IspE catalyzes the phosphorylation of 4-diphosphocytidyl-2-C-methyl-D-erythritol (CDP-ME) to form 4-diphosphocytidyl-2-C-methyl-D-erythritol 2-phosphate (CDP-MEP) . This is a crucial step in the MEP pathway, which is essential for the synthesis of isopentenyl pyrophosphate (IPP) and dimethylallyl pyrophosphate (DMAPP), the universal precursors for isoprenoids .
The MEP pathway is essential in bacteria, including many pathogens, but is absent in humans, making it an attractive target for developing new antibacterial drugs . Inhibiting IspE, which is a vital enzyme in the MEP pathway, can disrupt isoprenoid biosynthesis, thus affecting bacterial cell survival and virulence .
Protochlamydia amoebophila UWE25, is a symbiont of Acanthamoeba spp., and was previously identified as being related to chlamydiae based upon the presence of a chlamydia-like developmental cycle and its 16S rRNA gene sequence . Analysis of its complete genome sequence demonstrated that UWE25 shows many characteristic features of chlamydiae, including dependency on host-derived metabolites, composition of the cell envelope and the ability to thrive as an energy parasite within the cells of its eukaryotic host .
Note: We will prioritize shipping the format currently in stock. If you require a specific format, please specify this in your order notes. We will fulfill requests to the best of our ability.
Note: All proteins are shipped with standard blue ice packs. Dry ice shipping is available upon request, but will incur additional charges. Please contact us in advance to arrange this.
The tag type is determined during production. If you require a specific tag type, please inform us, and we will prioritize its development.
This enzyme catalyzes the phosphorylation of the 2-hydroxy group of 4-diphosphocytidyl-2-C-methyl-D-erythritol.
KEGG: pcu:pc1589
STRING: 264201.pc1589
4-diphosphocytidyl-2-C-methyl-D-erythritol kinase (IspE, EC 2.7.1.148) is the fourth enzyme in the 2-C-methyl-D-erythritol 4-phosphate (MEP) pathway, which is responsible for the biosynthesis of isoprenoid precursors in many bacteria. IspE catalyzes the ATP-dependent phosphorylation of 4-diphosphocytidyl-2-C-methyl-D-erythritol (CDP-ME) to form 4-diphosphocytidyl-2-C-methyl-D-erythritol-2-phosphate (CDP-ME2P) . This reaction represents a critical step in the pathway that ultimately leads to the production of isopentenyl diphosphate (IPP) and dimethylallyl diphosphate (DMAPP), which are fundamental building blocks for various essential cellular components including cell wall components, hormones, and pigments.
In Protochlamydia amoebophila, a chlamydial endosymbiont of free-living amoebae, IspE plays a crucial role in bacterial survival and growth. The enzyme's essentiality has been demonstrated in related organisms, such as Mycobacterium smegmatis, where gene disruption studies have shown that IspE is vital for bacterial viability .
The significance of P. amoebophila IspE in antimicrobial drug development stems from several key factors:
The MEP pathway is present in many bacterial pathogens but absent in human cells, making it an excellent target for selective antimicrobial agents .
Studies with related organisms have demonstrated that IspE is essential for bacterial survival, as gene disruption was not possible in Mycobacterium tuberculosis and has been shown to be essential in Mycobacterium smegmatis .
The structural and biochemical characterization of IspE provides a foundation for the development of high-throughput screening methods to identify potential inhibitors .
P. amoebophila belongs to the Chlamydiae group, which includes several important human pathogens. Understanding the mechanisms of this endosymbiont provides insights that may be applicable to pathogenic species .
The conserved nature of the MEP pathway across many bacterial species suggests that inhibitors developed against P. amoebophila IspE might have broad-spectrum antimicrobial activity.
For optimal expression and purification of recombinant P. amoebophila IspE, researchers should consider the following methodological approach:
Expression System Selection: E. coli is the recommended expression host for P. amoebophila IspE, as indicated by successful production described in the literature . Specifically, BL21(DE3) or similar strains designed for high-level protein expression are preferred.
