The query conflates two distinct enzymatic systems:
7-Cyano-7-deazaguanine synthase (queC): Catalyzes the ATP-dependent conversion of 7-carboxy-7-deazaguanine to 7-cyano-7-deazaguanine (preQ₀), a precursor in queuosine tRNA modification .
Cobalamin biosynthesis proteins (e.g., cobD): Involved in vitamin B₁₂ synthesis, including cobalamin biosynthesis pathways identified in L. interrogans serovar copenhageni .
| Enzyme/Pathway | Function | Relevance to L. interrogans copenhageni |
|---|---|---|
| queC | Queuosine tRNA modification | No evidence in provided sources |
| cobD | Cobalamin biosynthesis | Recombinant protein available |
The only recombinant protein explicitly linked to this serovar in the search results is cobD (cobalamin biosynthesis protein), not queC.
Cobalamin Biosynthesis:
Chromosome II contains 13 genes (e.g., cobC, cobD, cbiP) encoding enzymes for de novo B₁₂ synthesis .
LPS Biosynthesis:
A frameshift mutation in lic12008 distinguishes Icterohaemorrhagiae from Copenhageni strains .
Prevalence: Copenhageni dominates in the British Isles, with genetic diversity in MLVA patterns .
Virulence: Serovars Icterohaemorrhagiae and Copenhageni account for >50% of severe human leptospirosis cases .
While not present in L. interrogans copenhageni, queC is well-studied in thermophiles and pathogens:
Terminological Ambiguity:
The query may conflate cobD (a confirmed recombinant protein in L. interrogans copenhageni) with queC, which lacks supporting evidence in this context.
Research Gaps:
No genomic or proteomic data for queC in L. interrogans copenhageni.
queC’s role in leptospirosis pathogenesis remains unexplored.
Future Directions:
Validate queC’s presence via WGS or transcriptomics.
Compare cobD and queC functions in B₁₂ vs. tRNA modification.
KEGG: lic:LIC_10288
STRING: 267671.LIC10288
7-Cyano-7-deazaguanine synthase (EC 6.3.4.20) is an enzyme that catalyzes the formation of a nitrile from a carboxylic acid and ammonia at the expense of ATP, specifically converting 7-carboxy-7-deazaguanine to 7-cyano-7-deazaguanine (preQ0) . In Leptospira interrogans, queC is part of the queuosine biosynthetic pathway, which is essential for tRNA modification. This enzyme represents an important component in bacterial metabolism and potentially impacts pathogenesis through its role in translational fidelity.
While specific comparative data for Leptospira queC is limited, queC from other thermophilic bacteria like G. kaustophilus demonstrates high thermostability, a pH optimum of approximately 9.5, and an apparent temperature optimum around 60°C . Leptospira queC likely shares core catalytic mechanisms but may exhibit different biochemical characteristics adapted to the environmental conditions Leptospira encounters during its complex lifecycle. Sequence alignment analysis typically reveals conserved catalytic domains across bacterial species with adaptations in non-catalytic regions.
queC catalyzes the ATP-dependent conversion of 7-carboxy-7-deazaguanine to preQ0, releasing AMP and pyrophosphate as co-products . While specific kinetic parameters for Leptospira queC haven't been fully characterized, similar enzymes show strict substrate specificity for 7-carboxy-7-deazaguanine. The reaction involves ATP hydrolysis coupled to the amidation and subsequent dehydration to form the nitrile group. Experimental determination of Km, Vmax, and catalytic efficiency would require recombinant expression and biochemical assays under varying substrate concentrations and environmental conditions.
For recombinant Leptospira proteins, E. coli-based expression systems have proven effective, as demonstrated with other Leptospira proteins . For queC specifically, consider using the following approach:
Codon-optimized synthetic gene construct
Expression vector with T7 promoter system (pET series)
BL21(DE3) E. coli strain or derivatives
N-terminal His-tag fusion for purification
IPTG-inducible expression system
Recent advances in Leptospira genetics now allow for homologous expression using IPTG-inducible systems, which may provide proteins with native post-translational modifications . The choice between heterologous (E. coli) and homologous (Leptospira) expression depends on research goals and required protein authenticity.
