KEGG: lic:LIC_12841
STRING: 267671.LIC12841
DapL (EC 2.6.1.83) is an enzyme involved in the biosynthesis of the essential amino acid L-lysine through a recently discovered variant of the diaminopimelate (DAP) pathway. The significance of DapL in L. interrogans stems from its role in a single-step transamination reaction that converts tetrahydrodipicolinate (THDPA) directly to L,L-diaminopimelate (L,L-DAP), bypassing three enzymatic steps (DapD, DapC, and DapE) found in the acyl-DAP pathways .
This pathway is particularly important because:
It provides the essential metabolite meso-diaminopimelate (meso-DAP), which is critical for peptidoglycan crosslinking in bacterial cell walls
L-lysine produced in this pathway is essential for protein synthesis
The pathway is absent in mammals, making it an attractive target for antimicrobial development
DapL is found in several pathogenic bacteria, including Leptospira interrogans
The DapL enzyme functions as a homodimer, with each subunit approximately 50 kDa in size. Each active site requires coordination between both subunits for optimal function, involving one loop on the minor arm and two loops from the major arm in the dimer interface .
The DapL pathway represents one of four known variants of the diaminopimelate (DAP) pathway for lysine biosynthesis. The key differences between these pathways lie in how they synthesize meso-DAP:
The DapL pathway: Uses a single transamination reaction catalyzed by DapL to convert THDPA directly to L,L-DAP, bypassing three enzymatic steps present in other pathways
The acyl pathways (two variants): Require multiple enzymes (DapD, DapC, and DapE) to convert THDPA to L,L-DAP through acyl intermediates
The meso-diaminopimelate dehydrogenase (Ddh) pathway: Directly converts THDPA to meso-DAP in a single reductive amination step
The DapL pathway is distinguishable by its efficiency, requiring fewer enzymatic steps and energy expenditure. This streamlined process may confer evolutionary advantages in certain environments and makes the pathway an interesting target for both basic research and potential antimicrobial development .
Based on established protocols for recombinant leptospiral proteins, the following expression system and conditions are recommended for L. interrogans DapL:
Expression System:
Escherichia coli is the preferred heterologous expression host
Recommended strains include KS330 or BL21(DE3) derivatives for high-level expression
Vector Systems:
pET expression vectors (particularly pET-28a or pET-32a) have shown effectiveness for leptospiral proteins
pMalC2 vector expressing DapL as a fusion with maltose binding protein (MBP) provides advantages:
Expression Conditions:
Induction with IPTG (0.1-1.0 mM) when culture reaches OD600 of 0.5-0.7
Post-induction growth at lower temperatures (16-25°C) for 4-16 hours improves solubility
Supplementation with PLP (pyridoxal-5'-phosphate) may enhance proper folding of this aminotransferase
Purification Strategy:
For His-tagged proteins: Ni-NTA affinity chromatography
For MBP-fusion proteins: Amylose affinity chromatography
Further purification by ion-exchange and/or size-exclusion chromatography
Optimization of purification buffers should include 5-10% glycerol and low concentrations of reducing agents to maintain stability
When optimizing these conditions, expression levels of 10+ mg of purified recombinant protein per liter of culture can be achieved, similar to yields reported for other recombinant leptospiral proteins .
Verification of structural integrity and enzymatic activity of purified recombinant DapL involves multi-faceted analytical approaches:
Structural Integrity Assessment:
SDS-PAGE analysis: To confirm molecular weight and purity
Western blot: Using anti-His, anti-MBP, or anti-DapL antibodies for identity confirmation
Size-exclusion chromatography: To verify the dimeric state essential for DapL activity
Circular dichroism (CD) spectroscopy: For secondary structure analysis
Thermal stability assays: To evaluate protein folding and stability
Enzymatic Activity Verification:
Two-enzyme system assay: Measuring the physiological reverse activity in a coupled reaction with:
Three-enzyme system assay: For the physiologically relevant forward direction in a reaction containing:
These complementary approaches ensure both the structural integrity and functional activity of the purified recombinant DapL protein, critical for subsequent experimental applications.