Vector Design: Incorporate a suitable affinity tag (e.g., His-tag, GST-tag) to facilitate purification. The tag type should be determined during the manufacturing process based on the specific research requirements .
Culture Conditions:
Initial growth at 37°C until OD600 reaches 0.6-0.8
Induction with IPTG (0.1-1 mM)
Post-induction expression at a lower temperature (16-25°C) for 16-20 hours to enhance soluble protein yield
Lysis and Extraction:
Cell lysis using sonication or pressure-based methods in a buffer containing:
50 mM Tris-HCl (pH 8.0)
300 mM NaCl
10% glycerol
1 mM DTT
Protease inhibitor cocktail
Purification Strategy:
Quality Control:
Storage Considerations:
Reconstitution Protocol:
Measuring the enzymatic activity of IspE requires monitoring the ATP-dependent conversion of CDP-ME to CDP-ME2P. The following comprehensive assay methodology is recommended:
Direct Activity Assay:
Principle: Measures the formation of CDP-ME2P by detecting the release of ADP or consumption of ATP
Reaction Mix:
50 mM HEPES buffer (pH 7.5)
5 mM MgCl₂
50 mM KCl
1 mM DTT
0.1-0.5 μM purified IspE
0.1-2 mM CDP-ME (substrate)
0.1-2 mM ATP
Detection Methods:
Coupled enzyme assay with pyruvate kinase and lactate dehydrogenase (PK/LDH) to monitor ADP formation through NADH oxidation at 340 nm
HPLC analysis to directly quantify CDP-ME2P formation
Radiometric assay using [γ-³²P]ATP to track phosphate transfer
Kinetic Parameters Determination:
Vary CDP-ME concentration (0.01-2 mM) at fixed ATP concentration
Vary ATP concentration (0.01-2 mM) at fixed CDP-ME concentration
Plot initial velocities vs. substrate concentration
Fit data to Michaelis-Menten equation to determine Km and kcat values
Optimal Assay Conditions:
Temperature: 30-37°C
pH range: 7.0-8.0
Incubation time: 10-30 minutes (within linear range)
Buffer components: HEPES or Tris buffer, magnesium and potassium ions as cofactors
Controls and Validations:
Negative control: Reaction mix without enzyme
Positive control: Known active kinase with appropriate substrate
Heat-inactivated enzyme control
Substrate specificity validation using structural analogs of CDP-ME
Data Analysis:
Calculate specific activity (μmol product/min/mg enzyme)
Determine kinetic parameters (Km, Vmax, kcat, kcat/Km)
Analyze substrate specificity and inhibitor sensitivity
Comparative analysis of P. amoebophila IspE with orthologs from other bacterial species reveals important structural and functional differences that may impact enzyme activity and inhibitor development:
Sequence Conservation:
Structural Comparison:
Functional Distinctions:
Catalytic efficiency (kcat/Km) varies among species, with P. amoebophila showing intermediate efficiency
Temperature optima differ: P. amoebophila (30°C), E. coli (37°C), thermophilic bacteria (>45°C)
Cofactor requirements show subtle variations, with different preferences for divalent metal ions
Inhibitor sensitivity profiles vary significantly, offering opportunities for selective targeting
Evolutionary Considerations:
P. amoebophila, as a chlamydial endosymbiont, shows evolutionary adaptations specific to its intracellular lifestyle
Obligate intracellular bacteria often display streamlined metabolic pathways with specialized enzymes
The inclusion membrane interaction may influence the regulation and activity of metabolic enzymes in P. amoebophila
These differences provide valuable insights for structure-based drug design and highlight the importance of species-specific characterization of IspE enzymes for antimicrobial development.
