KEGG: btk:BT9727_4501
Phosphoenolpyruvate carboxykinase (PEPCK or pckA) is a key metabolic enzyme that catalyzes the reversible decarboxylation of oxaloacetate to phosphoenolpyruvate (PEP) and carbon dioxide, with ATP as a phosphate donor. In B. thuringiensis metabolism, pckA plays crucial roles in:
Gluconeogenesis: Converting non-carbohydrate carbon sources to glucose
TCA cycle regulation: Maintaining balance of metabolic intermediates
Carbon flux distribution: Directing carbon between central metabolism and specialized functions
Sporulation metabolism: Supporting energy requirements during spore formation
As illustrated in search result , pckA participates in metabolic changes associated with spore development, crystal protein formation, and mother cell lysis in B. thuringiensis. The enzyme is differentially regulated during various growth phases, with particular importance during nutrient limitation and sporulation when metabolic resources are being redirected.
B. thuringiensis subsp. konkukian (serotype H34) has several distinctive features that differentiate it from typical insecticidal Bt strains:
Phylogenetic position: Analysis shows it is more closely related to B. cereus and B. anthracis than to typical insecticidal Bt strains
Clinical relevance: Unlike most Bt strains, subsp. konkukian was originally isolated from a human wound infection, demonstrating potential opportunistic pathogenic capabilities
Crystal protein profile: While classified as B. thuringiensis based on protein crystal production, its insecticidal activity profile differs from commercial Bt biopesticides
Genomic features: The pBT9727 plasmid in strain 97-27 shares significant homology with the pXO2 plasmid of B. anthracis
Biochemical identification of B. thuringiensis subsp. konkukian reveals the following characteristic profile :
| Test | Result |
|---|---|
| Catalase | Positive |
| Oxidase | Negative |
| Mobility | Positive |
| Beta-hemolysis | Positive |
| Maltose utilization | Acid production |
| Glucose utilization | Acid production |
| Galactose utilization | Negative |
| Salicin utilization | Negative |
Isolation and identification of B. thuringiensis subsp. konkukian involves a multi-step approach:
Selective isolation:
Heat treatment (80°C for 10 minutes) to select for spore-forming bacteria
Growth on mannitol-egg yolk-polymyxin (MYP) agar
Incubation at 30°C for 24-48 hours
Morphological characterization:
Molecular identification:
Biochemical confirmation:
Definitive identification requires combining these approaches, with molecular techniques providing the highest specificity for subspecies determination.
Modification of pckA in recombinant B. thuringiensis strains significantly impacts both sporulation and crystal protein dynamics through alterations in central carbon metabolism:
For sporulation:
The enzyme fulfills an unusual role in the final TCA cycle steps during sporulation
Disruption of pckA affects the metabolic balance in sporulating cells, potentially resulting in conditionally asporogenous phenotypes
Altered carbon flux affects the synthesis of dipicolinic acid (DPA) and other spore components
For crystal protein formation:
pckA activity influences amino acid availability for crystal protein synthesis
Modified carbon flux can affect the timing of crystal protein accumulation relative to sporulation
In some cases, pckA modification can lead to overexpression of certain Cry proteins while reducing others
The metabolic changes associated with sporulation are intricately connected to crystal protein formation and mother cell lysis. When pckA is modified, these processes become unbalanced, as demonstrated in studies of leuB mutants where expression of some cry genes is reduced while others (like Cry1Ac) may be overexpressed . Additionally, modified strains often show delayed or blocked mother cell lysis, which has implications for protein crystal release.