Comparative analysis of L. interrogans serovar Copenhageni DapL (LiDapL) with other bacterial DapL orthologs reveals important structural and functional differences:
Structural Comparisons:
LiDapL shares the core structural features of the DapL family: a homodimeric arrangement with each monomer consisting of a major and minor domain
Multiple sequence alignments show conserved active site residues across DapL orthologs, including key catalytic lysine residues that interact with the PLP cofactor
The second shell of residues proximal to the active site show greater variation between orthologs, which may contribute to different inhibition profiles
Functional Differences:
Inhibition studies with five small molecule inhibitors from four structural classes (hydrazide, rhodanine, barbiturate, and thiobarbiturate) revealed varying sensitivities among DapL orthologs:
LiDapL showed similar inhibition patterns to Arabidopsis thaliana DapL (AtDapL)
Verrucomicrobium spinosum DapL (VsDapL) and Chlamydomonas reinhardtii DapL (CrDapL) displayed different inhibition patterns
CrDapL was notably insensitive to hydrazide-based inhibitors (IC50 >200 μM)
VsDapL showed highest sensitivity to barbiturate and thiobarbiturate inhibitors (IC50 ~5 μM)
Structural Basis for Functional Differences:
Molecular modeling based on the known AtDapL structure suggests that while active sites are conserved, variations in proximal residues affect inhibitor interactions
These differences may create altered binding pockets that influence substrate specificity and inhibitor sensitivity
The comparative analysis of DapL orthologs provides valuable insights into the structural basis of functional differences and can guide the development of species-specific inhibitors for antimicrobial applications.
Development of DapL-based diagnostic assays for leptospirosis requires careful consideration of several methodological aspects:
Recombinant Protein Design Strategies:
Multiepitope approach: Similar to successful recombinant multiepitope protein (r-LMP) strategies, designing recombinant DapL constructs that incorporate multiple immunogenic epitopes can enhance diagnostic sensitivity
Domain-specific constructs: Expressing specific domains rather than full-length DapL may improve specificity by eliminating cross-reactive epitopes
Epitope selection: Computational prediction of B-cell epitopes unique to pathogenic Leptospira can increase specificity
Assay Platforms:
ELISA-based detection:
Lateral flow assays:
Rapid point-of-care testing format
Incorporation of recombinant DapL as capture antigen
Sensitivity and Specificity Considerations:
Validation with sera from confirmed leptospirosis cases, both acute and convalescent phases
Testing against sera from other febrile illnesses to ensure specificity
Benchmarking against gold standard microscopic agglutination test (MAT)
Aim for sensitivity and specificity levels comparable to successful recombinant protein-based tests (>90%)
Performance Enhancement Strategies:
Using tandem repeats of DapL epitopes to amplify antigenic signal
Incorporating flexible linkers (e.g., tetraglycyl) between epitopes to ensure accessibility
Combining DapL with other leptospiral antigens like LipL32 for improved sensitivity
The recombinant protein approach offers advantages over whole-cell antigen-based assays, potentially circumventing drawbacks such as cross-reactivity and batch variability in traditional leptospirosis diagnostics .
Recombinant L. interrogans DapL (LiDapL) serves as a valuable platform for inhibitor discovery and antimicrobial development through the following methodological approaches:
High-Throughput Screening Systems:
Enzymatic activity-based screening:
Thermal shift assays:
Measuring changes in protein thermal stability upon ligand binding
Cost-effective and rapid approach for initial screening
Particularly useful for identifying fragments or scaffold molecules
Structure-Based Drug Design:
Homology modeling:
Fragment-based approaches:
Screening of fragment libraries against recombinant LiDapL
Building larger inhibitors by linking or growing fragments with confirmed binding
Known Inhibitor Classes and Optimization:
Previous studies have identified four main structural classes of DapL inhibitors:
IC50 ranges for these inhibitors against LiDapL can guide medicinal chemistry efforts:
Structure-activity relationship (SAR) studies based on existing scaffolds
Chemical modification to improve potency, selectivity, and pharmacokinetic properties
Focus on compounds with preferential activity against LiDapL compared to other DapL orthologs
In Vitro and In Vivo Validation:
Whole-cell assays:
Testing inhibitor efficacy against live L. interrogans
Correlation of enzyme inhibition with antimicrobial activity
Genetic validation:
Animal model testing:
Evaluation of promising inhibitors in animal models of leptospirosis
Assessment of efficacy, pharmacokinetics, and safety parameters
This systematic approach leverages the unique properties of LiDapL to develop targeted antimicrobials with potential advantages over broad-spectrum antibiotics for treating leptospirosis.
