Subunit c is a membrane-spanning component of the ATP synthase F₀ sector, forming a ring structure that facilitates proton translocation. This process drives ATP synthesis in K. pneumoniae and other Gram-negative bacteria .
Proton Translocation: Subunit c oligomerizes to create a pore for protons, enabling the synthesis of ATP via the ATP synthase F₁F₀ complex .
Antibiotic Target: Subunit c homologs in mycobacteria are validated targets for antibiotics like TMC207 (bedaquiline), which disrupts ATP synthesis .
Genetic Conservation: The atpE gene is highly conserved across Gram-positive bacteria (e.g., Staphylococcus aureus, Streptococcus pneumoniae), suggesting cross-species relevance .
ATP Synthase Assembly: Recombinant subunit c is used to study the structural dynamics of the F₀ sector and proton channel formation .
Membrane Stability: Deletion of atpE increases bacterial sensitivity to SDS-EDTA, highlighting its role in maintaining membrane integrity .
Target Validation: Subunit c inhibition disrupts ATP production, making it a candidate for novel antimicrobial therapies .
RamA Regulation: While not directly linked to atpE, RamA-mediated regulation of ABC transporters (e.g., MlaFEDCB) in K. pneumoniae affects multidrug resistance, underscoring the broader context of energy-dependent resistance mechanisms .
Recombinant K. pneumoniae ATP synthase subunit c is commercially available through specialized biotechnology suppliers.
Reconstitution: Suggested at 0.1–1.0 mg/mL in sterile water with 5–50% glycerol for long-term storage .
Storage: -20°C/-80°C, avoiding repeated freeze-thaw cycles .
KEGG: kpe:KPK_5539
ATP synthase subunit c (atpE) in K. pneumoniae is a membrane-spanning oligomeric protein that forms part of the F₀ domain of ATP synthase. This subunit is crucial for the flow of protons across the cytoplasmic membrane to facilitate ATP synthesis. The F₀ domain is embedded in the membrane and works in conjunction with the cytoplasmic F₁ domain to convert the energy from proton flow into ATP production .
Structurally, subunit c forms a ring-like structure within the membrane, which rotates during ATP synthesis. This rotation is coupled to proton translocation and drives conformational changes in the F₁ sector that enable ATP synthesis. Comparative analyses with E. coli have shown that the K. pneumoniae subunit c has only slight deviations in amino acid composition, suggesting highly conserved structural elements essential for its function .
To study its structure-function relationship, researchers commonly employ:
Site-directed mutagenesis to alter specific amino acids
Cryo-electron microscopy to visualize the protein in its native state
Chemical cross-linking studies to identify interaction partners
Molecular dynamics simulations to understand conformational changes
The recombinant K. pneumoniae ATP synthase subunit c is produced through heterologous expression systems, typically in E. coli, which may introduce several differences compared to the native form:
Feature | Native atpE | Recombinant atpE | Research Implications |
---|---|---|---|
Post-translational modifications | Contains natural bacterial modifications | May lack specific modifications depending on expression system | May affect functional studies |
Purity | Part of complete ATP synthase complex | Can be isolated at high purity | Enables specific binding and structural studies |
Tags | No artificial sequences | Often contains His, GST or other fusion tags | Facilitates purification but may affect function |
Folding | Native membrane-associated folding | Frequently forms inclusion bodies requiring refolding | Potential functional differences |
Oligomerization | Natural ring formation in membrane | May have altered oligomeric properties | Important consideration for structural studies |
When designing experiments with recombinant K. pneumoniae atpE, researchers should consider these differences, especially when studying interactions with potential inhibitors or analyzing structural properties .
