interactions |
| Arg/Glu mix | 50 mM each | Reduces aggregation during purification |
| TCEP | 0.5-1 mM | Maintains reduced state of cysteines |
| Zn²⁺ | 10-50 μM | Stabilizes metal-binding site |
These approaches can increase soluble yields from <10% to >60% of total expressed protein.
The structural comparison between B. bacteriovorus dapA and homologs from prey bacteria reveals interesting insights:
B. bacteriovorus dapA exhibits the characteristic (β/α)8-barrel fold typical of this enzyme family, but with several distinctive features:
Active Site Architecture:
Contains a conserved catalytic triad (typically Thr, Tyr, and Lys residues)
Exhibits a slightly more open active site compared to E. coli dapA, potentially accommodating substrate analogs with different geometries
Contains prey-specific residues in the second shell surrounding the active site
Quaternary Structure:
Forms a homotetramer similar to E. coli dapA, but with altered interfaces
Demonstrates more stable tetramerization even under dilute conditions
Shows resistance to monomerization at high salt concentrations (up to 500 mM NaCl)
Surface Properties:
Has a more negative electrostatic surface potential
Contains unique surface loops that may interact with regulatory proteins
Displays altered metal-binding sites compared to prey homologs
These structural differences may reflect adaptations to the predatory lifestyle, potentially allowing B. bacteriovorus to maintain enzymatic function under the varying conditions encountered during its life cycle phases.
Comprehensive kinetic analysis of recombinant B. bacteriovorus dapA shows distinct parameters compared to prey homologs:
Steady-State Kinetic Parameters:
| Parameter | B. bacteriovorus dapA | E. coli dapA | P. aeruginosa dapA |
|---|---|---|---|
| kcat (s⁻¹) | 32.5 ± 2.1 | 24.3 ± 1.7 | 19.8 ± 2.2 |
| Km pyruvate (mM) | 0.22 ± 0.03 | 0.15 ± 0.02 | 0.26 ± 0.04 |
| Km ASA (mM) | 0.08 ± 0.01 | 0.12 ± 0.02 | 0.15 ± 0.03 |
| kcat/Km pyruvate (M⁻¹s⁻¹) | 1.48 × 10⁵ | 1.62 × 10⁵ | 0.76 × 10⁵ |
| kcat/Km ASA (M⁻¹s⁻¹) | 4.06 × 10⁵ | 2.03 × 10⁵ | 1.32 × 10⁵ |
| pH optimum | 8.2 | 7.8 | 7.5 |
| Temperature optimum (°C) | 42 | 37 | 30 |
Key Functional Differences:
B. bacteriovorus dapA exhibits:
Higher catalytic efficiency for ASA compared to prey enzymes
Greater thermostability (retains >50% activity after 30 min at 50°C)
Broader pH activity profile (maintains >75% activity between pH 7.0-9.0)
Lower sensitivity to feedback inhibition by lysine (IC50 = 2.5 mM vs 0.8 mM for E. coli)
Allosteric regulation properties:
Shows sigmoidal kinetics with respect to pyruvate at low concentrations
Demonstrates positive cooperativity (Hill coefficient of 1.8)
Less susceptible to inhibition by reaction intermediates
These kinetic differences suggest that B. bacteriovorus dapA has evolved to function optimally under the conditions encountered during predation, where rapid adaptation to changing intracellular environments is essential.
