Recombinant Protochlamydia amoebophila 2-dehydro-3-deoxyphosphooctonate aldolase (kdsA)

Shipped with Ice Packs
In Stock

Description

Introduction to Recombinant Protochlamydia amoebophila 2-dehydro-3-deoxyphosphooctonate aldolase (KdsA)

Recombinant Protochlamydia amoebophila 2-dehydro-3-deoxyphosphooctonate aldolase (KdsA) refers to a specific enzyme that has been produced using recombinant DNA technology . KdsA, also known as 3-deoxy-D-manno-octulosonate 8-phosphate synthase (KDO8PS), catalyzes the condensation of D-arabinose 5-phosphate (A5P) and phosphoenolpyruvate (PEP) to form 3-deoxy-D-manno-octulosonate 8-phosphate (KDO8P) . This enzyme plays a crucial role in the biosynthesis of 3-deoxy-D-manno-octulosonate (KDO), a key component found in bacterial lipopolysaccharides and plant polysaccharides .

Function and Significance

KdsA is essential for the synthesis of KDO 8-P, a compound required for lipid A maturation and cellular growth . KDO is a vital component of the lipopolysaccharide (LPS) in Gram-negative bacteria, contributing to the structural integrity and functionality of the bacterial cell wall . Additionally, KDO is found in the rhamnogalacturonan II, a complex pectic polysaccharide in plants . The enzyme's activity is typically measured using either a discontinuous colorimetric assay or a continuous spectrophotometric assay .

Functional Characterization of Recombinant KdsA

The functional characterization of recombinant KdsA involves expressing the enzyme in a host organism like Escherichia coli, followed by purification and biochemical assays to determine its activity and kinetic parameters . Studies often include determining the Michaelis constant (K$$_m$$) for its substrates and the catalytic rate constant (k$$_cat$$) .

KdsA in Different Organisms

KdsA homologs have been identified and characterized in various organisms, including plants and bacteria. Plant KDOPSs may represent a distinct class compared to prokaryotic enzymes based on phylogenetic analysis and biochemical characterization . In Chlamydia pneumoniae, the kdtA gene encodes a KDO transferase involved in lipopolysaccharide biosynthesis .

Biotechnological and Pharmaceutical Relevance

Inhibitors of KdsA could potentially serve as antibacterial agents by disrupting the synthesis of LPS in Gram-negative bacteria . Understanding the structure and function of KdsA may aid in the development of novel therapeutics targeting bacterial infections.

Kinetic Studies of KdsA

Kinetic studies reveal important parameters about enzyme activity. For A. thaliana KDOPS, the enzyme follows a sequential mechanism with a $$K_m$$ = 3.6 μM for phosphoenolpyruvate and 3.8 μM for D-arabinose 5-phosphate, and a $$k_{cat}$$ = 5.9 $$s^{-1}$$ at 37 °C .

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference during order placement for customized preparation.
Lead Time
Delivery times vary depending on the purchasing method and location. Please consult your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard blue ice packs unless dry ice shipping is requested in advance. Additional fees apply for dry ice shipping.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to collect the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50%, which may serve as a guideline.
Shelf Life
Shelf life depends on several factors, including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized formulations have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during the manufacturing process.
The tag type is determined during production. If a specific tag type is required, please inform us; we will prioritize development of the specified tag.
Synonyms
kdsA; pc03622-dehydro-3-deoxyphosphooctonate aldolase; EC 2.5.1.55; 3-deoxy-D-manno-octulosonic acid 8-phosphate synthase; KDO-8-phosphate synthase; KDO 8-P synthase; KDOPS; Phospho-2-dehydro-3-deoxyoctonate aldolase
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-275
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Protochlamydia amoebophila (strain UWE25)
Target Names
kdsA
Target Protein Sequence
MRRIVVKDFA IGPKEPLVIM SGPCVIESET HCLKAAETLK NMFEKYNVSL IFKSSYDKAN RSAYDSFRGP GLEEGLRILE RIQKEFGLAV VTDVHSPQEA TTAGSVCEII QIPAFLCRQT DLILAAAQTG AIVSIKKGQF LAPWDMENVI RKMESGGNSN IILVDRGTTF GYNNLISDMR GIPIMQELGY PVCFDATHSV QKPGGLGSKS GGDREFIPIL AKAALAAGAN CLFIESHPNP SEAKSDAASV MDFKDLDQLL PQFKELYELI QKQGK
Uniprot No.

Target Background

Database Links

KEGG: pcu:pc0362

STRING: 264201.pc0362

Protein Families
KdsA family
Subcellular Location
Cytoplasm.

Q&A

What is the biological function of 2-dehydro-3-deoxyphosphooctonate aldolase in Protochlamydia amoebophila?

2-dehydro-3-deoxyphosphooctonate aldolase (kdsA) in Protochlamydia amoebophila catalyzes a crucial step in the biosynthesis of 3-deoxy-D-manno-octulosonic acid 8-phosphate (KDO-8-phosphate), which is an essential component of bacterial lipopolysaccharides (LPS) in the cell wall. Similar to other members of this enzyme family, such as the homologous enzyme in Vibrio vulnificus (EC 2.5.1.55), it facilitates the condensation of phosphoenolpyruvate and arabinose 5-phosphate to form KDO-8-phosphate . This reaction represents a critical pathway in cell wall biosynthesis for many Gram-negative bacteria and related organisms, playing a vital role in structural integrity and virulence.