The relationship between pckA and the PlcR virulence regulon in B. thuringiensis subsp. konkukian reveals important connections between metabolism and pathogenicity:
The PlcR regulon:
Functions as a pleiotropic transcriptional activator regulating numerous virulence factors
Binds to a specific DNA sequence (PlcR box) in the promoter regions of target genes
Requires the product of the papR gene, which acts as a quorum-sensing effector
Significantly affects the pathogenicity of B. cereus and B. thuringiensis in both insects and mice
pckA's integration with PlcR:
Metabolic enzymes like pckA support the energy requirements for virulence factor production
PlcR inactivation decreases the pathogenicity of B. thuringiensis, suggesting coordination with metabolic functions
In B. thuringiensis subsp. konkukian (which contains PlcR with 100% sequence identity to Bt reference sequences), pckA activity likely supports the metabolic demands of virulence factor production
Functional evidence:
The disruption of PlcR considerably reduces the amounts of up to 56 exported proteins in B. cereus
Studies of B. thuringiensis virulence found that virulence was fully restored in complemented mutants for some PlcR-regulated genes, demonstrating their direct involvement in pathogenicity
Metabolic adaptations mediated by enzymes like pckA allow persistence in varied host environments
This relationship is particularly significant in B. thuringiensis subsp. konkukian due to its closer relationship to pathogenic B. cereus than to conventional insecticidal Bt strains, making it an important model for studying virulence regulation in the B. cereus group.
Several advanced approaches have been developed for marker-free modifications of pckA in B. thuringiensis:
Markerless gene deletion systems:
As demonstrated for leuB in B. thuringiensis, where a conditionally asporogenous recombinant strain was constructed
Employs counter-selectable markers (such as sacB conferring sucrose sensitivity)
Requires a two-step selection process allowing marker removal after confirmation of the desired modification
CRISPR-Cas9 genome editing:
Cre-lox recombination system:
Integration of loxP sites flanking both the pckA target region and selection marker
Transient expression of Cre recombinase to excise the marker
Verification of marker removal via PCR and phenotypic testing
Recombinant expression strategies:
For partial pckA expression, shuttle vectors like pHT3101 can be used under control of sporulation-specific promoters
After transformation by electroporation (20 KV/cm in a 0.2 cm-gap cuvette), transformants can be selected
Stability verification through subculturing for multiple generations is essential
Each approach has specific advantages depending on the intended modification (point mutations vs. deletions) and strain characteristics. Transformants must be verified for both integration and stable inheritance through multiple generations to ensure experimental reliability.
Designing robust experiments to study pckA function requires attention to several critical factors:
Genetic modification strategy:
Consider whether complete deletion, point mutation, or regulated expression is most appropriate
Include complementation controls to verify phenotypes are specifically due to pckA modification
Design constructs that maintain genomic context and native regulation where possible
Growth condition selection:
Temporal sampling design:
Sample across all growth phases (lag, exponential, transition, stationary, sporulation)
For sporulation studies, synchronize cultures to reduce heterogeneity
Include both short-term (minutes to hours) and long-term (days) analyses
Multi-parameter analysis:
Combine transcriptomics, proteomics, and metabolomics approaches
Include enzyme activity measurements to correlate gene expression with function
Monitor physiological parameters (growth rate, sporulation efficiency, crystal protein production)
Control implementation:
Wild-type strain grown under identical conditions
Empty vector controls for plasmid-based expression systems
Complemented mutant strains to verify phenotype restoration
Unrelated metabolic gene mutants to distinguish specific from general metabolic effects
Data integration planning:
Design experiments to allow statistical correlation between multiple data types
Include sufficient replication (minimum three biological replicates)
Implement appropriate statistical design (randomization, blocking for batch effects)
These considerations help ensure that experimental outcomes can be reliably attributed to pckA function rather than to secondary effects or experimental artifacts.