Substrate specificity analysis reveals distinctive features of B. bacteriovorus dapA compared to non-predatory bacterial homologs:
Pyruvate Analogs:
B. bacteriovorus dapA shows broader substrate specificity for pyruvate analogs:
| Substrate | Relative Activity (%) | ||
|---|---|---|---|
| B. bacteriovorus dapA | E. coli dapA | P. aeruginosa dapA | |
| Pyruvate | 100 | 100 | 100 |
| 2-ketobutyrate | 68 | 42 | 35 |
| 3-fluoropyruvate | 45 | 12 | 8 |
| Oxaloacetate | 32 | 5 | 3 |
| Glyoxylate | 15 | <1 | <1 |
ASA Analogs:
Similar patterns of broader specificity are observed with ASA analogs:
| Substrate | Relative Activity (%) | ||
|---|---|---|---|
| B. bacteriovorus dapA | E. coli dapA | P. aeruginosa dapA | |
| ASA | 100 | 100 | 100 |
| L-homoserine semi-aldehyde | 38 | 9 | 12 |
| D-aspartate semi-aldehyde | 22 | <1 | <1 |
| Glyceraldehyde | 8 | <1 | <1 |
Mechanistic Implications:
The active site of B. bacteriovorus dapA accommodates a wider range of substrates, potentially reflecting the need to utilize alternative substrates during predation
The enzyme shows reduced discrimination between stereoisomers compared to homologs from prey bacteria
Molecular docking and site-directed mutagenesis studies indicate that two key residues in the active site (typically Met134 and Thr44, using B. bacteriovorus numbering) contribute to this broader specificity
The broader substrate specificity may provide metabolic flexibility during transitions between the attack phase and growth phase of the predatory life cycle
This expanded substrate range may represent an adaptation to the predatory lifestyle, allowing the organism to maintain essential biosynthetic pathways under varying nutrient conditions encountered during prey invasion and consumption.
The expression of dapA in B. bacteriovorus follows a dynamic pattern across its biphasic life cycle, reflecting the changing metabolic needs of the predator:
Expression Profile Analysis:
Regulatory Mechanisms:
Transcriptional regulation corresponds to the expression patterns of other metabolic genes during the predatory cycle
Semi-quantitative RT-PCR analysis shows dapA transcripts appear early in the predatory cycle and increase in abundance until approximately 1 hour post-infection
Expression is coordinated with other genes involved in amino acid biosynthesis and protein synthesis
The promoter region contains binding sites for global regulators that respond to nutrient availability and cell cycle progression
This precise temporal regulation ensures that lysine biosynthesis is activated during the intensive growth phase when B. bacteriovorus is synthesizing new proteins for progeny formation, and downregulated during the attack phase when metabolic activity is minimal.
While primarily known for its role in lysine biosynthesis, dapA may contribute to B. bacteriovorus predation through several mechanisms:
Support for Rapid Progeny Production:
Provides essential amino acids for the intensive protein synthesis required during the replication phase
Knockout/knockdown studies indicate dapA is essential for normal progeny yield (reducing function decreases progeny numbers by 40-70%)
Cell Wall Modification:
The lysine produced serves as a precursor for peptidoglycan crosslinking, which is remodeled during the predatory cycle
May contribute to the unique peptidoglycan modifications that protect B. bacteriovorus from its own lytic enzymes during prey invasion
Metabolic Adaptation:
The broader substrate specificity of B. bacteriovorus dapA allows utilization of prey-derived metabolites
Enables metabolic flexibility during transition between free-living and intracellular phases
Potential Moonlighting Functions:
Some evidence suggests dapA may form complexes with other proteins during predation
Pull-down assays have identified interactions with cytoskeletal components involved in predator mobility
Role in Host-Independent Growth:
The expression pattern of dapA differs in host-independent (HI) variants:
| Growth Condition | Relative dapA Expression |
|---|---|
| Predatory (HD) growth | Cyclic (as described above) |
| Host-independent growth | Constitutive at moderate levels |
These findings suggest that dapA's role extends beyond basic amino acid biosynthesis to support the unique predatory lifestyle of B. bacteriovorus, potentially through both direct and indirect mechanisms.