How does Protochlamydia amoebophila kdsA compare structurally to other bacterial aldolases?

While the complete structural characterization of P. amoebophila kdsA is still developing, comparative analysis with related enzymes from other bacterial species reveals conserved features. Analysis of sequence homology with the well-characterized 2-dehydro-3-deoxyphosphooctonate aldolase from Vibrio vulnificus (Q7MMY2) shows significant structural conservation in catalytic domains . Unlike the E. coli phospho-2-dehydro-3-deoxyheptonate aldolase (such as AroH), which participates in aromatic amino acid biosynthesis, the P. amoebophila kdsA belongs to a different functional class focused on cell wall component synthesis . Structural studies using X-ray crystallography techniques similar to those used for related aldolases (such as the rhombohedral crystals of DDG aldolase from E. coli K-12) would likely reveal a comparable quaternary structure with space group symmetry and unit-cell parameters that reflect its catalytic mechanism .

What metabolic pathways involve kdsA in Protochlamydia amoebophila, and how do they differ from pathways in related organisms?

The kdsA enzyme in P. amoebophila participates in the lipopolysaccharide biosynthesis pathway, which is critical for cell envelope formation. Recent metabolic studies of P. amoebophila elementary bodies have demonstrated that, unlike many other obligate intracellular bacteria, P. amoebophila maintains active metabolism even in the extracellular stage . The metabolic activity depends on D-glucose availability, which serves as an essential substrate to sustain enzymatic functions including those in cell wall biosynthesis pathways . This represents a significant difference from closely related Chlamydiaceae, whose extracellular elementary body (EB) stage was traditionally considered metabolically inert. The kdsA pathway in P. amoebophila appears to remain active even in EBs, which may explain the organism's ability to maintain infectivity for longer periods outside host cells compared to related species .

What are the optimal conditions for heterologous expression of recombinant P. amoebophila kdsA?

For optimal heterologous expression of recombinant P. amoebophila kdsA, an E. coli expression system using BL21(DE3) or similar strains is recommended, based on successful protocols for related aldolases. Expression should be conducted using a vector containing an N-terminal 6xHis-tag for subsequent purification, similar to the approach used for recombinant E. coli phospho-2-dehydro-3-deoxyheptonate aldolase . The optimal expression conditions include:

ParameterOptimal ConditionNotes
Induction temperature16-18°CLower temperatures reduce inclusion body formation
IPTG concentration0.1-0.5 mMHigher concentrations may not improve yield
Post-induction time16-20 hoursExtended time improves soluble protein yield
Media compositionLB with glucose supplement0.5% glucose supports metabolic activity
OD₆₀₀ at induction0.6-0.8Ensures cells are in exponential growth phase

When optimizing expression, it's crucial to monitor enzyme activity rather than simply protein yield, as higher expression levels may lead to non-functional protein through improper folding .

What purification strategies are most effective for obtaining high-purity recombinant P. amoebophila kdsA?

A multi-step purification strategy is recommended for obtaining high-purity recombinant P. amoebophila kdsA:

  • Initial capture: Immobilized metal affinity chromatography (IMAC) using Ni-NTA resin is effective for His-tagged kdsA. Elution should be performed with an imidazole gradient (20-250 mM) in a buffer containing 50 mM Tris-HCl pH 8.0, 300 mM NaCl .

  • Intermediate purification: Ion exchange chromatography using a Q-Sepharose column effectively separates the target protein from contaminants with similar molecular weights but different charge properties.

  • Polishing step: Size exclusion chromatography (Superdex 75 or 200) in 20 mM Tris-HCl pH 7.5, 150 mM NaCl, 5% glycerol buffer removes aggregates and yields >95% pure protein.

The purified protein should be stored in a buffer containing 5-50% glycerol at -80°C to maintain stability . Purity assessment by SDS-PAGE should achieve >85% purity, with enzymatic activity assays confirming functional integrity of the purified protein .

How can researchers assess the functional integrity of purified recombinant kdsA?

Functional integrity assessment of purified recombinant kdsA should employ multiple complementary approaches:

  • Enzymatic activity assay: Measure the forward reaction (condensation of phosphoenolpyruvate and arabinose 5-phosphate) by quantifying the production of KDO-8-phosphate using thiobarbituric acid assay. The specific activity should be expressed as μmol of product formed per minute per mg of enzyme under standard conditions (typically 37°C, pH 7.5) .

  • Thermal shift assay: Assess protein stability using differential scanning fluorimetry with SYPRO Orange dye. A well-folded kdsA typically shows a cooperative unfolding transition with Tm values between 45-60°C depending on buffer conditions.

  • Circular dichroism spectroscopy: Confirm proper secondary structure content, particularly α-helical and β-sheet elements consistent with the predicted structure based on homologous enzymes.

  • Size exclusion chromatography coupled with multi-angle light scattering (SEC-MALS): Verify the oligomeric state of the purified protein, as proper quaternary structure is essential for catalytic activity in many aldolases.

A fully functional enzyme should demonstrate Michaelis-Menten kinetics with KM values for phosphoenolpyruvate in the range of 10-100 μM, comparable to those reported for homologous enzymes from other bacterial species .

What crystallization conditions have proven successful for structural determination of bacterial aldolases similar to P. amoebophila kdsA?

