KEGG: pcu:pc0662
STRING: 264201.pc0662
Protochlamydia amoebophila UWE25 is an obligate intracellular symbiont belonging to the Chlamydiales order that thrives within the protozoan host Acanthamoeba sp . Unlike its pathogenic relatives in the Chlamydiaceae family (which cause human diseases), P. amoebophila represents an evolutionary important organism for understanding chlamydial biology.
The guanylate kinase (gmk) from P. amoebophila is significant for several reasons:
It plays an essential role in nucleotide metabolism, catalyzing the phosphorylation of GMP to GDP, which serves as a precursor for GTP synthesis
P. amoebophila lacks complete de novo nucleotide synthesis pathways, making nucleotide metabolism enzymes critical for survival
Studying gmk provides insights into the metabolic adaptation of obligate intracellular bacteria to their host environments
Comparative analysis with gmk from pathogenic chlamydiae can reveal evolutionary adaptations and potential antimicrobial targets
Guanylate kinase functions within a critically important pathway in P. amoebophila's metabolic network:
P. amoebophila cannot synthesize purine nucleotides de novo due to genomic reduction associated with its intracellular lifestyle
The organism depends on host-derived GMP, which is phosphorylated by gmk to produce GDP
GDP is subsequently converted to GTP by nucleoside-diphosphate kinase (ndk), also encoded in the P. amoebophila genome
GTP serves multiple essential functions:
Energy source for protein synthesis
Substrate for RNA synthesis
Precursor for dGTP used in DNA synthesis
Signaling molecule
This metabolic framework explains why gmk is an essential enzyme for P. amoebophila survival. The pathways are further integrated with the host cell metabolism through specialized nucleotide transport proteins, particularly the nucleotide transporter (NTT) family members that import nucleotides from the host .
Based on methodologies used for similar proteins, E. coli expression systems are most effective for recombinant P. amoebophila gmk production. The experimental procedure typically follows:
Gene amplification from P. amoebophila genomic DNA using PCR with primers containing appropriate restriction sites (e.g., XhoI and BamHI)
Cloning into an expression vector such as pET16b (Novagen) which provides:
Transformation into an E. coli expression strain:
BL21(DE3) for standard expression
Rosetta or Origami strains if codon usage or disulfide bond formation is problematic
Expression optimization typically requires testing multiple conditions:
Temperature (16°C, 25°C, 37°C)
IPTG concentration (0.1-1.0 mM)
Duration of induction (4-16 hours)
Media composition (LB, TB, or minimal media with supplements)
For optimal purification of enzymatically active recombinant P. amoebophila gmk, the following strategy has proven effective for similar enzymes:
Cell lysis:
Resuspend cells in buffer containing 50 mM Tris-HCl pH 7.5, 300 mM NaCl, 10 mM imidazole
Add protease inhibitors (e.g., PMSF or commercial cocktail)
Lyse using sonication or French press
Clarify lysate by centrifugation (20,000 × g, 30 min, 4°C)
Immobilized metal affinity chromatography (IMAC):
Load clarified lysate onto Ni-NTA column
Wash with buffer containing 20-30 mM imidazole
Elute with buffer containing 250-300 mM imidazole
Size exclusion chromatography:
Further purify using Superdex 75 or 200 column
Use buffer containing 50 mM Tris-HCl pH 7.5, 150 mM NaCl, 5 mM MgCl₂
Analyze fractions by SDS-PAGE
Quality control:
Assess purity by SDS-PAGE (>95% purity recommended)
Verify identity by Western blot or mass spectrometry
Confirm activity using enzyme assays (see section 3.1)
Determine protein concentration using Bradford assay or A₂₈₀ measurement
This protocol typically yields 5-10 mg of pure, active enzyme per liter of bacterial culture.