A multi-step purification strategy is recommended:
Immobilized metal affinity chromatography (IMAC) using Ni-NTA for initial capture of His-tagged queC
Size-exclusion chromatography to remove aggregates and impurities
Ion-exchange chromatography for final polishing
| Purification Step | Buffer Composition | Expected Yield | Purity |
|---|---|---|---|
| Crude lysate | 50 mM Tris-HCl pH 8.0, 300 mM NaCl, 10 mM imidazole | 100% | 10-20% |
| IMAC | 50 mM Tris-HCl pH 8.0, 300 mM NaCl, 250 mM imidazole | 60-70% | 70-80% |
| Size exclusion | 20 mM Tris-HCl pH 8.0, 150 mM NaCl | 40-50% | 85-90% |
| Ion exchange | 20 mM Tris-HCl pH 8.0, 0-500 mM NaCl gradient | 30-40% | >95% |
Storage recommendations include lyophilization or storage in Tris/PBS-based buffer with 6% trehalose at pH 8.0 with addition of 5-50% glycerol for long-term storage at -20°C/-80°C . Avoid repeated freeze-thaw cycles as this may compromise enzyme activity.
Multiple complementary approaches should be used:
Structural integrity assessment:
SDS-PAGE (expect >90% purity)
Circular dichroism spectroscopy to assess secondary structure
Thermal shift assays to determine protein stability
Limited proteolysis to assess proper folding
Activity assessment:
Enzyme kinetics should be determined under optimal conditions (likely pH ~9.5, temperature ~60°C based on homologous enzymes) .
While specific structural data for Leptospira queC is not available, inference from homologous proteins suggests:
ATP-binding domain characterized by Walker A and B motifs
Substrate-binding pocket with high specificity for 7-carboxy-7-deazaguanine
Catalytic residues mediating nitrile formation
Possible oligomerization interfaces
Structural prediction using AlphaFold or RoseTTAFold may provide preliminary structural insights pending experimental determination. Site-directed mutagenesis of predicted catalytic residues would help identify essential structural elements.
Based on homologous queC enzymes, Leptospira queC likely exhibits:
These parameters significantly impact experimental design:
Buffer selection: Bicine or CHES buffers for high pH conditions
Temperature control: Precise regulation during kinetic assays
Protein stability considerations: Addition of stabilizing agents for long-term storage
Assay development: pH-sensitive detection methods require careful calibration
A comprehensive pH-activity and temperature-activity profile should be established to determine optimal conditions specific to Leptospira queC.
queC enzymes typically demonstrate strict substrate specificity for 7-carboxy-7-deazaguanine . To investigate specificity determinants:
Test substrate analogs with modifications to the carboxylic acid position, purine ring, or substituents
Analyze binding pocket residues through homology modeling and site-directed mutagenesis
Conduct saturation mutagenesis of residues lining the active site
Utilize isothermal titration calorimetry (ITC) to measure binding affinities of substrates and analogs
Engineering altered specificity could lead to novel biotechnological applications or inhibitor development.
Recent advances in Leptospira genetics allow for powerful CRISPR/Cas9-based approaches:
Gene knockout: IPTG-inducible Cas9 expression combined with constitutive non-homologous end-joining (NHEJ) system facilitates queC knockout generation
Gene knockdown: IPTG-induced dead Cas9 (dCas9) expression enables validation of gene essentiality without complete disruption
Genome editing: Precise modifications to the queC gene to study structure-function relationships
Transcriptional regulation: Monitoring expression under various environmental conditions
These approaches can be implemented using recently developed IPTG-inducible heterologous protein expression systems in Leptospira, significantly expanding the genetic toolkit for studying this bacterium .
The connection between tRNA modification and bacterial pathogenesis is an emerging area of research. To investigate queC's role in Leptospira pathogenesis:
Compare queC expression levels between pathogenic and non-pathogenic Leptospira species
Construct conditional queC mutants and assess virulence in animal models
Identify transcriptional and translational effects of queC deficiency using RNA-seq and proteomics
Evaluate tRNA modification profiles and translational fidelity in queC mutants
Assess stress response and adaptation to host environments in queC-deficient strains
This research could provide insights into novel virulence mechanisms and potential therapeutic targets for leptospirosis, a disease responsible for over 1 million human cases annually .