Based on extensive research with ATP synthase components, several expression systems can be employed for recombinant K. pneumoniae atpE production, each with specific advantages:
E. coli expression system: Most commonly used due to its genetic tractability, rapid growth, and high protein yields. When expressing K. pneumoniae atpE in E. coli, consider the following:
Bacillus subtilis expression system: Useful when studying functional aspects, as Bacillus has been shown to be sensitive to ATP synthase inhibitors and may provide a more native-like membrane environment
Cell-free expression systems: Beneficial for membrane proteins like atpE, allowing direct incorporation into liposomes or nanodiscs
The methodological approach should include optimization of:
Induction conditions (temperature, inducer concentration, induction time)
Growth media composition (particularly important for membrane proteins)
Codon optimization for the expression host
Addition of membrane-mimetic environments during expression
Purification of recombinant K. pneumoniae ATP synthase subunit c presents challenges due to its hydrophobic nature and membrane localization. A systematic purification strategy involves:
Inclusion body isolation and solubilization:
Bacterial lysis through sonication or high-pressure homogenization
Washing inclusion bodies with detergents (typically 0.5-2% Triton X-100)
Solubilization using chaotropic agents (6-8M urea or 6M guanidine hydrochloride)
Affinity chromatography:
Immobilized metal affinity chromatography (IMAC) for His-tagged proteins
Detergent selection is critical (common choices: DDM, LDAO, or Fos-choline)
Refolding strategies:
Dialysis against decreasing concentrations of denaturant
Incorporation into liposomes or nanodiscs for functional studies
On-column refolding during affinity purification
Quality assessment:
To maintain function, researchers should consider incorporating the purified subunit c into proteoliposomes, which better mimic its native membrane environment and allow for functional assays measuring proton translocation or ATP synthesis activities.
Recombinant K. pneumoniae ATP synthase subunit c has emerged as a valuable tool in antibiotic discovery due to its critical role in bacterial energy metabolism and identification as a binding site for compounds like diarylquinolines . Researchers can implement the following methodological approaches:
High-throughput screening assays:
Develop binding assays using fluorescently labeled atpE or FRET-based approaches
Establish ATP synthesis inhibition assays using reconstituted ATP synthase containing recombinant atpE
Implement thermal shift assays to identify compounds that alter protein stability upon binding
Structure-based drug design:
Utilize purified recombinant atpE for co-crystallization studies with potential inhibitors
Perform computational docking studies against the atpE structure
Design rational modifications of existing inhibitors based on binding site interactions
Resistance mechanism studies:
Generate site-directed mutants of recombinant atpE corresponding to clinical resistance mutations
Evaluate binding affinities of compounds to wild-type versus mutant proteins
Conduct biochemical characterization of how mutations alter inhibitor interactions
Target validation approaches:
Develop assays comparing compound activity against purified ATP synthase versus whole bacterial cells
Perform competition binding studies between novel compounds and known atpE-binding molecules
Utilize genetic approaches with controlled expression of atpE to validate phenotypic effects
These methodologies leverage the research finding that the transmembrane F₀ part of ATP synthase, which includes subunit c, plays a crucial role in interaction with certain antibacterial compounds .
Based on established methodologies in ATP synthase research, several experimental approaches can identify and characterize resistance mutations in the K. pneumoniae atpE gene:
In vitro selection of resistant mutants:
Site-directed mutagenesis and complementation studies:
Generate a library of atpE mutants based on predicted binding sites or known resistance mutations
Express mutant genes in an atpE-deficient or knockdown strain
Evaluate resistance profiles through growth inhibition assays
Biochemical characterization of mutant proteins:
Express and purify recombinant mutant and wild-type atpE proteins
Compare binding affinities of inhibitors using surface plasmon resonance or isothermal titration calorimetry
Assess functional consequences of mutations on ATP synthesis activity
Structural analysis:
Perform comparative structural analysis of wild-type versus mutant proteins
Map mutations onto the three-dimensional structure to understand mechanism of resistance
Use molecular dynamics simulations to evaluate conformational changes induced by mutations
When analyzing potential resistance mutations, researchers should consider:
Conservation of the mutated residues across bacterial species
Proximity to predicted binding sites for inhibitors
Impact on protein stability and function
Cross-resistance patterns to different ATP synthase inhibitors
Comparative analyses between K. pneumoniae and E. coli ATP synthase subunit c reveal important structural and functional similarities that inform research approaches:
Structural similarities:
SDS-gel electrophoresis of purified F₁F₀ complexes demonstrates identical subunit patterns for both organisms
Protease digestion of individual subunits generates identical cleavage patterns for subunit c in both species
Slight deviations in amino acid composition have been observed, but these appear to have minimal functional impact
Immunological cross-reactivity:
Antibodies raised against E. coli F₁ complex and purified F₀ subunits recognize corresponding polypeptides in both E. coli and K. pneumoniae, indicating significant epitope conservation
This cross-reactivity enables the use of E. coli-derived antibodies for detection and purification of K. pneumoniae ATP synthase components
Functional conservation:
Feature | K. pneumoniae atpE | E. coli atpE | Research Implications |
---|---|---|---|
Protease digestion pattern | Identical to E. coli | Reference pattern | Can use same purification protocols |
Antibody cross-reactivity | Recognized by anti-E. coli antibodies | Source of antibodies | Enables immunological detection methods |
Amino acid composition | Slight deviations from E. coli | Reference composition | Minimal impact on functional studies |
Genetic organization | Similar operon structure | Well-characterized | Allows parallel genetic approaches |
These similarities facilitate the adaptation of established E. coli-based methodologies for studying K. pneumoniae ATP synthase subunit c, including expression systems, purification protocols, and functional assays .