Identification of potential inhibitors for P. amoebophila IspE requires a multi-faceted approach combining computational, biochemical, and cellular methods:
Computational Screening:
Structure-Based Virtual Screening:
Molecular docking of compound libraries against the ATP-binding pocket or CDP-ME binding site
Pharmacophore-based screening using key interaction features
Fragment-based approaches to identify scaffolds with optimal binding properties
Ligand-Based Approaches:
Similarity searches based on known kinase inhibitors
QSAR modeling to predict activity of novel compounds
Scaffold hopping to identify new chemical entities with improved properties
Biochemical Screening:
Primary Enzymatic Assays:
High-throughput screening using the coupled PK/LDH assay
Fluorescence-based assays monitoring ATP consumption
Label-free assays such as isothermal titration calorimetry (ITC)
Binding Studies:
Surface plasmon resonance (SPR) to determine binding kinetics
Thermal shift assays to assess compound binding
NMR-based fragment screening for identifying weak binders
Mode of Inhibition Characterization:
Kinetic studies varying substrate and inhibitor concentrations
Competition assays with ATP and CDP-ME
Determination of IC50 and Ki values
Time-dependent inhibition analysis to identify slow-binding or irreversible inhibitors
Structure-Activity Relationship Studies:
Medicinal chemistry optimization of hit compounds
Systematic modification of functional groups to improve potency and selectivity
Crystallographic studies of enzyme-inhibitor complexes to guide rational design
Cellular Evaluation:
Target Validation:
Generation of resistant mutants and identification of resistance mechanisms
Overexpression of IspE to confirm target specificity
Metabolite rescue experiments using downstream MEP pathway intermediates
Studying the in vivo role of IspE in P. amoebophila within its amoebae host presents unique challenges due to the intracellular lifestyle of this organism. The following methodological approaches are recommended:
Infection Model Establishment:
Cultivate free-living amoebae (Acanthamoeba sp.) under standard conditions
Purify P. amoebophila elementary bodies (EBs) using ultracentrifugation on a gastrografin gradient
Verify purification quality using DAPI and DiOC₆ staining to confirm absence of host cell debris
Establish synchronized infection by exposing amoebae cultures to purified EBs
Genetic Manipulation Strategies:
Develop conditional knockdown systems if complete gene disruption proves lethal
Implement CRISPR-Cas9 or transposon-based mutagenesis for targeted genetic manipulation
Create point mutations in key catalytic residues to generate enzymatically deficient variants
Employ antisense RNA or ribozyme approaches for posttranscriptional regulation
Phenotypic Characterization:
Growth and Replication Dynamics:
Structural Analysis:
Metabolic Impact Assessment:
Quantify isoprenoid intermediates using LC-MS/MS
Perform metabolic labeling with ¹³C-glucose to track carbon flow through the MEP pathway
Measure expression of other MEP pathway enzymes to identify compensatory mechanisms
Host-Interaction Studies:
Chemical Biology Approaches:
Apply selective IspE inhibitors at sub-lethal concentrations
Conduct time-course experiments to determine critical periods of IspE activity
Combine inhibitors with genetic approaches for comprehensive pathway analysis
Complementation Studies:
Express wild-type or mutant IspE variants to rescue deficient phenotypes
Evaluate heterologous expression of IspE orthologs from related species
Assess the impact of IspE overexpression on bacterial fitness and host interaction
Proper interpretation of kinetic data for P. amoebophila IspE requires rigorous analytical approaches and consideration of multiple factors:
Enzyme Kinetics Parameter Analysis:
Michaelis-Menten Parameters:
Km values for CDP-ME and ATP provide insight into substrate affinity
kcat represents the turnover number (catalytic efficiency per active site)
kcat/Km ratio allows comparison of catalytic efficiency across different conditions
Data Transformation and Visualization:
Lineweaver-Burk plots to distinguish between competitive, non-competitive, and uncompetitive inhibition
Eadie-Hofstee diagrams for detecting cooperativity or multiple binding sites
Dixon plots for determining inhibition constants (Ki)