Development of queC inhibitors could follow this strategic pathway:
High-throughput screening of compound libraries against recombinant Leptospira queC
Structure-based drug design using computational modeling
Fragment-based drug discovery focusing on the ATP binding site
Natural product screening for queC inhibitors
Validation in cellular systems and animal models
| Inhibitor Development Stage | Key Considerations | Success Metrics |
|---|---|---|
| Target validation | Essentiality of queC for Leptospira growth/virulence | Growth inhibition in knockout/knockdown strains |
| Assay development | Robust, reproducible activity measurement | Z-factor >0.7, low coefficient of variation |
| Primary screening | Diverse chemical libraries, natural products | Hit rate ~0.1-1% |
| Hit validation | Counter-screens, dose-response curves | IC50 <10 μM, selectivity index >10 |
| Lead optimization | Improve potency, selectivity, ADMET properties | IC50 <1 μM, acceptable PK profile |
| In vivo validation | Animal models of leptospirosis | Reduced bacterial burden, improved survival |
Given the annual impact of over 1 million human leptospirosis cases , novel therapeutic approaches targeting queC could be significant.
When encountering expression or solubility issues:
Expression optimization:
Test multiple E. coli strains (BL21, Arctic Express, Rosetta)
Vary induction conditions (temperature, IPTG concentration, duration)
Consider fusion partners (MBP, SUMO, TrxA) to enhance solubility
Co-express with molecular chaperones (GroEL/GroES)
Solubility enhancement:
Optimize buffer conditions (pH, ionic strength, additives)
Include stabilizing agents (trehalose, glycerol, reducing agents)
Consider detergents for membrane-associated forms
Implement refolding protocols if inclusion bodies form
Alternative approaches:
To enhance assay performance:
Substrate purity:
Synthesize or obtain high-purity 7-carboxy-7-deazaguanine
Validate substrate identity by NMR and mass spectrometry
Store under appropriate conditions to prevent degradation
Assay optimization:
Data analysis:
Apply appropriate kinetic models (Michaelis-Menten, substrate inhibition)
Use statistical methods to identify and manage outliers
Implement quality control metrics for assay validation
For structural determination through crystallography:
Protein preparation:
Achieve >95% purity through rigorous purification
Verify monodispersity by dynamic light scattering
Remove flexible regions identified through limited proteolysis
Stabilize protein through ligand binding (substrate analogs, ATP analogs)
Crystallization screening:
Implement sparse matrix screens with varying precipitation agents
Test co-crystallization with substrates, products, and cofactors
Explore pH range 7.0-10.0 based on enzyme stability and activity profiles
Vary protein concentration (5-15 mg/mL) and temperature (4-20°C)
Crystal optimization:
Fine-tune promising conditions through grid screens
Apply seeding techniques for improved crystal quality
Consider surface entropy reduction mutations
Explore crystallization additives to improve order
Comparative analysis between pathogenic (e.g., L. interrogans) and non-pathogenic (e.g., saprophytic) Leptospira species may reveal important differences:
Gene expression patterns:
Transcriptional regulation under various environmental conditions
Response to host-associated signals (temperature, osmolarity, serum)
Enzyme characteristics:
Substrate affinity and catalytic efficiency
Temperature and pH optima reflecting ecological niches
Allosteric regulation and protein-protein interactions
Physiological roles:
Impact on tRNA modification patterns
Contribution to stress response mechanisms
Influence on translation of specific virulence factors
This comparative approach can provide insights into Leptospira evolution and adaptation to different lifestyles, leveraging genome sequencing data from diverse Leptospira species .
Analyzing the genomic neighborhood of queC provides crucial contextual information:
Gene clustering:
Co-localization with other queuosine biosynthesis genes (queD, queE, queF)
Presence of tRNA modification enzymes
Proximity to metabolic pathway genes
Regulatory elements:
Promoter analysis and transcription factor binding sites
Presence of riboswitches or attenuators
Small RNA interactions
Evolutionary conservation:
Synteny analysis across Leptospira species
Horizontal gene transfer signatures
Selection pressure analysis
Whole genome sequencing of diverse Leptospira isolates, as prioritized by the leptospirosis research community , facilitates these comparative genomic approaches.
Integrating multiple omics technologies provides comprehensive insights:
Genomics:
Whole genome sequencing to identify queC variants across strains
Comparative genomics to assess evolutionary conservation
Transcriptomics:
RNA-seq to determine expression patterns under different conditions
Ribosome profiling to assess translational impacts
Proteomics:
Global protein expression analysis in queC mutants
Protein-protein interaction studies to identify functional partners
Post-translational modification analysis
Metabolomics:
Quantification of queuosine pathway intermediates
Global metabolic shifts in queC mutants
Structural biology:
Protein structure determination
Molecular dynamics simulations of enzyme mechanism
This multi-omics strategy aligns with research priorities for understanding Leptospira biology and developing improved diagnostic and preventive measures for leptospirosis .