When investigating the regulation of the atpE gene in K. pneumoniae, researchers should consider several methodological approaches based on recent findings about ATP synthase regulation:
Transcriptional regulation analysis:
The OmpR response regulator has been shown to directly bind the promoter region of the atp operon in K. pneumoniae, suggesting important regulatory control
Design chromatin immunoprecipitation (ChIP) experiments to identify additional transcription factors that bind the atp operon promoter
Perform reporter gene assays using the atp operon promoter region fused to fluorescent or enzymatic reporters
Environmental regulation studies:
Investigate expression changes under varying energy states, pH conditions, or oxygen availability
Analyze growth phase-dependent expression patterns using qRT-PCR
Evaluate the impact of different carbon sources on atpE expression levels
Post-transcriptional regulation:
Examine mRNA stability under different conditions
Investigate the role of small RNAs in regulating atpE expression
Analyze translation efficiency using ribosome profiling
Systems biology approaches:
Conduct transcriptomic analysis (RNA-Seq) under conditions where atpE expression is altered
Perform network analysis to identify genes co-regulated with atpE
Integrate transcriptomic and proteomic data to assess correlation between mRNA and protein levels
When designing these experiments, researchers should consider:
The polycistronic nature of the atp operon, which may complicate individual gene analysis
Potential polar effects when manipulating upstream genes in the operon
Cross-regulation between energy metabolism pathways
The role of two-component systems, particularly OmpR/EnvZ, in sensing environmental signals that influence atpE expression
To characterize the functional activity of recombinant K. pneumoniae ATP synthase subunit c, researchers can employ several complementary methodologies:
Reconstitution in proteoliposomes:
Purify recombinant atpE and reconstitute with other ATP synthase subunits
Measure ATP synthesis activity using luciferin/luciferase assays
Assess proton translocation using pH-sensitive fluorescent dyes
Quantify ATP hydrolysis activity through phosphate release assays
Membrane potential measurements:
Monitor membrane potential changes using potential-sensitive dyes like DiSC3(5)
Compare wild-type versus mutant atpE incorporation to assess functional impact
Measure effects of inhibitors on membrane potential in reconstituted systems
Drug binding assays:
Develop fluorescence-based binding assays using intrinsic tryptophan fluorescence
Perform isothermal titration calorimetry to obtain binding constants
Utilize surface plasmon resonance to measure binding kinetics of inhibitors to reconstituted atpE
Conduct thermal shift assays to assess stabilization upon inhibitor binding
Genetic complementation approaches:
Express K. pneumoniae atpE in heterologous systems with atpE deficiencies
Measure restoration of growth under conditions requiring ATP synthase function
Assess complementation efficiency with wild-type versus mutant atpE variants
When conducting these assays, researchers should control for:
Proper incorporation and orientation of atpE in membrane systems
Complete reconstitution of all necessary ATP synthase components
Buffer conditions that mimic physiological environments
Potential artifacts from fusion tags or expression system modifications
When evaluating inhibitor specificity for K. pneumoniae ATP synthase subunit c, researchers should implement a comprehensive assessment strategy:
Binding selectivity assays:
Mutational analysis:
Generate point mutations in key residues predicted to be involved in inhibitor binding