Successful crystallization of bacterial aldolases similar to P. amoebophila kdsA has been achieved using the following conditions:

For related 2-dehydro-3-deoxygalactarate aldolase from E. coli K-12, rhombohedral crystals were obtained that belong to space group R32 with unit-cell parameters a = 93 Å, α = 85 degrees . These crystals diffracted to beyond 1.8 Å resolution on a Cu Kα rotating-anode generator . The crystallization conditions included:

ComponentConcentration/Condition
Protein concentration10-15 mg/mL
Precipitant1.6-2.0 M ammonium sulfate
Buffer0.1 M Tris-HCl, pH 7.5-8.0
Additives5-10 mM divalent cations (Mg²⁺ or Mn²⁺)
Temperature18-20°C
MethodHanging drop vapor diffusion

The asymmetric unit of these crystals contained two molecules, corresponding to a packing density of 1.34 ų Da⁻¹ . When attempting crystallization of P. amoebophila kdsA, researchers should consider both substrate-free and substrate-bound forms to capture different conformational states relevant to the catalytic mechanism.

How can site-directed mutagenesis be used to investigate the catalytic mechanism of P. amoebophila kdsA?

Site-directed mutagenesis provides a powerful approach to investigate the catalytic mechanism of P. amoebophila kdsA. Based on sequence alignments with homologous enzymes like the 2-dehydro-3-deoxyphosphooctonate aldolase from Vibrio vulnificus (Q7MMY2) , researchers should target the following residues for mutagenesis:

  • Catalytic residues: Based on the conserved active site architecture of KDO-8-phosphate synthases, mutagenesis of putative catalytic residues (typically lysine and aspartate residues involved in Schiff base formation and proton transfer) to alanine or less reactive amino acids should substantially reduce or eliminate activity.

  • Substrate binding residues: Mutation of residues predicted to interact with phosphoenolpyruvate or arabinose 5-phosphate to assess their contribution to substrate specificity and binding affinity.

  • Metal coordination sites: If the enzyme requires divalent cations for activity (as many aldolases do), mutation of metal-coordinating residues (typically aspartate, glutamate, or histidine) to evaluate their role in catalysis.

The mutant proteins should be characterized using:

  • Steady-state kinetics to determine changes in KM and kcat values

  • Isothermal titration calorimetry to directly measure substrate binding affinities

  • Structural analysis (X-ray crystallography or cryo-EM) to visualize conformational changes

This approach has successfully elucidated catalytic mechanisms in related aldolases and would provide valuable insights into the specific functional properties of P. amoebophila kdsA.

What computational approaches can be employed to predict substrate specificity and catalytic efficiency of P. amoebophila kdsA?

Several computational approaches can effectively predict substrate specificity and catalytic efficiency of P. amoebophila kdsA:

  • Homology modeling and molecular docking: Using the crystal structures of homologous enzymes (such as Vibrio vulnificus KDO-8-phosphate synthase, Q7MMY2) as templates, researchers can construct a detailed homology model of P. amoebophila kdsA. Subsequently, molecular docking studies with various substrates can predict binding affinities and productive binding modes. Software packages like MODELLER for homology modeling and AutoDock Vina for docking are recommended.

  • Molecular dynamics simulations: Simulating the enzyme-substrate complex over nanosecond to microsecond timescales can reveal dynamic aspects of catalysis, including:

    • Conformational changes upon substrate binding

    • Water networks in the active site

    • Proton transfer pathways during catalysis

    • Substrate orientation fluctuations

  • Quantum mechanics/molecular mechanics (QM/MM) approaches: For detailed understanding of the reaction mechanism, QM/MM calculations can model the electronic structure of the active site during catalysis while treating the rest of the protein with molecular mechanics. This approach can predict:

    • Energy barriers for different reaction pathways

    • Transition state structures

    • Effects of active site mutations on catalytic efficiency

  • Sequence-based machine learning approaches: Training algorithms on datasets of characterized aldolases can predict substrate specificity and catalytic parameters based solely on primary sequence. This is particularly valuable when structural information is limited.

These computational predictions should be validated experimentally through enzyme kinetics assays with multiple substrates to establish structure-function relationships unique to P. amoebophila kdsA.

How can recombinant P. amoebophila kdsA be utilized as a tool for studying bacterial cell wall synthesis?

Recombinant P. amoebophila kdsA serves as a valuable tool for studying bacterial cell wall synthesis through several applications:

  • In vitro reconstitution of KDO biosynthesis pathway: The purified enzyme can be combined with other enzymes in the pathway to study the coordinated synthesis of KDO and its incorporation into lipopolysaccharides. This system allows researchers to identify rate-limiting steps and regulatory points in cell wall component synthesis.

  • Development of specific inhibitors: The recombinant enzyme provides a platform for high-throughput screening of small molecule libraries to identify selective inhibitors of KDO-8-phosphate synthesis. Such inhibitors could serve as chemical probes to study the consequences of disrupting this pathway in live bacteria.

  • Investigation of chlamydial elementary body survival: As P. amoebophila elementary bodies maintain metabolic activity outside host cells , the role of continuous KDO synthesis in extracellular survival can be studied by manipulating kdsA activity through genetic or chemical approaches.

  • Comparative biochemistry: Side-by-side characterization of kdsA enzymes from P. amoebophila and related pathogenic species can reveal adaptations specific to the intracellular lifestyle of these bacteria. Differences in substrate specificity, catalytic efficiency, or regulation may reflect evolutionary adaptations to different host environments.

  • Isotope labeling studies: Using the recombinant enzyme with isotopically labeled substrates allows tracking of newly synthesized cell wall components, providing insights into cell wall turnover and remodeling during the developmental cycle of Chlamydia-related bacteria.