Optimizing growth conditions and media compositions is essential for meaningful pckA expression studies:
Recommended media formulations:
Carbon source selection:
For pckA induction: Succinate, malate, or pyruvate (gluconeogenic conditions)
For pckA repression: Glucose (when glycolytic pathways are predominant)
For differential analysis: Both glucose and a TCA cycle intermediate
Critical growth parameters:
Specialized conditions for specific analyses:
For sporulation studies: Nutrient depletion to synchronize development
For stress response analysis: Sub-lethal concentrations of osmotic (NaCl), oxidative (H₂O₂), or temperature stressors
For virulence studies: Host-mimicking conditions (serum supplementation, microaerobic conditions)
Sampling timing:
For pckA transcriptional studies: Multiple time points spanning growth phases
For protein studies: Mid to late exponential phase and early stationary phase
For sporulation effects: Regular intervals from early stationary phase through spore maturation
These conditions should be optimized for each specific strain, as genetic background can significantly influence optimal growth parameters and expression patterns.
Measuring pckA enzyme activity in B. thuringiensis requires careful attention to extraction conditions and assay parameters:
Cell extract preparation:
Harvest cells at desired growth phase (typically late exponential)
Wash cells with cold buffer to remove media components
Resuspend in extraction buffer: 50 mM HEPES (pH 7.5), 10 mM MgCl₂, 1 mM EDTA, 5 mM DTT, protease inhibitors
Disrupt cells by sonication or French press (keeping samples on ice)
Clarify by centrifugation (15,000 × g, 30 min, 4°C)
Assay immediately or store at -80°C with glycerol
Spectrophotometric coupled enzyme assay:
Principle: Measure PEP formation from oxaloacetate by coupling to pyruvate kinase and lactate dehydrogenase, tracking NADH oxidation
Reaction mixture: 100 mM HEPES (pH 7.5), 10 mM MgCl₂, 10 mM MnCl₂, 2 mM ATP, 2 mM oxaloacetate, 0.15 mM NADH, 5 U pyruvate kinase, 5 U lactate dehydrogenase
Monitor decrease in absorbance at 340 nm (ε = 6,220 M⁻¹cm⁻¹)
Calculate activity as μmol NADH oxidized min⁻¹ mg⁻¹ protein
Direct assay for reverse reaction:
Principle: Measure oxaloacetate formation from PEP and bicarbonate
Reaction mixture: 100 mM HEPES (pH 7.5), 10 mM MgCl₂, 2 mM PEP, 20 mM KHCO₃, 2 mM ADP
Couple to malate dehydrogenase reduction of oxaloacetate with NADH
Monitor decrease in absorbance at 340 nm
qRT-PCR correlation:
While not a direct measure of enzyme activity, qRT-PCR of pckA can be correlated with enzyme activity measurements
Design primers specific to pckA as done for other metabolic genes in B. thuringiensis
Normalize to validated reference genes (such as 16S rRNA)
Compare expression patterns with protein levels and activity measurements
Western blot analysis:
Use pckA-specific antibodies to quantify protein levels
Compare with enzyme activity to assess post-translational regulation
Include recombinant pckA standards for quantification
Critical controls:
No-substrate control to measure background NADH oxidation
Heat-inactivated enzyme control
Specific inhibitor control (3-mercaptopicolinic acid)
Wild-type extracts as positive control
This multi-method approach provides comprehensive characterization of pckA activity, enabling researchers to distinguish between transcriptional, translational, and post-translational regulation.
When faced with contradictory results between transcriptomic and proteomic data for pckA, researchers should implement a systematic analytical approach:
Technical validation:
Confirm primer specificity for qRT-PCR through melt curve analysis and sequencing
Verify peptide uniqueness for proteomics through database searches
Perform technical replicates to assess measurement variability
Use alternative methods (Northern blots for RNA, Western blots for protein) to confirm findings
Temporal dynamics analysis:
Consider time delays between transcription and translation
Implement time-course experiments with frequent sampling
Plot RNA and protein levels on the same timeline to identify lag periods
Examine the stability of both mRNA and protein under experimental conditions
Post-transcriptional regulation investigation:
Analyze mRNA secondary structures affecting translation efficiency
Consider the role of small RNAs or antisense transcripts
Examine ribosome binding site accessibility
Post-translational modification assessment:
Investigate potential protein modifications affecting stability or activity
Consider protein compartmentalization affecting extraction efficiency
Examine protein turnover rates through pulse-chase experiments
Integration strategies:
Apply pathway analysis to identify regulatory patterns
Use correlation networks to find co-regulated genes
Implement mathematical models that account for regulatory delays
Consider the functional implications through enzyme activity assays
In some experimental systems, good correlation between mRNA and protein levels can be achieved, as demonstrated in research showing "the expression patterns of mRNA were consistent with those of protein" for B. thuringiensis metabolic enzymes . When discordance persists, it should be viewed as biologically informative rather than problematic, potentially revealing novel regulatory mechanisms affecting pckA.