Measure binding affinities and inhibitory potencies against mutant proteins
Map resistance mutations to the three-dimensional structure to identify binding sites
Comparative species analysis:
Test inhibitor activity against ATP synthases from different bacterial species
Correlate inhibitory potency with sequence variations in the atpE gene
Assess activity against human mitochondrial ATP synthase to evaluate potential toxicity
Functional specificity:
Compare effects on ATP synthesis versus other ATP synthase functions
Conduct kinetic analyses to determine mechanism of inhibition
Measure impact on proton translocation to confirm F₀ domain targeting
Assay Type | Methodology | Expected Results for Specific Inhibitors | Limitations |
---|---|---|---|
Binding assays | Fluorescence polarization with labeled inhibitors | Displacement by known subunit c ligands | Requires labeled compounds |
Structural studies | Co-crystallization or NMR studies | Direct evidence of binding site | Technical challenges with membrane proteins |
Cellular assays | ATP depletion in bacterial cultures | Correlation with in vitro binding | Multiple targets may deplete ATP |
Resistance selection | Generation of resistant mutants | Mutations mapping to atpE gene | Resistance may involve other mechanisms |
For experimental design, consider that the F₀ part of ATP synthase plays a crucial role in interaction with certain chemical classes, as demonstrated for diarylquinolines, and binding specificity can be assessed by comparing whole ATP synthase complex versus isolated domains .
Recombinant K. pneumoniae ATP synthase subunit c serves as a powerful tool for investigating bacterial energy metabolism and antibiotic resistance through several methodological approaches:
Energy metabolism studies:
Down-regulation of atpE expression has been shown to dramatically impair bacterial growth, indicating its essential role in energy production
Researchers can employ controlled expression systems to modulate atpE levels and measure:
Changes in intracellular ATP concentrations
Shifts in metabolic pathways (through metabolomics)
Adaptations in respiratory chain components
Alterations in membrane potential and proton motive force
Antibiotic resistance mechanism investigation:
atpE mutations have been identified as resistance determinants for certain antibiotics
Research methodologies should include:
Creation of libraries of atpE variants through site-directed mutagenesis
Assessment of cross-resistance patterns between different ATP synthase inhibitors
Evaluation of fitness costs associated with resistance mutations
Investigation of compensatory mutations that restore fitness in resistant strains
Regulatory network analysis:
The OmpR response regulator directly binds the promoter region of the atp operon, suggesting regulatory connections between environmental sensing and energy metabolism
Experimental approaches include:
ChIP-seq to identify binding sites for regulatory factors
Reporter gene assays to quantify transcriptional responses
Analysis of how signals from two-component systems modulate ATP synthase expression
Bacterial adaptation studies:
ATP synthase inhibition creates significant metabolic stress, potentially triggering adaptive responses
Researchers can investigate:
Transcriptional responses to ATP synthase inhibition
Metabolic rewiring in response to energy limitation
Development of tolerance mechanisms distinct from target-based resistance
These approaches help elucidate the interconnection between energy metabolism and antibiotic resistance, particularly how bacteria adapt to perturbations in ATP synthesis and how these adaptations might influence antimicrobial susceptibility.
