KEGG: lic:LIC_11454
STRING: 267671.LIC11454
Pantothenate synthetase (PanC; EC 6.3.2.1) is an enzyme encoded by the panC gene that catalyzes the essential adenosine triphosphate (ATP)-dependent condensation of D-pantoate and beta-alanine to form pantothenate in bacteria, including Leptospira interrogans. Pantothenate is a key precursor for the biosynthesis of coenzyme A (CoA) and acyl carrier protein (ACP) .
The significance of PanC in research stems from several critical factors:
PanC is absent in mammals but essential for bacterial growth, making it an attractive target for antimicrobial development
Both CoA and ACP are essential cofactors for bacterial metabolism
L. interrogans, particularly serovar Copenhageni, is a major causative agent of leptospirosis, a global zoonotic disease with significant public health impact
The enzyme represents a potential target for developing selective inhibitors that could be effective against leptospirosis
The organization of the panC gene in L. interrogans shows distinctive characteristics compared to other bacterial species:
Unlike many bacterial genes that are organized into operons, L. interrogans genes, including panC, are typically scattered across chromosome I
The L. interrogans serovar Copenhageni genome consists of two circular chromosomes (chromosome I and chromosome II)
The genome contains various metabolic pathways distributed across both chromosomes with some functional links between them
Genome analysis reveals that L. interrogans has complex regulatory systems and signal transduction mechanisms that likely control metabolic genes like panC in response to environmental stimuli
The biochemical properties of L. interrogans PanC reflect its role in bacterial metabolism:
Enzymatic Mechanism: PanC catalyzes a two-step reaction:
Activation of pantoate with ATP to form pantoyl-adenylate
Nucleophilic attack by beta-alanine to form pantothenate with release of AMP
Optimal Conditions: The enzyme functions optimally under conditions that match L. interrogans physiological parameters:
Cofactor Requirements:
Requires divalent metal ions (usually Mg²⁺) for ATP coordination
May interact with components of L. interrogans' complex metabolic network
PanC functions within a broader metabolic network in L. interrogans:
The pantothenate synthesis pathway is critical because L. interrogans requires long-chain fatty acids for growth and metabolism, and these processes depend on CoA, which is derived from pantothenate .
Based on current research practices, the following methodology has proven effective:
Expression System:
E. coli expression system using BL21(DE3) or similar strains
Expression vector with N-terminal or C-terminal His-tag for purification
Optimal induction conditions: 0.5-1 mM IPTG at 30°C for 4-6 hours or 18°C overnight
Purification Protocol:
Cell lysis using sonication or pressure-based methods in buffer containing 50 mM Tris-HCl pH 8.0, 300 mM NaCl, 10% glycerol, and protease inhibitors
Initial purification by Ni-NTA affinity chromatography
Secondary purification by size exclusion chromatography
Final polishing step using ion-exchange chromatography if needed
Quality Assessment:
SDS-PAGE analysis for purity (>90% purity is typically achievable)
Enzymatic activity assay measuring ATP utilization via coupled enzyme reactions
Thermal shift assay to assess protein stability
Dynamic light scattering to confirm monodispersity
Storage:
Optimal storage in buffer containing 20 mM Tris pH 8.0, 150 mM NaCl, 6% trehalose at -80°C
Addition of glycerol (final concentration 20-50%) for preventing freeze-thaw damage
Avoiding repeated freeze-thaw cycles to maintain enzymatic activity
Optimizing high-throughput screening for PanC inhibitors involves several key approaches:
Enzymatic Assay Development:
A coupled enzymatic reaction can be used to monitor PanC activity by measuring the reduction in absorbance at 340 nm due to NADH oxidation
This coupled assay involves:
PanC reaction producing AMP from ATP
Myokinase converting AMP to ADP
Pyruvate kinase converting ADP to ATP with phosphoenolpyruvate
Lactate dehydrogenase converting pyruvate to lactate with NADH oxidation
Assay Optimization Parameters:
Signal-to-background ratio optimization: ≥3:1
Z'-factor: ≥0.7 for robust screening
DMSO tolerance: up to 2% final concentration
Reaction components optimization:
Enzyme concentration: typically 10-50 nM
Substrate concentrations: at or slightly below Km values
Buffer composition: typically 50 mM HEPES pH 7.5, 10 mM MgCl₂, 50 mM KCl
Screening Strategy:
Primary screen at single concentration (10-20 μM)
Confirmation of hits in dose-response format
Counter-screening against coupling enzymes to eliminate false positives
Evaluation of potential inhibitors in whole-cell assays against L. interrogans
Hit Evaluation:
IC₅₀ determination for promising compounds
Mechanism of inhibition studies (competitive, noncompetitive, uncompetitive)
Structure-activity relationship analysis
Assessment of selectivity against mammalian enzymes
This approach has successfully identified inhibitor classes such as 3-biphenyl-4-cyanopyrrole-2-carboxylic acids for Mycobacterium tuberculosis PanC, which could inform similar work on L. interrogans PanC .