Analyzing genetic diversity of DapL among different L. interrogans serovars requires a multifaceted approach combining molecular techniques with bioinformatic analysis:
Molecular Characterization Methods:
Gene sequencing and alignment:
Restriction enzyme analysis (REA):
Multiple-locus variable-number tandem repeat analysis (MLVA):
Whole genome sequencing and comparative genomics:
Complete genome sequencing of multiple isolates
Identification of dapL sequence variants in genomic context
Analysis of surrounding genetic elements that may influence expression
Bioinformatic Analysis Approaches:
Sequence conservation analysis:
Calculation of sequence identity and similarity percentages
Identification of conserved versus variable regions
Mapping of variation to functional domains
Structural prediction and modeling:
Selection pressure analysis:
Calculation of dN/dS ratios to identify patterns of selection
Identification of positively selected sites that may contribute to functional differences
Correlation with Phenotypic Characteristics:
Association of dapL variants with virulence differences between serovars
Relationship between dapL sequence and enzyme kinetic parameters
Correlation with serovar-specific epidemiological patterns
By applying these complementary approaches, researchers can develop a comprehensive understanding of DapL genetic diversity across L. interrogans serovars, potentially revealing insights into strain-specific pathogenicity and evolutionary adaptations.
Distinguishing between cross-reactivity and specific serological responses to DapL in leptospirosis patients requires sophisticated methodological approaches:
Pre-analytical Considerations:
Serum pre-adsorption:
Carefully selected control groups:
Analytical Methods to Minimize Cross-Reactivity:
Recombinant protein design:
Expression of unique regions of DapL that lack homology with proteins from other organisms
Site-directed mutagenesis of potential cross-reactive epitopes
Creation of chimeric constructs focusing on leptospira-specific epitopes
Western blot analysis:
Competitive ELISA approaches:
Pre-incubation of sera with soluble homologous or heterologous antigens
Quantification of inhibition to determine specificity
Calculation of cross-reactivity percentages
Data Analysis and Interpretation:
Statistical approaches:
Comparative antibody profiling:
Analysis of IgM versus IgG responses to differentiate acute from convalescent cases
Evaluation of antibody avidity to distinguish between primary and secondary infections
Monitoring of response patterns over time
Validation Studies:
Testing against panels of sera from microscopic agglutination test (MAT)-confirmed cases
Evaluation against different L. interrogans serogroups to determine cross-serovar reactivity
Analysis across acute and convalescent phases of illness
Based on studies with other leptospiral recombinant antigens, IgG responses may be more reliable than IgM for recombinant protein-based assays, with sensitivities of 56-94% from acute to convalescent phases for highly immunogenic proteins like LipL32 .
Enhancing the immunogenicity of recombinant DapL for vaccine development requires a multifaceted approach addressing antigen design, delivery systems, and adjuvant selection:
Antigen Design Strategies:
Chimeric construct development:
Epitope optimization:
Post-translational modifications:
Expression in systems that allow for proper glycosylation if applicable
Lipidation to enhance innate immune activation
Consideration of the native lipidation status of the protein in vivo
Expression/Delivery Systems:
Bacterial expression platforms:
E. coli-based systems for cost-effective production
Alternative expression hosts if protein folding or modifications are concerns
Advanced delivery vehicles:
Adjuvant Selection and Optimization:
The choice of adjuvant is critical for maximizing immune responses to recombinant proteins. Options include:
Aluminum-based adjuvants:
Oil-in-water emulsions:
Enhanced immune stimulation compared to alum alone
Potential for balanced Th1/Th2 responses
Pattern recognition receptor agonists:
TLR agonists (e.g., MPLA, CpG)
NOD-like receptor agonists
Combination adjuvant systems targeting multiple innate pathways
Immunological Assessment:
Antibody response characterization:
Titer determination by ELISA
Isotype profiling (IgG1, IgG2a, etc.)
Neutralization assays
Opsonophagocytic activity
Cellular immunity evaluation:
T-cell proliferation assays
Cytokine profiling (IFN-γ, IL-4, IL-17, etc.)