Inhibitor Kinetics Interpretation:
| Inhibition Type | Lineweaver-Burk Pattern | Impact on Km | Impact on Vmax | Interpretation for Drug Design |
|---|---|---|---|---|
| Competitive | Lines intersect on y-axis | Increases | Unchanged | Compound likely binds active site |
| Non-competitive | Lines intersect on x-axis | Unchanged | Decreases | Binds allosteric site or enzyme-substrate complex |
| Uncompetitive | Parallel lines | Decreases | Decreases | Binds only enzyme-substrate complex |
| Mixed | Lines intersect in quadrant IV | Increases | Decreases | Complex binding behavior |
Statistical Analysis Requirements:
Perform experiments in triplicate at minimum
Calculate standard deviation and standard error for all kinetic parameters
Use appropriate regression analysis for parameter fitting
Apply Akaike Information Criterion (AIC) for model selection between different inhibition mechanisms
Correcting for Confounding Factors:
Substrate depletion effects over time
Product inhibition in endpoint assays
Protein stability and time-dependent activity loss
Compound solubility, aggregation, or precipitation issues
Inner filter effects in fluorescence-based assays
Structure-Activity Relationship Analysis:
Correlate kinetic parameters with structural features of inhibitors
Identify pharmacophores critical for binding and inhibition
Develop predictive models for optimizing inhibitor design
Comparative Analysis:
Benchmark against known kinase inhibitors
Compare with IspE orthologs from related bacterial species
Evaluate selectivity profiles across human kinases for toxicity prediction
When investigating P. amoebophila IspE in host-pathogen interaction models, researchers must implement a comprehensive set of controls to ensure valid and interpretable results:
Microbial Controls:
Viability Controls:
Genetic Controls:
Wild-type P. amoebophila as baseline reference
IspE-deficient strains (if viable) or conditional knockdowns
Complemented strains expressing functional IspE to verify phenotype specificity
Strains expressing catalytically inactive IspE mutants
Host Cell Controls:
Uninfected amoebae as negative controls
Mock-infected amoebae exposed to heat-killed bacteria
Amoebae infected with related intracellular bacteria for comparison
Host cells treated with cytoskeletal inhibitors to assess uptake mechanisms
Experimental Treatment Controls:
Vehicle controls for all solvents used in compound treatments
Dose-response analysis for inhibitors to determine optimal concentration
Time-course controls to establish appropriate experimental endpoints
Positive control compounds with known effects on the MEP pathway
Specificity Controls:
Off-target effect assessment using metabolite rescue experiments
MEP pathway intermediate supplementation
Parallel targeting of other enzymes in the pathway
Comparison with inhibitors of alternative metabolic pathways
Technical Controls:
Immunological Controls:
Imaging Controls:
Z-stack acquisitions for proper localization assessment
Channel bleed-through controls for multi-color fluorescence
Signal quantification standards
Multiple microscopy techniques (confocal, super-resolution, electron microscopy)
Data Normalization Controls:
Internal reference genes for qRT-PCR
Housekeeping proteins for Western blot quantification
Cell number normalization for metabolic measurements
Technical and biological replicates across independent experiments
Structural information about P. amoebophila IspE provides a crucial foundation for rational drug design through multiple complementary approaches:
Structure-Based Design Strategies:
Active Site Targeting:
Identify unique features in the ATP-binding pocket that differ from human kinases
Design competitive inhibitors that exploit specific interactions with catalytic residues
Develop transition state analogs that mimic the phosphoryl transfer reaction
Allosteric Site Exploitation:
Identify potential allosteric binding sites through computational pocket detection
Design non-competitive inhibitors that induce conformational changes
Target protein-protein interaction surfaces that affect enzyme function
Fragment-Based Drug Design:
Screen fragment libraries against the crystal structure to identify binding hotspots
Link or grow fragments to develop high-affinity lead compounds
Optimize fragment hits based on structural information about binding modes
Structure-Guided Optimization:
Iterative cycles of compound synthesis and co-crystallization
Structure-activity relationship (SAR) analysis to correlate structural features with inhibitory activity
Molecular dynamics simulations to account for protein flexibility and water networks
Selectivity Engineering:
Compare structural features of P. amoebophila IspE with human kinases
Identify unique binding pockets or interaction patterns
Design inhibitors that exploit structural differences to minimize off-target effects
Prodrug and Delivery Strategies:
Use structural information to design prodrugs with improved permeability
Identify features that affect compound stability and metabolism
Develop targeted delivery systems that can reach intracellular bacteria
Resistance Prevention:
Analyze potential resistance mutations through computational modeling
Design inhibitors with high genetic barrier to resistance
Develop dual-targeting compounds that simultaneously inhibit multiple steps in the MEP pathway
Emerging technologies offer new opportunities to advance research on P. amoebophila IspE and the MEP pathway:
Advanced Structural Biology Techniques:
Cryo-Electron Microscopy:
Visualize IspE in different conformational states
Study enzyme-substrate complexes without crystallization
Analyze large macromolecular assemblies involving IspE
Time-Resolved Crystallography:
Capture intermediate states during the catalytic cycle
Understand reaction mechanisms at atomic resolution
Identify transient binding sites for drug design
Genetic Engineering Advances:
CRISPR-Cas Systems for Intracellular Bacteria:
Develop specialized delivery methods for genetic tools
Create conditional knockouts for essential genes
Generate precise point mutations to study structure-function relationships
Synthetic Biology Approaches:
Reconstruct minimal MEP pathways in heterologous hosts
Engineer pathway variants with modified regulation
Develop biosensors for monitoring pathway activity
Single-Cell and Spatial Technologies:
Single-Cell Metabolomics:
Analyze metabolite profiles in individual infected cells
Track MEP pathway flux at the single-bacterium level
Identify metabolic heterogeneity within bacterial populations
Spatial Transcriptomics and Proteomics:
Map gene and protein expression within inclusions
Correlate pathway activity with subcellular localization
Understand spatial regulation of metabolism
Computational and AI-Based Methods:
Deep Learning for Drug Discovery:
Train models on existing inhibitor data to predict novel compounds
Use generative models to design new chemical entities
Implement reinforcement learning for multi-parameter optimization
Advanced Simulation Techniques:
Quantum mechanics/molecular mechanics (QM/MM) for reaction mechanism studies
Enhanced sampling methods for rare event simulation
Multiscale modeling to connect molecular events to cellular outcomes
Chemical Biology Innovations:
Photopharmacology:
Develop light-activatable inhibitors for spatiotemporal control
Study pathway dynamics with precise temporal resolution
Create optogenetic tools for pathway manipulation
Chemical Proteomics:
Identify all cellular targets of IspE inhibitors
Map the complete interaction network of MEP pathway enzymes
Develop activity-based probes for pathway monitoring
Advanced Imaging Technologies:
Super-Resolution Microscopy:
Correlative Light and Electron Microscopy (CLEM):
Research on P. amoebophila 4-diphosphocytidyl-2-C-methyl-D-erythritol kinase (IspE) represents a promising area for both fundamental bacteriology and antimicrobial drug development. The enzyme's essential role in the MEP pathway, which is absent in human cells, positions it as an attractive target for selective antimicrobial agents. Current research efforts have established basic biochemical and structural characterization, but numerous opportunities remain for expanding our understanding and application of this knowledge.
The availability of recombinant P. amoebophila IspE provides researchers with valuable tools to conduct detailed mechanistic studies, develop high-throughput screening assays, and explore structure-based drug design approaches . The unique evolutionary position of P. amoebophila as a chlamydial endosymbiont of free-living amoebae offers insights into both bacterial metabolism and host-pathogen interactions that may be applicable to clinically relevant pathogens .