For accurate determination of kinetic parameters of recombinant P. amoebophila gmk, several complementary approaches can be employed:
Coupled enzyme assay:
Measure ADP production using pyruvate kinase and lactate dehydrogenase
Monitor NADH oxidation at 340 nm
Reaction mixture: 50 mM Tris-HCl pH 7.5, 50 mM KCl, 5 mM MgCl₂, 1 mM phosphoenolpyruvate, 0.2 mM NADH, 2 units pyruvate kinase, 2 units lactate dehydrogenase, varying GMP concentrations (5-500 μM), fixed ATP (4 mM), and purified gmk
Direct measurement of product formation by HPLC:
Separate nucleotides on reverse-phase or ion-exchange column
Monitor at 260 nm
Quantify GDP formation over time at different substrate concentrations
Radiochemical assay:
Use [α-³²P]-GMP as substrate
Separate products by thin-layer chromatography
Quantify radioactive products using phosphorimager
For data analysis:
Plot initial velocity versus substrate concentration
Fit data to Michaelis-Menten equation to determine Km and Vmax
Calculate kcat by dividing Vmax by enzyme concentration
For inhibition studies, determine Ki using appropriate inhibition models
Based on data from similar bacterial gmk enzymes, expected kinetic parameters might fall within these ranges :
kcat: 20-70 sec⁻¹
Km (GMP): 20-150 μM
Km (ATP): 100-500 μM
While specific data on P. amoebophila gmk regulation by (p)ppGpp is not directly available, comparative analysis with other bacterial gmk enzymes provides valuable insights:
(p)ppGpp is a bacterial alarmone that mediates stress responses, and its interaction with gmk represents an important regulatory mechanism. In many bacteria, (p)ppGpp competitively inhibits gmk activity, preventing GMP conversion to GDP and subsequently limiting GTP synthesis during stress conditions .
The structural determinants for (p)ppGpp binding likely include:
Binding pocket residues: Conserved residues in the GMP binding site, particularly a tyrosine residue (equivalent to Tyr78 in S. aureus gmk) that contributes significantly to (p)ppGpp binding affinity
Mode of inhibition: Competitive inhibition with respect to GMP, indicating that (p)ppGpp binds to the GMP binding site
Phylogenetic patterns: The sensitivity to (p)ppGpp varies across bacterial phyla:
Since P. amoebophila is evolutionarily distinct from the well-characterized bacterial groups, determining its gmk sensitivity to (p)ppGpp would provide valuable information about the conservation of this regulatory mechanism across bacterial lineages.
Based on patterns observed in other bacteria, the inhibition constant (Ki) for P. amoebophila gmk could range from 1-60 μM for pppGpp, with potential variation between ppGpp and pppGpp .
For optimal characterization of P. amoebophila gmk activity, the following environmental parameters should be systematically evaluated:
pH dependence:
Test activity across pH range 6.0-9.0 using appropriate buffers
Most bacterial gmk enzymes show optimal activity at pH 7.0-8.0
Use overlapping buffers (MES, HEPES, Tris) to eliminate buffer-specific effects
Temperature effects:
Divalent cation requirements:
Gmk requires divalent cations for activity, typically Mg²⁺
Test various concentrations (1-20 mM) of Mg²⁺, Mn²⁺, Ca²⁺, and other divalent cations
Determine if monovalent cations (K⁺, Na⁺) affect activity
Stability conditions:
Evaluate enzyme stability in different storage buffers
Test effects of additives (glycerol, DTT, BSA) on long-term stability
Determine freeze-thaw stability
These parameters are critical for establishing reproducible assay conditions and for comparing P. amoebophila gmk properties with those of other bacterial gmk enzymes.
P. amoebophila gmk likely exhibits several differences from gmk enzymes in pathogenic Chlamydiaceae, reflecting their evolutionary divergence and adaptation to different host environments:
These differences could be exploited for developing selective inhibitors targeting pathogenic Chlamydiaceae without affecting related environmental species.
Phylogenetic analysis of gmk provides valuable insights into the evolution of nucleotide metabolism in Chlamydiales:
Conservation of essential function:
Adaptation to intracellular niche:
Co-evolution with nucleotide transporters:
Regulatory diversification:
Variations in gmk regulation across Chlamydiales lineages may reflect adaptation to different host environments
The (p)ppGpp regulatory system shows phylogenetic patterns that might extend to Chlamydiales
Horizontal gene transfer assessment:
Analysis of gmk sequences can help identify potential horizontal gene transfer events in the evolution of Chlamydiales
Unusual sequence features or unexpected phylogenetic placement would suggest horizontal acquisition
This evolutionary analysis provides context for understanding P. amoebophila gmk function and its role in the adaptation to an intracellular lifestyle.