These applications collectively contribute to a deeper understanding of the unique aspects of cell wall biology in obligate intracellular bacteria.

What is the significance of studying P. amoebophila kdsA in the context of Protochlamydia's role as an emerging pathogen?

Studying P. amoebophila kdsA has significant implications in understanding Protochlamydia's role as an emerging pathogen:

  • Target for antimicrobial development: As KDO-8-phosphate synthase catalyzes an essential step in LPS biosynthesis that has no human homolog, it represents a potential target for developing selective antimicrobials against Protochlamydia and related pathogens. Characterizing the unique structural and functional properties of P. amoebophila kdsA can guide structure-based drug design efforts.

  • Understanding persistence mechanisms: The recent discovery that Protochlamydia elementary bodies remain metabolically active outside host cells, with D-glucose metabolism being essential for maintaining infectivity , suggests that kdsA activity may be crucial for extracellular survival. This contradicts the traditional view of chlamydial elementary bodies as metabolically inert and may explain how these organisms persist in the environment.

  • Host-pathogen interaction studies: KDO-containing molecules on the bacterial surface interact with host immune receptors. Characterizing P. amoebophila kdsA and its products could reveal how these bacteria evade or modulate host immune responses, particularly in the context of respiratory infections where Protochlamydia naegleriophila (a close relative) has been identified as a potential etiologic agent of pneumonia .

  • Diagnostic marker development: The unique properties of P. amoebophila kdsA could be exploited for developing specific diagnostic tests, similar to how specific PCR for Protochlamydia spp. has been applied to bronchoalveolar lavage samples to detect these bacteria in patients with pneumonia .

  • Evolutionary insights: Comparative analysis of kdsA across the Chlamydiales order can provide insights into the evolution of pathogenicity in this group, potentially identifying genetic changes that correlate with shifts in host range or tissue tropism.

These research directions are particularly important given the growing evidence that environmental chlamydiae, including Protochlamydia species, may be emerging human pathogens associated with respiratory infections .

How can metabolic studies of P. amoebophila elementary bodies inform our understanding of kdsA function in extracellular survival?

Recent metabolic studies of P. amoebophila elementary bodies have revolutionized our understanding of chlamydial biology and have specific implications for kdsA function:

  • Active metabolism in extracellular stage: P. amoebophila elementary bodies (EBs) exhibit respiratory activity and can import D-glucose in the absence of host cells, challenging the traditional view that chlamydial EBs are metabolically inert . This suggests that kdsA and other enzymes remain active during the extracellular phase.

  • D-glucose dependency: Experiments have demonstrated that D-glucose availability is essential for maintaining metabolic activity in P. amoebophila EBs, with replacement by non-metabolizable L-glucose leading to rapid decline in infectious particles . This dependency suggests that glucose is channeled into multiple pathways, potentially including KDO synthesis via kdsA activity.

  • Quantitative assessment of metabolic activity: Flow cytometry and fluorescent glucose analog (2-NBDG) uptake experiments have shown that a high proportion of EBs (comparable to the percentage showing respiratory activity) can import glucose . This correlation supports the hypothesis that imported glucose fuels metabolic pathways including those involving kdsA.

  • Comparative analysis with pathogenic chlamydiae: Similar to P. amoebophila, Chlamydia trachomatis (a major human pathogen) shows decreased infectivity in the absence of nutrients , suggesting that metabolic activity in the extracellular stage is a conserved feature with biological significance across the Chlamydiales order.

  • Implications for cell wall maintenance: The continued activity of kdsA during the extracellular stage would enable ongoing synthesis of KDO-8-phosphate, allowing for maintenance and potential remodeling of the cell envelope during environmental exposure. This could be a critical adaptation for survival outside the host cell and for maintaining infection potential.

These findings collectively suggest that kdsA function may be critical not only during intracellular replication but also during extracellular survival, representing a potential target for interventions aimed at reducing environmental persistence of these bacteria.

What are the challenges and solutions for studying enzyme kinetics of recombinant P. amoebophila kdsA?

Studying enzyme kinetics of recombinant P. amoebophila kdsA presents several challenges with corresponding methodological solutions:

ChallengeSolutionMethodology Details
Limited substrate availabilityChemical synthesis of arabinose 5-phosphateUtilize the optimized synthesis route via arabinose 1,5-bisphosphate intermediate with subsequent selective dephosphorylation
Low sensitivity of product detectionCoupled enzyme assaysLink KDO-8-phosphate production to NAD(P)H oxidation via auxiliary enzymes for spectrophotometric monitoring at 340 nm
Enzyme instabilityBuffer optimizationScreen additives (10-20% glycerol, 1-5 mM DTT, divalent cations) to enhance stability during kinetic measurements
Bi-substrate reaction mechanismInitial velocity pattern analysisPerform initial velocity studies with varying concentrations of both substrates to determine if the mechanism is sequential or ping-pong
Potential product inhibitionProduct inhibition analysisInclude varying concentrations of purified KDO-8-phosphate to assess competitive or non-competitive inhibition patterns
Temperature sensitivityTemperature-controlled kineticsMeasure activity across 20-50°C range to determine temperature optimum and stability profile
pH dependencepH-rate profile determinationDetermine pH optimum and identify ionizable groups in the active site by measuring activity across pH 5.5-9.5

For accurate kinetic parameter determination, researchers should use global fitting of initial velocity data to appropriate bi-substrate models rather than simplified Lineweaver-Burk analyses. This approach provides more reliable estimates of kcat, KM values for both substrates, and their potential interaction terms.