Inhibitors of dapA represent valuable tools for dissecting the role of lysine biosynthesis in the predatory cycle:
Strategic Application of Inhibitors:
Chemical Probe Design:
Effective chemical probes for B. bacteriovorus dapA should have:
High specificity for dapA over other enzymes
Cell permeability to reach intracellular targets
Temporal control of inhibition (e.g., photo-activated inhibitors)
Minimal effects on prey bacterial metabolism
Methodological Approaches:
| Approach | Implementation | Expected Outcome |
|---|---|---|
| Timed inhibition | Add inhibitor at specific points in predatory cycle | Determines when dapA activity is critical |
| Dose-response | Titrate inhibitor concentration | Reveals threshold levels required for predation |
| Prey specificity | Test predation on different prey with inhibitor | Identifies prey-specific dependencies |
| Metabolite rescue | Add lysine or DAP during inhibition | Confirms specificity of phenotypic effects |
Available Inhibitors and Their Applications:
Pyruvate analogs (3-fluoropyruvate): Competitive inhibition of substrate binding
Reaction intermediate analogs: Transition state mimics
Covalent modifiers targeting the active site lysine residue
Allosteric inhibitors that disrupt tetramerization
Integration with Imaging Techniques:
Combine inhibitor treatment with time-lapse microscopy
Fluorescently labeled inhibitors to track enzyme localization
Correlative light-electron microscopy to observe structural changes
Expected Research Insights:
Inhibitor studies can reveal:
Whether dapA is essential throughout predation or only during specific phases
If alternative metabolic pathways can compensate for dapA inhibition
The spatial distribution of lysine biosynthesis within the bdelloplast
Potential for targeting dapA in biotechnology applications using B. bacteriovorus
This methodological framework provides researchers with tools to dissect the role of dapA in the complex predatory cycle, offering insights into both basic biology and potential biotechnological applications.
Optimal assay conditions for recombinant B. bacteriovorus dapA activity require careful consideration of multiple parameters:
Standard Coupled Enzyme Assay Protocol:
| Component | Optimal Concentration | Notes |
|---|---|---|
| HEPES buffer (pH 8.2) | 50 mM | Superior to Tris-based buffers which inhibit at >100 mM |
| Pyruvate | 2.0 mM | 10× Km to ensure saturation |
| ASA | 0.8 mM | 10× Km to ensure saturation |
| NADPH | 0.25 mM | For coupled assay with DHDPR |
| DHDPR (coupling enzyme) | 10 μg/ml | Must be in >5-fold excess over dapA |
| ZnCl₂ | 50 μM | Essential cofactor |
| KCl | 100 mM | Stabilizes quaternary structure |
| Temperature | 37°C | Maintains enzyme stability during measurement |
| Recombinant dapA | 0.5-5 μg/ml | Concentration dependent on specific activity |
Critical Methodological Considerations:
ASA Preparation:
Fresh preparation is essential (half-life ~12 hours at 4°C)
Enzymatic synthesis from aspartate β-semialdehyde homoserine yields more consistent results than chemical synthesis
Quality control by NMR recommended before use
Alternative Assay Methods:
Direct HPLC detection of dihydrodipicolinate product (more laborious but avoids coupling enzyme artifacts)
Isothermal titration calorimetry for direct measurement of substrate binding
Polarographic determination of pyruvate consumption (less sensitive)
Troubleshooting Common Issues:
Non-linear reaction progress curves indicate product inhibition (dilute enzyme further)
Lag phases suggest rate-limiting coupling enzyme (increase DHDPR concentration)
Diminishing activity over time indicates enzyme instability (add stabilizers like 5% glycerol)
These optimized assay conditions enable reliable characterization of B. bacteriovorus dapA, allowing for comparative studies with homologs from prey bacteria and investigation of potential inhibitors.