Memory B and T cell assessment
Protection studies:
The most successful approaches are likely to combine multiple strategies, optimizing both the antigen and the immune context in which it is presented.
Investigating the role of DapL in L. interrogans pathogenesis and host-pathogen interactions requires a comprehensive research approach spanning molecular, cellular, and in vivo methodologies:
Genetic Manipulation Strategies:
Gene knockout or knockdown:
Targeted mutagenesis of dapL if feasible in L. interrogans
Conditional expression systems to control DapL levels
CRISPR-Cas9 approaches if applicable
Note: Essential gene status may necessitate conditional approaches
Complementation studies:
Site-directed mutagenesis:
Targeted modification of active site residues
Alteration of surface-exposed regions potentially involved in host interactions
Creation of catalytically inactive variants for separating enzymatic and non-enzymatic functions
Protein Localization and Interaction Studies:
Subcellular localization:
Host molecule binding assays:
Protein-protein interaction mapping:
Co-immunoprecipitation with host or bacterial proteins
Yeast two-hybrid or bacterial two-hybrid screening
Pull-down assays with recombinant DapL
Proximity labeling approaches in infected cells
Functional Studies:
Cell adhesion and invasion assays:
Immune modulation assessment:
Cytokine profiling in response to DapL stimulation
Evaluation of inflammasome activation
Analysis of neutrophil and macrophage responses
Complement interaction studies
Metabolic role investigations:
In Vivo Approaches:
Animal infection models:
Comparison of wild-type and DapL-modified strains
Assessment of bacterial burden in tissues
Histopathological evaluation
Immune response characterization
Anti-DapL immunization studies:
Vaccination with recombinant DapL
Challenge with virulent L. interrogans
Evaluation of protective efficacy
Passive transfer of anti-DapL antibodies
Through this multi-dimensional approach, researchers can build a comprehensive understanding of DapL's potential dual roles in both essential metabolism and host-pathogen interactions during L. interrogans infection.
Researchers working with recombinant L. interrogans DapL encounter several technical challenges. The following troubleshooting guide addresses these issues with practical solutions:
Expression and Solubility Issues:
| Challenge | Potential Causes | Solutions |
|---|---|---|
| Low expression yield | Codon bias, toxicity, mRNA instability | - Use codon-optimized gene synthesis - Try different E. coli strains (BL21, Rosetta) - Use tightly controlled induction systems - Reduce incubation temperature (16-20°C) |
| Protein insolubility/inclusion bodies | Misfolding, hydrophobic regions, improper disulfide formation | - Express as fusion with solubility tags (MBP, SUMO, TrxA) - Co-express with chaperones - Use lysis buffers with mild detergents - Optimize induction conditions (lower IPTG, longer expression time) |
| Protein degradation | Protease activity, intrinsic instability | - Add protease inhibitor cocktails - Include stabilizing agents (glycerol, arginine) - Maintain samples at 4°C - Consider rapid purification protocols |
Purification Challenges:
| Challenge | Potential Causes | Solutions |
|---|---|---|
| Poor binding to affinity resins | Tag inaccessibility, interference from contaminants | - Place tag at opposite terminus - Use longer linkers between protein and tag - Optimize binding buffers (adjust salt, pH) - Include low concentrations of detergent |
| Co-purification of contaminants | Non-specific binding, protein-protein interactions | - Include step gradients in elution - Add secondary purification steps (ion exchange, size exclusion) - Try different affinity tags - Consider on-column refolding protocols |
| Loss of enzymatic activity | Cofactor loss, oxidation, improper folding | - Add PLP cofactor during purification - Include reducing agents (DTT, β-mercaptoethanol) - Avoid freeze-thaw cycles - Store with glycerol at -80°C |
Assay-Related Issues:
Structural Analysis Challenges:
| Challenge | Potential Causes | Solutions |
|---|---|---|
| Difficulty obtaining crystals | Flexibility, heterogeneity, aggregation | - Remove flexible regions (construct optimization) - Use surface entropy reduction - Try crystallization chaperones - Consider alternative structural methods (cryo-EM, SAXS) |
| Instability during biophysical studies | Buffer incompatibility, oligomerization, aggregation | - Screen buffer conditions systematically - Add stabilizing agents (amino acids, polyols) - Use analytical SEC to monitor oligomeric state - Consider chemical crosslinking for stabilization |
By applying these targeted solutions to specific challenges, researchers can improve the reliability and reproducibility of their work with recombinant L. interrogans DapL, advancing both basic understanding and applied research in leptospirosis.