What advanced spectroscopic techniques can provide insights into substrate binding and catalytic mechanism of P. amoebophila kdsA?

Several advanced spectroscopic techniques can elucidate substrate binding and catalytic mechanisms of P. amoebophila kdsA:

  • Nuclear Magnetic Resonance (NMR) Spectroscopy:

    • ¹H-¹⁵N HSQC titrations with substrates can map binding interactions by monitoring chemical shift perturbations

    • ³¹P NMR can track phosphate group changes during catalysis

    • Saturation transfer difference (STD) NMR can identify which substrate moieties make closest contact with the enzyme

  • Electron Paramagnetic Resonance (EPR) Spectroscopy:

    • If the enzyme utilizes metal cofactors, EPR can characterize the electronic environment around paramagnetic centers

    • Site-directed spin labeling combined with EPR can measure distances between specific residues during catalysis

  • Vibrational Spectroscopy:

    • Fourier-transform infrared (FTIR) spectroscopy can detect formation/breakage of specific bonds during catalysis

    • Raman spectroscopy can monitor conformational changes associated with substrate binding and product formation

    • Time-resolved vibrational spectroscopy can capture transient intermediates in the catalytic cycle

  • X-ray Absorption Spectroscopy (XAS):

    • Extended X-ray absorption fine structure (EXAFS) analysis can provide detailed geometric information about metal coordination sites if present in the enzyme

    • X-ray absorption near edge structure (XANES) can determine oxidation states of metal cofactors during different stages of catalysis

  • Hydrogen-Deuterium Exchange Mass Spectrometry (HDX-MS):

    • Maps regions of conformational change upon substrate binding by measuring differential solvent exposure

    • Can identify allosteric networks that connect substrate binding to distant regions of the protein

These techniques provide complementary information that, when integrated, creates a comprehensive understanding of the enzyme's structure-function relationships and reaction mechanism at molecular resolution.

How can researchers develop effective inhibitors of P. amoebophila kdsA for use as research tools?

Developing effective inhibitors of P. amoebophila kdsA as research tools requires a systematic multi-stage approach:

  • Structure-based rational design:

    • Generate a high-quality homology model based on related aldolases if crystal structure is unavailable

    • Perform virtual screening of compound libraries targeting the active site

    • Design transition state analogs that mimic the geometry and charge distribution of the reaction's transition state

    • Employ fragment-based approaches to identify building blocks with affinity for different regions of the active site

  • Synthesis and initial screening:

    • Synthesize top candidates from virtual screening

    • Develop a medium-throughput enzymatic assay for initial activity screening

    • Determine IC₅₀ values for promising candidates

  • Mechanistic characterization:

    • Perform detailed kinetic analysis to determine inhibition mode (competitive, noncompetitive, uncompetitive, or mixed)

    • Determine Ki values under standardized conditions

    • Assess time-dependent inhibition to identify potential covalent or slow-binding inhibitors

  • Structural validation:

    • Confirm binding mode through X-ray crystallography or NMR studies of enzyme-inhibitor complexes

    • Use this structural information to guide iterative optimization

  • Selectivity profiling:

    • Test inhibitors against related aldolases to ensure specificity

    • Create a selectivity profile across a panel of enzymes involved in similar or adjacent metabolic pathways

  • Cellular validation:

    • Evaluate inhibitor uptake and target engagement in cellular contexts

    • Confirm on-target effects by complementary genetic approaches (e.g., overexpression of kdsA should increase inhibitor concentrations required for effects)

A particularly promising approach involves designing bisubstrate analog inhibitors that simultaneously occupy both the phosphoenolpyruvate and arabinose 5-phosphate binding sites, potentially achieving higher affinity and specificity than compounds targeting either substrate site alone.

How does P. amoebophila kdsA differ from homologous enzymes in other Chlamydia-related bacteria?

Comparative analysis reveals several key differences between P. amoebophila kdsA and homologous enzymes in other Chlamydia-related bacteria:

  • Sequence divergence: Phylogenetic analysis demonstrates that P. amoebophila kdsA shares approximately 91-93% sequence similarity with other Parachlamydiaceae family members, but only 85-88% similarity with other Chlamydiales order members . This sequence divergence reflects the evolutionary distance between these bacterial groups and may indicate functional adaptations.

  • Substrate specificity: While the core catalytic function is conserved, subtle variations in active site residues may confer differences in substrate preference or catalytic efficiency. For instance, differences in the binding pocket architecture could affect the affinity for phosphoenolpyruvate or arabinose 5-phosphate between species.

  • Metabolic context: P. amoebophila maintains active metabolism in its elementary body stage, with glucose import capabilities that support continued enzymatic activity . This contrasts with some pathogenic Chlamydia species whose elementary bodies were traditionally considered metabolically dormant. The metabolic activity of kdsA likely reflects this broader difference in extracellular metabolic capacity.

  • Structural stability: Protein stability parameters may differ between species, reflecting adaptation to different host environments. P. amoebophila, as an environmental chlamydia primarily infecting amoebae, likely exhibits different stability characteristics compared to human pathogens like C. trachomatis, which must withstand different environmental stresses.

  • Regulatory mechanisms: The expression patterns and regulation of kdsA may differ significantly between species based on their life cycles and host adaptation. Genomic context analysis suggests differences in operon organization and potential regulatory elements controlling kdsA expression across Chlamydia-related bacteria.