While PanC is not a classical virulence factor, its contribution to L. interrogans pathogenesis is multifaceted:
Metabolic Support for Pathogenesis:
PanC enables CoA synthesis, which is essential for fatty acid metabolism - the primary energy source for L. interrogans during infection
L. interrogans' ability to survive in host tissues depends on essential metabolic pathways supported by PanC
Relationship to Virulence Mechanisms:
L. interrogans has a complex array of virulence factors including LigA and LigB proteins, LipL32, and Loa22
While PanC doesn't directly interact with these virulence factors, it provides the metabolic foundation for their expression and function
The energy derived from CoA-dependent pathways supports motility via the periplasmic flagella, which is crucial for bacterial invasion and dissemination
Host-Pathogen Interaction Context:
L. interrogans infection triggers host immune responses including TLR2 and TLR4 activation
The metabolic activity supported by PanC enables the bacteria to persist despite these immune responses
L. interrogans can cause biphasic illness with both anicteric and icteric phases, requiring sustained metabolic activity throughout infection
Research indicates that metabolic enzymes like PanC represent potential vulnerability points in bacterial pathogens, as they are essential for survival during infection but are absent in mammalian hosts .
Comparative analysis of PanC across Leptospira species reveals important distinctions:
Interspecies Comparison:
Serovar-Level Variations:
Evolutionary Considerations:
Pathogenic Leptospira species form a monophyletic group distinct from non-pathogenic species
L. interrogans serovar Copenhageni is closely related to L. interrogans serovar Lai and L. kirschneri
Unlike L. borgpetersenii, which shows genome reduction and increased host dependence, L. interrogans maintains more metabolic flexibility
Targeting PanC for therapeutic development presents both opportunities and challenges:
Advantages:
PanC is absent in mammals, minimizing potential off-target effects
It is essential for bacterial growth and survival of L. interrogans
The enzyme is part of a conserved metabolic pathway, allowing for potential broad-spectrum activity
High-throughput screening methods have been successfully developed for PanC inhibitor discovery
Challenges:
L. interrogans has a complex double membrane structure that may limit inhibitor access
The bacterium possesses numerous efflux mechanisms as part of its transport system
Achieving appropriate tissue distribution to target L. interrogans in infected kidneys and other tissues
Developing inhibitors that can function in the physiological conditions where L. interrogans thrives
Potential Approaches:
Structure-based drug design using recombinant L. interrogans PanC
Development of prodrugs that can penetrate bacterial membranes
Combination therapy with agents that increase membrane permeability
Exploration of natural products with anti-leptospiral activity as starting points
Current Status:
PanC inhibitors have been identified for other bacterial species such as Mycobacterium tuberculosis
These include 3-biphenyl-4-cyanopyrrole-2-carboxylic acids that show activity against both the enzyme and whole bacteria
Similar approaches could be adapted for L. interrogans PanC, though species-specific optimization would be necessary
Current leptospirosis treatment relies on antibiotics like penicillin and doxycycline
Recombinant L. interrogans PanC has potential applications in leptospirosis diagnostics:
Current Diagnostic Landscape:
The microscopic agglutination test (MAT) is the gold standard but has limitations including reduced sensitivity in early disease
PCR methods are effective in early disease but require specialized equipment
Current recombinant antigen-based diagnostics utilize proteins like LipL32, LigA/B, and GroEL
Potential Applications of Recombinant PanC:
Serological Assays:
ELISA-based detection of anti-PanC antibodies in patient sera
Integration into multi-antigen panels to improve sensitivity and specificity
Development of rapid lateral flow assays for point-of-care testing
Protein Microarray Technology:
Chimeric Protein Constructs:
Implementation Considerations:
Validation with serum panels from confirmed leptospirosis cases and appropriate controls
Assessment of cross-reactivity with other infectious diseases (dengue, malaria, etc.)