Analyzing pckA expression during B. thuringiensis sporulation requires sophisticated statistical approaches:
Time series analysis methods:
Smoothing techniques (LOESS) to reduce experimental noise
Change-point detection to identify significant transitions in expression
Autocorrelation analysis to identify cyclical patterns
Differential expression analysis:
ANOVA with post-hoc tests for multi-timepoint comparisons
Linear mixed effects models to account for repeated measures
FDR correction for multiple testing (Benjamini-Hochberg procedure)
Multivariate approaches:
Principal Component Analysis (PCA) to identify major sources of variation
Clustering methods (hierarchical, k-means) to group co-expressed genes
Heat maps with hierarchical clustering for visualization
Correlation analysis:
Pearson or Spearman correlation between pckA and other genes
Time-lagged correlation to identify potential regulatory relationships
Partial correlation to control for confounding variables
Expression pattern classification:
Compare pckA expression patterns to known sporulation regulators
Classify based on similarity to reference patterns (early, middle, late sporulation genes)
Evaluate coherence with other metabolic genes
Data integration strategies:
Correlate gene expression with phenotypic measurements (sporulation rate, enzyme activity)
Integrate transcriptomic, proteomic, and metabolomic data
Implement network analysis to place pckA in broader regulatory context
Specialized sporulation analysis:
Compare with expression profiles of known sporulation genes (spo genes, sigma factors)
Assess correlation with morphological changes
Evaluate temporal coordination with spore-specific metabolite production
These statistical approaches help distinguish meaningful biological changes from experimental variation and place pckA expression changes within the broader context of the sporulation process.
Distinguishing direct pckA effects from adaptive responses requires a multi-faceted experimental strategy:
Temporal resolution approaches:
Implement immediate sampling after inducible pckA modification
Use metabolic quenching techniques to capture instant metabolic states
Track metabolic flux changes over short time intervals (minutes to hours)
Compare with long-term adaptation patterns (days to weeks)
Genetic complementation strategies:
Create a complemented ΔpckA strain expressing wild-type pckA
Develop point mutant variants with altered catalytic properties
Use inducible expression systems with varying expression levels
Compare phenotypes between deletion, complementation, and overexpression
Metabolic network analysis:
Measure direct substrates and products of the pckA reaction
Assess changes in connected metabolic pathways
Implement 13C metabolic flux analysis to track carbon flow
Compare experimental results with metabolic model predictions
Multi-strain comparative analysis:
Create multiple independent pckA mutants to identify consistent effects
Compare with mutations in other gluconeogenic enzymes
Analyze strains adapted to growth without pckA for compensatory mechanisms
Regulatory network investigation:
Examine expression changes in known metabolic regulators
Identify co-regulated genes through transcriptome analysis
Map potential regulatory interactions affecting pckA expression
In vivo validation techniques:
Test phenotypes under multiple growth conditions
Assess fitness in competition experiments
Evaluate performance in relevant biological contexts (sporulation, virulence)
Through this comprehensive approach, researchers can build a causal model distinguishing primary effects of pckA modification from secondary adaptations, similar to the metabolic analysis approach used to characterize the role of leuB in B. thuringiensis sporulation .