These differences highlight the evolutionary adaptations of this enzyme across the Chlamydiales order and may provide insights into host specificity and environmental persistence mechanisms.

What evolutionary insights can be gained from studying the conservation patterns in kdsA across bacterial phyla?

Studying conservation patterns in kdsA across bacterial phyla provides valuable evolutionary insights:

  • Essential nature reflected in conservation: The high degree of sequence conservation in catalytic residues across diverse bacterial phyla underscores the essential nature of KDO-8-phosphate synthesis. This conservation extends across Gram-negative bacteria, suggesting strong purifying selection maintaining enzyme function throughout bacterial evolution.

  • Taxonomic signatures: Despite core functional conservation, specific sequence motifs can serve as taxonomic signatures. For instance, Protochlamydia species show characteristic sequence patterns that distinguish them from other bacterial phyla, potentially reflecting adaptation to their unique intracellular lifestyle .

  • Horizontal gene transfer assessment: Phylogenetic analysis of kdsA sequences can reveal potential horizontal gene transfer events. Incongruence between kdsA-based phylogeny and species phylogeny based on ribosomal RNA genes would suggest horizontal transmission rather than vertical inheritance.

  • Selection pressure mapping: Calculating the ratio of non-synonymous to synonymous substitutions (dN/dS) across the enzyme structure identifies regions under different selection pressures. Typically, active site residues show strong purifying selection (low dN/dS), while surface-exposed regions may exhibit higher variability.

  • Co-evolution with other pathway components: Correlation analysis between evolutionary patterns of kdsA and other enzymes in the KDO biosynthesis pathway can reveal co-evolutionary relationships. Such analysis can identify functionally coupled residues that have co-evolved to maintain protein-protein interactions or substrate channeling.

  • Host adaptation signatures: Comparing kdsA sequences from bacteria with different host ranges (from environmental to strict human pathogens) can identify adaptive changes potentially associated with host switching events or specialization to particular host environments.

These evolutionary insights contribute to our understanding of bacterial cell wall evolution and may guide the development of species-specific interventions targeting this essential pathway.

How does metabolic activity in Protochlamydia elementary bodies change our understanding of enzyme function across the bacterial life cycle?

The discovery of metabolic activity in Protochlamydia elementary bodies significantly transforms our understanding of enzyme function across the bacterial life cycle:

  • Paradigm shift in developmental biology: The traditional model of chlamydial development portrayed elementary bodies (EBs) as metabolically inert spore-like particles. The observation that P. amoebophila EBs actively import D-glucose and maintain respiratory activity challenges this model and suggests that enzymes like kdsA remain functional throughout the developmental cycle.

  • Continuous enzymatic activity: The demonstration that a high proportion of P. amoebophila EBs (comparable to the percentage showing respiratory activity) can import the fluorescent glucose analog 2-NBDG indicates that metabolic pathways remain active . This suggests that kdsA and other enzymes do not undergo complete inactivation during the EB stage, contrasting with previous assumptions.

  • Metabolic adaptation for environmental persistence: The metabolic activity in EBs likely represents an adaptation for survival outside host cells. The continued function of kdsA would allow for maintenance and potential remodeling of the cell envelope during environmental exposure, potentially explaining the extended infectious potential of these bacteria.

  • Differential regulation rather than inactivation: Instead of complete enzyme inactivation during developmental transitions, the findings suggest differential regulation of metabolic pathways. Enzymes may be maintained in a functionally competent state but with altered activity levels or substrate access depending on developmental stage.

  • Evolutionary implications: The maintenance of metabolic activity in Protochlamydia EBs, compared to the more limited activity in pathogenic Chlamydia species, may represent an ancestral trait that has been partially lost in highly adapted pathogens. This suggests that enzyme function across the life cycle has been shaped by evolutionary pressures related to host adaptation.

  • Research methodology implications: These findings emphasize the importance of studying enzyme function in context-specific ways that account for the bacterial developmental stage. Traditional in vitro characterization may not capture the regulated nature of enzyme activity throughout the bacterial life cycle.

This new understanding has profound implications for research approaches, potentially leading to the identification of metabolic vulnerabilities that could be targeted across different developmental stages.

What emerging technologies could advance our understanding of P. amoebophila kdsA structure and function?

Several emerging technologies show particular promise for advancing our understanding of P. amoebophila kdsA:

  • Cryo-electron microscopy (cryo-EM): Recent advances in single-particle cryo-EM now allow near-atomic resolution structures without the need for crystallization. This could be particularly valuable for capturing different conformational states of kdsA during catalysis and for determining structures of complexes with substrates, products, or inhibitors that have been challenging to crystallize.

  • AlphaFold and deep learning structure prediction: The revolutionary accuracy of AlphaFold and related deep learning approaches for protein structure prediction could provide high-quality structural models of P. amoebophila kdsA, especially when combined with sparse experimental constraints from techniques like crosslinking mass spectrometry.

  • Time-resolved X-ray crystallography: Advanced synchrotron and X-ray free-electron laser (XFEL) sources enable capturing structural snapshots of enzymes during catalysis at femtosecond to millisecond timescales. This could reveal transient states in the kdsA reaction mechanism that have been inaccessible to traditional structural biology approaches.

  • Nanobody-assisted structural biology: Developing specific nanobodies against P. amoebophila kdsA could stabilize specific conformational states for structural studies and provide tools for tracking the enzyme in cellular contexts.

  • In-cell NMR spectroscopy: This emerging technique allows for studying protein structure and dynamics in living cells, potentially revealing how the intracellular environment affects kdsA function compared to in vitro conditions.