Determination of sensitivity in different phases of infection
Evaluation of serovar cross-reactivity across different Leptospira interrogans serovars
Research on other leptospiral antigens has demonstrated that recombinant proteins can achieve high specificity (90-97%) in distinguishing leptospirosis from other conditions, suggesting similar potential for properly characterized recombinant PanC .
Comprehensive assessment of recombinant PanC immunogenicity requires multiple complementary approaches:
In Vitro Immunological Assays:
Antibody Recognition Studies:
Western blotting using sera from leptospirosis patients to detect recognition of recombinant PanC
ELISA to quantify antibody binding and determine titers
Epitope mapping to identify immunodominant regions
Cell-Based Assays:
Peripheral blood mononuclear cell (PBMC) stimulation to assess T-cell responses
Cytokine profiling following stimulation with recombinant PanC
Antigen presentation assays with dendritic cells
Animal Immunization Studies:
Protocol Design:
Immunological Evaluation:
Protection Assessment:
Challenge studies with virulent L. interrogans
Monitoring survival, bacterial burden, and histopathological changes
Evaluation of sterilizing immunity using culture and PCR methods
This approach follows similar methodologies used successfully with other leptospiral proteins such as LigA, LigB, and chimeric constructs like rChi2 .
Structure-function studies should combine multiple experimental approaches:
Structural Analysis:
X-ray Crystallography:
Crystallization of purified recombinant PanC (apo-enzyme)
Co-crystallization with substrates, product, and potential inhibitors
Analysis of active site architecture and substrate binding determinants
Alternative Structural Methods:
Nuclear Magnetic Resonance (NMR) for dynamic studies
Cryo-electron microscopy for larger complexes
Small-angle X-ray scattering (SAXS) for solution conformation
Functional Characterization:
Enzyme Kinetics:
Determination of key kinetic parameters (Km, kcat, kcat/Km) for both substrates
Assessment of potential allosteric regulation
pH and temperature dependence profiles
Metal ion requirements and effects
Mutagenesis Studies:
Site-directed mutagenesis of catalytic residues
Alanine-scanning of substrate binding pocket
Creation of chimeric enzymes with PanC from other species
Molecular Dynamics and Computational Approaches:
Simulation Studies:
Molecular dynamics simulations to analyze protein flexibility
Substrate binding and product release pathways
Identification of potential allosteric sites
Virtual Screening:
Structure-based virtual screening for potential inhibitors
Docking studies to predict binding modes
Quantum mechanical/molecular mechanical (QM/MM) studies of reaction mechanism
Correlation with Biological Function:
Gene Knockout/Complementation:
Generation of L. interrogans panC mutants (if technically feasible)
Complementation studies with wild-type and mutant versions
Assessment of impact on bacterial physiology and virulence
Expression Analysis:
Examination of panC expression under various environmental conditions
Effect of host factors on expression levels
Correlation with expression of related metabolic genes
Development of selective PanC inhibitors requires a systematic approach:
Target Validation and Assay Development:
Biochemical Validation:
Confirmation that L. interrogans PanC is essential for bacterial growth
Development of reliable enzymatic assays for inhibitor screening
Establishment of appropriate positive controls and assay parameters
Assay Cascade Design:
Primary biochemical screen
Secondary whole-cell activity assessment
Selectivity against human enzymes
ADME and toxicity evaluation
Inhibitor Discovery Strategies:
High-Throughput Screening:
Diverse compound libraries (>100,000 compounds)
Fragment-based screening approaches
Natural product libraries
Structure-Based Design:
Utilization of X-ray crystal structures
Virtual screening of in silico libraries
Fragment growing/linking strategies
Structure-activity relationship development
Optimization Parameters:
Potency Considerations:
IC₅₀ < 1 μM against purified enzyme
MIC < 10 μg/mL against L. interrogans
Selectivity index >50 (ratio of cytotoxicity to antibacterial activity)
Physicochemical Properties:
Compliance with Lipinski's rules for oral bioavailability
Optimization for penetration of the leptospiral outer membrane
Stability in physiological conditions
Pharmacokinetic Considerations:
Distribution to tissues where Leptospira localizes (particularly kidneys)
Appropriate half-life to maintain effective concentrations
Limited metabolism and excretion
This approach has been successful for M. tuberculosis PanC, where 3-biphenyl-4-cyanopyrrole-2-carboxylic acids were identified as potent inhibitors with whole-cell activity . A similar methodology could be adapted for L. interrogans PanC.