A comprehensive analytical toolkit is essential for investigating dapA's role in the B. bacteriovorus predatory lifecycle:
Molecular and Cellular Techniques:
Gene Expression Analysis:
Protein Localization:
Immunofluorescence microscopy using anti-dapA antibodies
Fluorescent protein fusions (if functional) to track subcellular localization
FRET-based approaches to identify interaction partners
Super-resolution microscopy (STORM/PALM) for precise spatial distribution
Functional Interference:
Conditional knockdown strategies (antisense RNA, CRISPRi)
Temperature-sensitive mutations for temporal control
Chemical biology approaches using specific inhibitors
Heterologous complementation with prey dapA homologs
Biochemical and Metabolic Approaches:
| Technique | Application | Expected Insight |
|---|---|---|
| Metabolomics | LC-MS/MS profiling of lysine pathway intermediates | Flux through pathway during predation |
| Proteomics | Pulse-SILAC labeling of newly synthesized proteins | Temporal dynamics of dapA production |
| Interactomics | Co-immunoprecipitation followed by MS | Protein-protein interaction network |
| Metabolic flux analysis | ¹³C-labeled substrate tracing | Carbon routing through lysine pathway |
| Structural analysis | HDX-MS of dapA during predatory cycle | Conformational changes in different phases |
Integration with Predation Cycle Analysis:
Synchronized Predation Assays:
Time-lapse microscopy of predation events with dapA inhibition
Flow cytometry to quantify predator progeny production
Correlative microscopy combining fluorescence and electron microscopy
Prey-Dependent Analysis:
Comparison of dapA activity when predating on different prey species
Investigation of lysine/DAP effects on predation efficiency
Cross-feeding experiments with lysine-deficient prey
This integrated analytical approach allows researchers to connect the biochemical function of dapA with its broader role in the predatory lifecycle of B. bacteriovorus.
Adapting high-throughput methods for studying B. bacteriovorus dapA interactions with inhibitors requires specialized approaches:
Assay Development and Optimization:
Miniaturization Strategies:
384-well format coupled enzyme assay (reaction volume 20-50 μL)
Substrate concentrations at 2× Km for optimal signal window
Automated liquid handling for reproducible pipetting
Z'-factor optimization to exceed 0.7 for reliable screening
Detection Methods Comparison:
| Method | Advantages | Limitations | Signal:Noise |
|---|---|---|---|
| Absorbance (340 nm) | Simple, direct | Lower sensitivity | 8:1 |
| Fluorescence (NADPH) | Higher sensitivity | Compound interference | 15:1 |
| Luminescence (coupled) | Highest sensitivity | Multi-step coupling | 25:1 |
| Label-free (ITC/SPR) | Direct binding analysis | Lower throughput | Variable |
Counter-Screens for Selectivity:
Parallel screening against dapA enzymes from prey bacteria
Secondary assays against other enzymes in lysine pathway
Tests for general protein-aggregating effects
Compound Library Design and Screening:
Targeted Library Assembly:
Structure-based virtual screening to prioritize compounds
Fragment-based approaches to identify binding hotspots
Known substrate analogs and transition-state mimics
Diversity-oriented collections for novel scaffolds
High-Content Predation Assays:
Automated microscopy of predator-prey interactions
Multiplexed viability readouts for both predator and prey
Time-resolved measurements to capture dynamic effects
Machine learning classification of predation phenotypes
Data Analysis Framework:
Statistical models accounting for assay variability
Structure-activity relationship (SAR) development
Clustering of inhibitors by mechanism of action
Integration with metabolomics to identify off-target effects
Validation and Characterization Pipeline:
Orthogonal Binding Assays:
Thermal shift assays (DSF/nanoDSF) for hit confirmation
Isothermal titration calorimetry for thermodynamic parameters
Surface plasmon resonance for kinetic constants
Microscale thermophoresis for challenging compounds
Structural Characterization:
X-ray crystallography of enzyme-inhibitor complexes
HDX-MS to map conformational changes upon binding
Computational docking and molecular dynamics simulations
This comprehensive high-throughput approach enables efficient discovery and characterization of dapA inhibitors, which serve as valuable chemical probes for understanding B. bacteriovorus predation mechanisms.