  • Single-molecule enzymology: Advanced fluorescence techniques like single-molecule FRET could reveal the conformational dynamics of kdsA during substrate binding and catalysis, providing insights into reaction mechanisms at unprecedented resolution.

  • Microfluidic approaches for high-throughput biochemistry: Droplet-based microfluidic systems could enable testing thousands of reaction conditions to optimize activity and discover new substrates or inhibitors with minimal protein consumption.

These technologies, either individually or in combination, have the potential to significantly advance our understanding of P. amoebophila kdsA structure-function relationships and catalytic mechanism.

How might systems biology approaches integrate kdsA function into broader metabolic networks of Protochlamydia?

Systems biology approaches offer powerful frameworks for integrating kdsA function into the broader metabolic context of Protochlamydia:

  • Genome-scale metabolic modeling:

    • Construction of a genome-scale metabolic model for P. amoebophila, incorporating all known metabolic reactions including those catalyzed by kdsA

    • Flux balance analysis to predict how perturbations in kdsA activity would affect global metabolic fluxes

    • Identification of synthetic lethal interactions where simultaneous inhibition of kdsA and another enzyme would be catastrophic for bacterial viability

  • Metabolomics integration:

    • Untargeted metabolomics to identify unexpected metabolites affected by kdsA activity

    • Stable isotope labeling experiments to trace carbon flux through the KDO biosynthesis pathway and connected pathways

    • Integration of metabolomic data with transcriptomic and proteomic datasets to create multi-omics models of cell wall biosynthesis regulation

  • Protein-protein interaction networks:

    • Affinity purification coupled with mass spectrometry to identify protein interaction partners of kdsA

    • Bacterial two-hybrid or proximity labeling approaches to map the "interactome" of cell wall biosynthesis enzymes

    • Network analysis to identify hub proteins and regulatory nodes in cell envelope biosynthesis

  • Transcriptional regulatory networks:

    • ChIP-seq to identify transcription factors regulating kdsA expression

    • RNA-seq under various conditions to understand coordinated expression patterns of kdsA with other genes

    • Construction of gene regulatory networks to predict how environmental signals are transduced to changes in cell wall biosynthesis

  • Host-pathogen interface modeling:

    • Integration of bacterial and host cell metabolic models to understand metabolic dependencies and competition

    • Prediction of how kdsA activity influences host immune recognition through changes in cell surface components

    • Modeling of metabolic adaption during transition between host-associated and environmental stages

These systems approaches would place kdsA in its proper biological context, revealing both its direct catalytic role and its broader significance in bacterial physiology and host interaction.

What innovative experimental designs could address contradictory findings regarding kdsA function in different bacterial species?

To address contradictory findings regarding kdsA function across bacterial species, several innovative experimental approaches could be employed:

  • Heterologous complementation with controlled expression:

    • Engineer conditional knockout strains of kdsA in model organisms like E. coli

    • Complement with kdsA orthologs from different species under identical, tightly controlled expression levels

    • Quantitatively assess growth rates, cell morphology, and cell wall composition to determine functional equivalence or differences

    • This approach controls for confounding variables like expression level differences that often complicate cross-species comparisons

  • Domain-swapping chimeras:

    • Create chimeric enzymes containing domains from kdsA orthologs of different species

    • Systematically characterize these chimeras to map species-specific functional differences to specific protein regions

    • This approach can identify which structural elements underlie contradictory findings across species

  • Parallel multi-omics across species:

    • Apply identical experimental conditions and analytical methods to study kdsA function across multiple bacterial species

    • Integrate transcriptomic, proteomic, and metabolomic data to create comparable, comprehensive profiles

    • Use computational approaches to identify species-specific regulatory or metabolic contexts that might explain functional differences

  • Controlled in vivo labeling:

    • Develop parallel systems for metabolic labeling of KDO biosynthesis in different bacterial species

    • Use click-chemistry compatible precursors to track KDO incorporation into cell walls under defined conditions

    • Quantitatively compare flux through this pathway across species using identical methodology

  • Single-cell analyses:

    • Apply microfluidic or flow cytometry approaches to analyze cell-to-cell variability in kdsA function

    • Determine if contradictory findings might represent differences in population heterogeneity rather than species-level differences

  • Evolutionary reconstruction:

    • Resurrect ancestral kdsA sequences using ancestral sequence reconstruction

    • Characterize these ancestral enzymes to determine which functional properties were present in the last common ancestor

    • Map evolutionary events that led to divergent functions in different lineages

  • Standardized environmental perturbation panels:

    • Subject multiple bacterial species to identical panels of environmental stresses

    • Measure how kdsA expression, activity, and metabolic impact respond across species

    • Identify condition-specific functional differences that might explain contradictory literature findings

These approaches collectively provide a systematic framework for resolving contradictions in the literature and developing a unified understanding of kdsA function across bacterial diversity.

What are the most significant recent advances in our understanding of Protochlamydia aldolases and their roles in bacterial metabolism?

The most significant recent advances in understanding Protochlamydia aldolases include:

  • Metabolic activity in elementary bodies: The paradigm-shifting discovery that Protochlamydia elementary bodies (EBs) maintain active metabolism outside host cells, with D-glucose serving as an essential substrate . This finding contradicts the long-held view that chlamydial EBs are metabolically inert and suggests that aldolases like kdsA remain functional throughout the developmental cycle.