When investigating the relationship between K. pneumoniae ATP synthase subunit c and bacterial virulence, researchers should consider several experimental design elements:
Genetic manipulation approaches:
Complete deletion of atpE may be lethal, necessitating conditional knockdown systems:
In vitro virulence assays:
Assess the impact of atpE modulation on:
Biofilm formation capacity
Adhesion to epithelial cells
Resistance to host defense mechanisms
Production of virulence factors
Growth in various nutrient-limited conditions mimicking host environments
In vivo infection models:
Mouse pneumonia models have been used successfully to study K. pneumoniae virulence factors
Design considerations include:
Comparison of wild-type strains versus atpE-modulated strains
Assessment of bacterial burden in different tissues
Monitoring host immune responses
Evaluation of survival rates and disease progression
Testing efficacy of ATP synthase inhibitors in infection models
Integration with regulatory networks:
When designing these experiments, researchers should be aware that:
Perturbations in energy metabolism may have pleiotropic effects on multiple virulence traits
The relationship between ATP synthase function and virulence may vary depending on infection site
Host factors may influence the requirement for fully functional ATP synthase during infection
Environmental conditions during infection may alter ATP synthase expression and importance
Researchers working with recombinant K. pneumoniae ATP synthase subunit c frequently encounter several technical challenges that require specific troubleshooting approaches:
Expression challenges:
Problem: Low expression levels in heterologous systems
Solutions:
Problem: Toxicity to expression host
Solutions:
Implement tight expression control using repressible promoters
Reduce induction temperature (16-20°C)
Use lower inducer concentrations
Consider cell-free expression systems
Purification challenges:
Problem: Formation of inclusion bodies
Solutions:
Optimize solubilization conditions (detergent screening)
Develop effective refolding protocols
Co-express with chaperones
Test fusion partners that enhance solubility
Problem: Loss of structural integrity during purification
Solutions:
Determine optimal detergent for extraction and purification
Include stabilizing additives (glycerol, specific lipids)
Minimize exposure to harsh conditions
Verify structural integrity through circular dichroism
Functional reconstitution challenges:
Problem: Improper incorporation into membranes
Solutions:
Optimize lipid composition for reconstitution
Control protein-to-lipid ratios
Ensure correct orientation in proteoliposomes
Develop functional assays to verify activity after reconstitution
Quality control issues:
Problem: Heterogeneity in purified preparations
Solutions:
Implement rigorous size exclusion chromatography steps
Utilize analytical ultracentrifugation to assess oligomeric state
Apply mass spectrometry for detailed characterization
Develop activity assays to correlate with structural properties
Each of these challenges requires systematic optimization and validation to ensure that the recombinant protein maintains native-like properties relevant for subsequent experimental applications.
When investigating interactions between inhibitors and K. pneumoniae ATP synthase subunit c, researchers may encounter data inconsistencies that require careful analysis and troubleshooting:
Binding assay inconsistencies:
Problem: Discrepancies between different binding assay formats
Methodological solutions:
Employ multiple orthogonal binding assays (SPR, ITC, fluorescence-based)
Evaluate potential interference from buffer components or detergents
Control for non-specific binding to membranes or detergent micelles
Standardize protein preparation methods across experiments
Problem: Variable binding constants across experiments
Analytical approaches:
Ensure protein stability throughout binding studies
Control temperature and other experimental conditions rigorously
Account for potential ligand depletion effects
Validate binding stoichiometry through careful data fitting
Functional assay discrepancies:
Problem: Differences between biochemical and cellular inhibition potencies
Investigative strategies:
Assess compound permeability in cellular systems
Evaluate potential efflux mechanisms in whole cells
Determine compound stability under assay conditions
Consider potential off-target effects in cellular contexts
Problem: Inconsistent structure-activity relationships
Systematic approaches:
Verify compound purity and identity before each assay
Control for potential aggregation of inhibitors
Ensure consistent reconstitution of atpE in functional assays
Carefully analyze kinetic mechanisms of inhibition
Resistance mutation interpretation challenges:
Problem: Mutations that confer resistance without obvious structural explanation
Analytical methods:
Perform comprehensive molecular modeling of inhibitor binding
Consider allosteric effects that may not be immediately apparent
Evaluate effects of mutations on protein stability and oligomerization
Analyze potential changes in protein dynamics rather than just static structure
Species-specific differences:
Problem: Inhibitor efficacy varying between closely related bacterial species
Comparative approaches:
Conduct detailed sequence and structural alignments
Create chimeric proteins to identify critical regions for binding
Consider differences in membrane composition between species
Evaluate potential differences in drug uptake or efflux
By systematically addressing these challenges, researchers can develop more robust and reproducible methods for characterizing interactions between inhibitors and K. pneumoniae ATP synthase subunit c, ultimately leading to more reliable data for drug discovery applications.