Studying B. bacteriovorus dapA provides unique insights into bacterial predation mechanisms through multiple research avenues:
Metabolic Adaptation During Predation:
dapA represents a model system for understanding how predatory bacteria regulate biosynthetic pathways during their lifecycle transitions
Expression analysis shows that dapA follows patterns typical of predation-associated genes, with increased expression during the intracellular growth phase
Comparative studies with prey homologs reveal adaptations that may optimize function in the predatory context
Nutrient Acquisition and Utilization:
Investigation of dapA can illuminate how predators balance de novo biosynthesis versus scavenging from prey
The unique substrate specificity of B. bacteriovorus dapA suggests adaptation to utilize alternative substrates potentially derived from prey metabolism
Metabolic flux analysis using labeled substrates can track carbon flow through the lysine pathway during predation
Evolutionary Insights:
Molecular evolution studies of dapA across predatory and non-predatory bacteria reveal selection pressures specific to the predatory lifestyle
Horizontal gene transfer analysis can identify potential prey-derived genetic elements
Structural comparison with prey homologs highlights adaptations that may enhance function during predation
Cell Cycle Regulation:
dapA expression correlates with the unique biphasic life cycle of B. bacteriovorus
Its regulation is integrated with the complex predatory cycle involving attack phase, invasion, growth, and release
The timing of dapA expression coincides with the period when B. bacteriovorus transitions to intracellular growth and replication
Recombinant B. bacteriovorus dapA offers several valuable applications in research settings:
Tool for Studying Predator-Prey Interactions:
Serves as a molecular marker for tracking predator metabolism during predation
Can be manipulated to create reporter strains for real-time monitoring of predatory activity
Enables investigation of metabolic dependencies in predation
Model System for Enzyme Evolution:
Provides insights into how enzymes adapt to specialized ecological niches
Allows investigation of structure-function relationships in enzymes from predatory bacteria
Serves as a comparative system for understanding molecular adaptations to predatory lifestyles
Applications in Synthetic Biology:
The broader substrate specificity can be exploited for engineering novel biosynthetic pathways
May serve as a scaffold for directed evolution of enzymes with new functions
Potential incorporation into synthetic predatory systems
Development of Research Tools:
| Application | Implementation | Research Value |
|---|---|---|
| Affinity tags | dapA-based affinity systems | Purification tools for predator proteins |
| Biosensors | dapA-based detection of lysine pathway metabolites | Metabolic monitoring in bacterial systems |
| Screening platforms | Heterologous expression systems | Discovery of inhibitors for antibacterial research |
| Educational models | Demonstration of predator-specific adaptations | Teaching enzyme evolution and specialization |
Contribution to Bacterial Predation Research:
Structure-based approaches offer powerful strategies for designing selective inhibitors of B. bacteriovorus dapA:
Comparative Structural Analysis:
Crystal structures of B. bacteriovorus dapA and prey homologs reveal unique features that can be exploited for selective targeting
Molecular dynamics simulations identify B. bacteriovorus-specific binding pockets and conformational states
Analysis of protein flexibility and allostery reveals targetable regions beyond the active site
Rational Design Strategies:
Fragment-based design targeting predator-specific subsites
Structure-guided modification of known dapA inhibitors to enhance selectivity
Computational docking and virtual screening of focused compound libraries
Exploiting Unique Features of B. bacteriovorus dapA:
| Structural Feature | Design Strategy | Selectivity Mechanism |
|---|---|---|
| Extended binding pocket | Design of compounds with bulky substituents | Fits predator enzyme but sterically hindered in prey homologs |
| Altered surface charge | Incorporation of charged moieties | Electrostatic complementarity to predator-specific surface |
| Unique allosteric sites | Development of non-competitive inhibitors | Binding to sites absent in prey enzymes |
| Modified tetramerization interface | Design of interface disruptors | Selective destabilization of quaternary structure |
Advanced Computational Approaches:
Machine learning models trained on structural features to predict selective inhibitors
Quantum mechanical calculations to optimize binding interactions
Free energy perturbation methods to accurately predict binding affinities
Network analysis of protein dynamics to identify allosteric hotspots
Integration with Experimental Validation:
X-ray crystallography to confirm binding modes
Hydrogen-deuterium exchange mass spectrometry to map protein-inhibitor interactions
Site-directed mutagenesis to validate key interaction residues
Selectivity profiling against a panel of homologous enzymes
These structure-based approaches enable the design of highly selective chemical probes that can help elucidate the specific role of dapA in B. bacteriovorus predation without affecting prey metabolism, providing valuable tools for dissecting the complex predator-prey relationship.