Site-directed mutagenesis of recombinant P. amoebophila gmk can provide significant insights into structure-function relationships through strategic amino acid substitutions:
Catalytic site mutations:
Identify conserved residues involved in GMP binding by sequence alignment with characterized gmk enzymes
Create alanine substitutions of putative catalytic residues
Measure effects on Km and kcat to determine contribution to substrate binding and catalysis
(p)ppGpp sensitivity determinants:
Domain interface mutations:
If P. amoebophila gmk functions as an oligomer, mutate residues at subunit interfaces
Analyze effects on oligomerization and cooperative kinetics
Correlate structural changes with functional alterations
Substrate specificity engineering:
Design mutations to alter nucleotide specificity (e.g., modify GMP binding site to accept other nucleotides)
Test activity with non-canonical substrates
Evaluate evolutionary constraints on substrate specificity
A methodical mutagenesis strategy might include:
Sequence alignment to identify conserved and divergent residues
Homology modeling to predict structural impacts of mutations
Creation of single and multiple mutations using PCR-based methods
Parallel purification and characterization of multiple variants
Circular dichroism to verify proper folding of variants
Detailed kinetic analysis of each variant
For effective screening of potential P. amoebophila gmk inhibitors, a comprehensive strategy incorporating multiple approaches is recommended:
High-throughput primary screening:
Adapt the coupled enzyme assay to 96 or 384-well format
Screen diverse chemical libraries at single concentration (10-50 μM)
Define hit criteria (e.g., >50% inhibition)
Include positive controls (known nucleotide analogs) and negative controls
Dose-response confirmation:
Test hits in dose-response format (0.1-100 μM)
Determine IC₅₀ values and Hill coefficients
Eliminate compounds with poor dose-response relationships
Mechanism of action studies:
Determine inhibition mode (competitive, uncompetitive, noncompetitive)
Measure Ki values with respect to GMP and ATP
Evaluate time-dependence of inhibition
Selectivity assessment:
Test inhibitors against human gmk and gmk from related bacteria
Calculate selectivity indices
Prioritize compounds with high selectivity for P. amoebophila gmk
Structure-activity relationship analysis:
Group inhibitors by chemical scaffold
Identify pharmacophore features correlated with activity
Guide synthetic optimization of promising leads
Advanced validation methods:
Thermal shift assays to confirm direct binding
Surface plasmon resonance to measure binding kinetics
Crystallography of enzyme-inhibitor complexes (if feasible)
This systematic approach enables identification of potent, selective inhibitors with well-characterized mechanisms of action, which could serve as chemical probes for studying P. amoebophila metabolism or as starting points for antimicrobial development.
Addressing stability challenges with recombinant P. amoebophila gmk requires a multi-faceted approach:
Expression optimization:
Lower induction temperature (16-20°C) to slow folding and prevent aggregation
Reduce IPTG concentration (0.1-0.2 mM) to decrease expression rate
Co-express with molecular chaperones (GroEL/ES, DnaK/J) to assist folding
Use specialized E. coli strains (Arctic Express, SHuffle) designed for difficult proteins
Buffer optimization:
Screen multiple buffer systems (HEPES, phosphate, Tris) at different pH values (6.5-8.5)
Include stabilizing additives:
Glycerol (10-20%) to prevent aggregation
DTT or β-mercaptoethanol (1-5 mM) to maintain reduced state
NaCl or KCl (50-300 mM) to shield surface charges
MgCl₂ (1-5 mM) to stabilize nucleotide binding site
Storage conditions:
Test protein stability at different temperatures (4°C, -20°C, -80°C)
Evaluate cryoprotectants (glycerol, sucrose) for freeze-thaw stability
Determine if flash-freezing in liquid nitrogen preserves activity better than slow freezing
Consider lyophilization with appropriate excipients for long-term storage
Fusion partners and solubility tags:
Test expression with different tags:
Thioredoxin (TrxA) or glutathione S-transferase (GST) for solubility enhancement
SUMO or MBP for improved folding and solubility
Consider tag position (N-terminal vs. C-terminal) effects on folding
Enzyme stabilization techniques:
Add substrate analogs or product during purification to stabilize active conformation
Test chemical cross-linking for stabilizing quaternary structure
Consider consensus engineering to improve intrinsic stability
Systematic application of these approaches can significantly improve the stability and handling properties of recombinant P. amoebophila gmk.