Generating highly specific antibodies against L. interrogans DapL with minimal cross-reactivity requires strategic approaches throughout the antibody production pipeline:
Antigen Design and Selection:
Epitope analysis and selection:
Perform in silico analysis to identify DapL-specific regions with low homology to other bacterial proteins
Use algorithms to predict surface-exposed epitopes
Avoid highly conserved regions such as active sites that may be similar across aminotransferases
Recombinant constructs for immunization:
Generate truncated fragments containing unique regions rather than full-length protein
Remove tags (e.g., MBP, His) before immunization or use cleavable linkers
Consider synthetic peptides corresponding to unique regions
Use both domain-specific constructs and full-length protein to compare specificity
Immunization Strategies:
Host selection:
Choose species phylogenetically distant from bacteria (rabbits, guinea pigs, or goats)
Consider using multiple species to obtain diverse antibody repertoires
For monoclonal antibodies, optimize mouse strain selection based on MHC haplotype
Adjuvant and protocol optimization:
Use adjuvants that promote high-affinity antibody development
Implement extended immunization schedules with gradually decreasing antigen doses
Monitor antibody titers and specificity throughout the immunization process
Employ DNA prime-protein boost strategies for enhanced responses
Antibody Purification and Characterization:
Affinity purification approaches:
Use recombinant DapL coupled to solid support for specific antibody isolation
Implement negative selection against potential cross-reactive proteins
Consider epitope-specific purification using synthetic peptides
Elute with mild conditions to preserve antibody activity
Cross-reactivity elimination:
Validation and Characterization:
Comprehensive specificity testing:
Western blot against whole cell lysates from various bacterial species
Test against different leptospiral serovars to determine cross-serovar reactivity
Include recombinant protein panels of related aminotransferases
Perform immunoprecipitation followed by mass spectrometry to identify all targets
Functional validation:
Documentation of cross-reactivity patterns:
Systematically catalog any cross-reactivity with other leptospiral proteins, particularly LRR-containing proteins
Quantify relative affinities for specific versus cross-reactive targets
Create detailed epitope maps to explain observed patterns
Share characterization data to benefit the research community
By implementing these comprehensive strategies, researchers can produce antibodies with enhanced specificity for DapL, facilitating more precise studies of its expression, localization, and function in L. interrogans.
The study of recombinant L. interrogans DapL presents numerous exciting opportunities for future research in both fundamental science and applied fields:
Basic Science Directions:
Structural Biology and Enzymology:
Metabolic Integration:
Systems biology approaches to understand DapL's role in the broader metabolic network
Flux analysis of the lysine biosynthetic pathway under different growth conditions
Investigation of potential moonlighting functions beyond lysine biosynthesis
Exploration of metabolic interactions with host environments during infection
Evolutionary and Comparative Genomics:
Phylogenetic analysis of DapL across Leptospira species and other bacteria
Investigation of horizontal gene transfer events in DapL evolution
Analysis of selection pressures on dapL gene across pathogenic and non-pathogenic species
Comparative genomics of the DapL pathway in different bacterial lineages
Translational Research Opportunities:
Diagnostic Applications:
Therapeutic Development:
Vaccine Research:
Emerging Technologies and Approaches:
CRISPR-Based Methods:
Development of CRISPR interference approaches for conditional dapL knockdown
CRISPR-based screens to identify genetic interactions with dapL
Gene editing to introduce point mutations in endogenous dapL
Single-Cell Technologies:
Investigation of dapL expression heterogeneity within Leptospira populations
Single-cell metabolomics to understand metabolic consequences of dapL manipulation
Spatial transcriptomics to map dapL expression in infected tissues
Advanced Imaging Approaches:
Super-resolution microscopy to visualize DapL localization in leptospiral cells
Correlative light-electron microscopy to connect function with ultrastructure
Live-cell imaging with fluorescent DapL fusions to track dynamics