The genomic context provides important insights for research design:
Genomic Organization:
Unlike many bacteria, leptospiral genes including panC are not organized into operons but are scattered across chromosome I
This dispersed organization may indicate unique regulatory mechanisms that should be considered in expression studies
Comparative genomics between L. interrogans and other spirochetes like Borrelia and Treponema can highlight important differences in metabolic organization
Regulatory Considerations:
Promoter Analysis:
Identification of potential regulatory elements in the panC promoter region
Analysis of binding sites for known transcription factors
Reporter gene assays to assess promoter activity under different conditions
Expression Coordination:
Investigation of potential co-regulation with other metabolic genes
RNA-seq analysis under various environmental conditions
Correlation with expression of other genes in the CoA biosynthetic pathway
Evolutionary Context:
Phylogenetic Analysis:
Comparison of panC sequences across Leptospira species and serovars
Assessment of selection pressure on different domains
Correlation with host range and pathogenicity
Genome Reduction Analysis:
Functional Genomics Approaches:
Transposon Mutagenesis:
Assessment of panC essentiality through transposon insertion site analysis
Identification of genetic interactions through synthetic lethality screens
Conditional expression systems to regulate panC levels
CRISPR-Cas Systems:
The genomic context analysis helps inform experimental design by highlighting L. interrogans-specific features that distinguish it from model organisms where PanC has been studied previously.
Evaluating PanC-host immune interactions requires multiple complementary approaches:
Antigen Presentation and Recognition:
B-cell Epitope Mapping:
Peptide array analysis to identify immunodominant epitopes
Phage display libraries to define antibody binding sites
Structural analysis of antibody-antigen complexes
T-cell Response Analysis:
Identification of MHC-binding epitopes using prediction algorithms and validation assays
T-cell proliferation assays with synthetic peptides
Cytokine profiling to characterize Th1/Th2/Th17 responses
Animal Model Studies:
Immune Response Characterization:
Detailed analysis of antibody isotypes and subclasses following immunization
Assessment of cell-mediated immunity through adoptive transfer experiments
Cytokine responses in different tissues following challenge
Protection Mechanisms:
Passive transfer of anti-PanC antibodies to evaluate protection
Depletion of specific immune cell populations to determine their contribution
Correlation of immune parameters with protection against challenge
Human Studies:
Patient Sample Analysis:
Screening of sera from leptospirosis patients for anti-PanC antibodies
Comparison of acute and convalescent phases
Correlation with disease severity and outcomes
Cross-reactivity Assessment:
Evaluation of potential cross-reactivity with human proteins
Assessment of autoimmune potential
Analysis of cross-reactivity with other bacterial PanC enzymes
Immunomodulatory Effects:
Innate Immune Response:
Assessment of interaction with pattern recognition receptors
Analysis of dendritic cell maturation and antigen presentation
Evaluation of NK cell activation
Adaptive Immune Programming:
Impact on T-cell differentiation pathways
Effects on B-cell affinity maturation
Long-term memory formation
Similar approaches with other leptospiral antigens have demonstrated their utility in understanding immune responses to proteins such as LigA/B, LipL32, and chimeric constructs .