Accurately assessing the biological impact of P. amoebophila gmk inhibition requires specialized approaches due to the organism's obligate intracellular lifestyle:
In vitro enzyme inhibition studies:
Establish dose-response relationships for inhibitors
Determine inhibition mechanism and kinetic parameters
Compare effects on P. amoebophila gmk versus host (Acanthamoeba) gmk
Cell culture models:
Acanthamoeba infected with P. amoebophila provides the most relevant model
Establish methods to quantify bacterial growth:
qPCR targeting P. amoebophila-specific genes
Immunofluorescence microscopy to enumerate bacterial inclusions
Transmission electron microscopy for detailed morphological assessment
Metabolic impact assessment:
Measure nucleotide pools using LC-MS/MS after inhibitor treatment
Monitor GTP/GDP/GMP levels to confirm on-target effects
Assess global metabolic changes to identify compensatory mechanisms
Genetic approaches (if feasible):
Attempt conditional knockdown of gmk expression in P. amoebophila
Create point mutations in gmk to generate inhibitor-resistant variants
Express heterologous gmk to test for functional complementation
Host-pathogen interaction analysis:
Evaluate effects on bacterial developmental cycle
Assess changes in host cell responses
Monitor bacterial stress responses (e.g., (p)ppGpp accumulation)
Data integration:
Correlate enzymatic inhibition with bacterial growth inhibition
Establish pharmacokinetic/pharmacodynamic relationships
Build mathematical models to predict effective inhibition parameters
These approaches collectively provide robust evidence for the biological consequences of gmk inhibition and help validate gmk as a potential therapeutic target.
Studying P. amoebophila gmk provides unique insights into the evolution of host-pathogen interactions through several perspectives:
Metabolic dependency relationships:
Evolutionary adaptation signatures:
Comparison of P. amoebophila gmk with gmk from pathogenic Chlamydiaceae reveals adaptations to different host environments (amoeba vs. mammalian cells)
Sequence changes in substrate binding regions may reflect adaptation to different intracellular nucleotide concentrations or compositions
Regulatory network evolution:
Changes in gmk regulation (e.g., (p)ppGpp sensitivity) between environmental Chlamydiae and pathogenic species illuminate the evolution of stress responses
Transcriptional regulation patterns during the developmental cycle provide insights into adaptation to complex host environments
Reductive evolution principles:
P. amoebophila represents an intermediate state between free-living bacteria and highly reduced obligate pathogens
The conservation of gmk across this evolutionary spectrum highlights its essential nature
The functional analysis of gmk helps define the minimal metabolic requirements for intracellular survival
Horizontal gene transfer assessment:
Unusual features in P. amoebophila gmk sequence or structure might indicate horizontal gene transfer events
Such events could represent pivotal moments in the evolution of host adaptation
Ancient symbiosis model:
P. amoebophila's relationship with amoebae represents an ancient host-symbiont relationship
Understanding how gmk functions in this system provides insights into the early evolution of intracellular lifestyles
This research contributes to our fundamental understanding of how metabolic dependencies evolve during the transition to an intracellular lifestyle, with broader implications for host-pathogen co-evolution.
For effective structural prediction and virtual screening of P. amoebophila gmk, several computational approaches can be leveraged:
Homology modeling:
Identify suitable templates from structurally characterized bacterial gmk enzymes (>30% sequence identity preferred)
Use multiple templates for challenging regions
Employ advanced modeling tools such as:
AlphaFold2 or RoseTTAFold for enhanced accuracy
MODELLER for template-based modeling with refinement
SWISS-MODEL for automated modeling
Model refinement and validation:
Refine using molecular dynamics simulations (50-100 ns)
Validate using:
PROCHECK for stereochemical quality
VERIFY3D for structural compatibility with sequence
QMEANDisco for distance-dependent model quality estimation
Active site analysis:
Identify conserved catalytic residues through multiple sequence alignment
Characterize binding pocket properties:
Volume and shape using CASTp or POCASA
Electrostatic properties using APBS
Hydrophobicity distribution
Virtual screening workflow:
Prepare diverse compound libraries:
Known nucleotide analogs and kinase inhibitors
Natural product databases
Fragment libraries
Implement hierarchical screening:
Pharmacophore filtering based on key interactions
Molecular docking using Glide, AutoDock Vina, or GOLD
MM-GBSA rescoring for improved ranking
Advanced simulation techniques:
Molecular dynamics for binding mode stability assessment
Free energy calculations (MM-PBSA, FEP) for binding affinity estimation
Residence time prediction for promising inhibitors
Machine learning integration:
Develop predictive models for activity using known kinase inhibitor data
Implement deep learning methods for binding affinity prediction
Use active learning to guide subsequent experimental testing
Visualization and interpretation:
Analyze protein-ligand interactions using:
Hydrogen bond networks
Hydrophobic contacts
π-stacking interactions
Compare binding modes across multiple compounds to define structure-activity relationships
These computational approaches provide a comprehensive framework for structure-based inhibitor discovery targeting P. amoebophila gmk, accelerating the identification of potent and selective compounds.