  • Structural insights from related aldolases: Crystallographic studies of related bacterial aldolases have provided templates for understanding the structural basis of substrate specificity and catalytic mechanism in Protochlamydia enzymes . The rhombohedral crystal structure of related aldolases, diffracting to beyond 1.8 Å resolution, has revealed critical details about active site architecture and oligomeric assembly .

  • Phylogenetic classification and diversity: Rigorous genetic and phylogenetic analyses have established the taxonomic position of Protochlamydia species, such as P. naegleriophila, revealing their relationship to other Chlamydia-related bacteria . These analyses provide an evolutionary framework for understanding aldolase diversity and adaptation.

  • Potential pathogenic relevance: The identification of Protochlamydia in clinical samples from patients with pneumonia has highlighted the potential medical significance of these organisms and their metabolic enzymes . This finding suggests that understanding aldolases in these bacteria may have implications for human health.

  • Co-culture methodologies: The development of amoeba co-culture systems for growing previously uncultivable Protochlamydia strains has enabled biochemical and functional studies that were previously impossible . These methodological advances have opened new avenues for direct characterization of Protochlamydia enzymes.

These advances collectively provide a foundation for future research on Protochlamydia aldolases and their roles in bacterial metabolism, with implications for understanding both fundamental bacterial biology and potential pathogenic mechanisms.

How does integrating structural, functional, and evolutionary studies of kdsA contribute to a comprehensive understanding of Protochlamydia biology?

Integrating structural, functional, and evolutionary studies of kdsA creates a comprehensive understanding of Protochlamydia biology through multiple interconnected insights:

  • Structural-functional correlations: By mapping sequence conservation patterns onto three-dimensional structures, researchers can identify which structural features are indispensable for function across evolutionary time. This approach reveals that while the catalytic core of kdsA is highly conserved, peripheral regions show greater variability, potentially reflecting adaptations to different cellular environments or regulatory mechanisms.

  • Evolutionary adaptation signatures: Comparative analysis of kdsA sequences across the Chlamydiales order reveals signatures of selection that correlate with host range transitions. The distinctive sequence patterns in Protochlamydia kdsA compared to other bacterial phyla likely reflect adaptation to their unique intracellular lifestyle in amoebae and potential association with human respiratory infections .

  • Metabolic context interpretation: Functional studies demonstrating the importance of D-glucose availability for maintaining P. amoebophila elementary body viability gain deeper meaning when integrated with structural knowledge of how glucose-derived metabolites feed into pathways involving kdsA. This integration explains why specific metabolic capabilities have been maintained through evolution.

  • Host-interaction insights: The surface-exposed products of pathways involving kdsA interact with host immune systems. Understanding the structural determinants of these interactions, combined with evolutionary analysis of selective pressures, reveals mechanisms by which these bacteria may evade host recognition.

  • Developmental biology framework: Integrating these perspectives helps explain the unique developmental biology of Protochlamydia, particularly the maintenance of metabolic activity in elementary bodies . This activity represents an evolutionary adaptation that can be understood through the lens of enzyme function and regulation across different life stages.

  • Taxonomic classification support: The genetic and phylogenetic analysis of genes like kdsA has contributed to defining the taxonomic position of organisms like Protochlamydia naegleriophila, with serological differentiation indices (SDIs) and sequence similarity analyses supporting its classification .

This integrated perspective enables researchers to move beyond isolated characterizations toward a systems-level understanding of how kdsA function is embedded within and contributes to the broader biology of these fascinating organisms.

What are the most promising therapeutic or diagnostic applications emerging from research on Protochlamydia aldolases?

Research on Protochlamydia aldolases is revealing several promising therapeutic and diagnostic applications:

  • Novel antimicrobial targets: As essential enzymes with no human homologs, aldolases like kdsA represent attractive targets for developing selective antimicrobials against Protochlamydia and related pathogens. The distinct active site architecture of these enzymes, compared to other bacterial aldolases, provides opportunities for developing highly specific inhibitors that don't affect human metabolism or beneficial microbiota.

  • Diagnostic PCR development: The development of specific diagnostic PCR assays targeting Protochlamydia species has already demonstrated utility in clinical settings. In one study, such a PCR assay applied to bronchoalveolar lavage samples identified Protochlamydia in a patient with pneumonia . Refined molecular diagnostics based on genes encoding aldolases could further improve detection specificity and sensitivity.

  • Metabolic activity-based diagnostics: The discovery that Protochlamydia elementary bodies remain metabolically active, with D-glucose uptake capabilities , suggests potential diagnostics based on metabolic activity rather than just DNA detection. Fluorescent substrate analogs that are processed by aldolases could enable activity-based detection of viable bacteria in clinical or environmental samples.

  • Environmental persistence intervention: Understanding the role of aldolases in maintaining extracellular viability of Protochlamydia could lead to interventions that reduce environmental persistence of these potential pathogens. Targeting the metabolic activity that sustains infectious potential outside host cells represents a novel approach to infection control.

  • Vaccine development platforms: The structural characterization of aldolases provides targets for rational vaccine design. Surface-exposed epitopes of these enzymes, or their metabolic products that appear on the bacterial surface, could serve as vaccine candidates, particularly if they are conserved across strains but sufficiently different from human or commensal bacterial proteins.

  • Microbiome monitoring tools: As our understanding of the potential pathogenic role of environmental chlamydiae grows, aldolase-based detection methods could be incorporated into microbiome monitoring systems for water sources, air handling systems, and other environments to assess risk factors for respiratory infections.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.