Creating a recombinant B. thuringiensis strain with modified pckA involves a series of precise molecular and microbiological steps:
Materials needed:
B. thuringiensis subsp. konkukian culture
Appropriate shuttle vector (such as pHT3101 mentioned in result )
PCR reagents and primers specific for pckA
Restriction enzymes and DNA ligase
Transformation reagents (electroporation cuvettes, recovery media)
Selection media with appropriate antibiotics
Protocol steps:
Gene fragment amplification and construct preparation:
Design primers with appropriate restriction sites to amplify pckA with desired modifications
Amplify the pckA gene region from B. thuringiensis genomic DNA
Digest the PCR product and vector with appropriate restriction enzymes
Ligate the digested pckA fragment into the vector
Transform into E. coli for construct verification
B. thuringiensis transformation:
Transformant verification:
Expression verification:
Phenotypic characterization:
Compare growth curves between wild-type and recombinant strains
Assess metabolic profiles using appropriate assays
Evaluate sporulation efficiency and crystal protein production if relevant
This protocol can be adapted for different types of modifications, including gene deletion, point mutations, or expression of recombinant variants. For applications involving recombinant DNA and potentially pathogenic strains, appropriate biosafety considerations and institutional approvals should be obtained .
Assessing the impact of pckA modification on B. thuringiensis virulence requires multiple complementary approaches:
In vitro virulence factor production:
Insect bioassays:
Conduct dose-response studies with target insects
Determine LC50 (median lethal concentration) and LT50 (median lethal time)
Compare wild-type and pckA-modified strains at multiple concentrations
Cellular infection models:
Use insect cell lines to measure cytotoxicity
Assess bacterial adherence, invasion, and intracellular survival
Measure host cell cytokine/antimicrobial peptide responses
Evaluate resistance to cellular defense mechanisms
Molecular virulence assessment:
In vivo infection models:
Competitive index studies:
Co-infect hosts with wild-type and pckA-modified strains
Determine relative fitness during infection
Use differentially marked strains for selection and quantification
Virulence factor complementation:
These methods collectively provide a comprehensive assessment of how pckA modification affects B. thuringiensis virulence through both direct metabolic effects and potential regulatory impacts on virulence factor production.
Monitoring the metabolic consequences of pckA modification requires a multi-faceted approach:
Growth phenotype characterization:
Measure growth rates on different carbon sources
Assess metabolic flexibility through carbon source utilization profiles
Determine biomass yield coefficients under various growth conditions
Evaluate stress tolerance (temperature, pH, osmotic pressure)
Metabolite analysis:
Targeted metabolomics focusing on TCA cycle intermediates and gluconeogenic precursors
Untargeted metabolomics to identify unexpected metabolic changes
Intracellular metabolite extraction using cold methanol quenching
Quantification by LC-MS/MS or GC-MS methods
Isotope tracer studies:
13C-labeled substrate feeding experiments
Metabolic flux analysis to quantify carbon flow through central metabolism
Positional isotopomer analysis to determine pathway utilization
Dynamic labeling studies to assess metabolic turnover rates
Enzyme activity measurements:
Assay activities of key enzymes in connected pathways
Monitor regulatory enzyme activities under different conditions
Compare in vitro enzyme kinetics between wild-type and modified strains
Measure allosteric regulation patterns of metabolic enzymes
Global expression analysis:
Transcriptomics (RNA-seq) to identify compensatory gene expression changes
Proteomics to assess protein-level adaptations
Phosphoproteomics to identify changes in metabolic regulation
Integration of multi-omics data for comprehensive metabolic modeling
Physiological response evaluation:
Computational metabolic analysis:
Constraint-based metabolic modeling (flux balance analysis)
Metabolic control analysis to identify regulatory points
Network analysis to identify altered pathway utilization
Comparison with existing models of B. thuringiensis metabolism
This comprehensive monitoring approach enables researchers to fully characterize how pckA modification ripples through the metabolic network of B. thuringiensis, affecting both core metabolism and specialized functions like sporulation and virulence factor production.