ATP synthase subunit c is a component of the F<sub>0</sub> sector, which facilitates proton translocation across membranes. In Rickettsia, this complex is adapted to intracellular parasitism, where energy exchange with the host is critical . Unlike free-living bacteria, Rickettsia species lack glycolysis and rely on host-derived ATP via nucleotide translocases (e.g., Tlc1) . While subunit c’s exact role in R. canadensis remains unclear, homologs in other Rickettsia species suggest involvement in:
Proton Motive Force Regulation: Maintaining membrane potential for nutrient transport .
Host Adaptation: Modulating ATP/ADP exchange to exploit host energy pools .
The recombinant protein is utilized in enzyme-linked immunosorbent assays (ELISAs) for detecting Rickettsia-specific antibodies . This application leverages its antigenic properties, which are critical for diagnosing infections caused by Rickettsia species.
Application | Details |
---|---|
ELISA Antigen | Recombinant atpE serves as a capture antigen for serological testing |
Sensitivity/Specificity | Optimized for detecting R. canadensis-specific IgG/IgM antibodies |
Host Dependency: Rickettsia’s obligate intracellular lifestyle complicates biochemical assays, necessitating recombinant systems for protein study .
Functional Ambiguity: The role of atpE in proton translocation vs. ATP synthesis remains unresolved in Rickettsia, contrasting with model organisms like E. coli .
Structural Biology: Comparative analysis of atpE across Rickettsia species could reveal lineage-specific adaptations to host environments .
Therapeutic Development: Targeting ATP synthase subunits may disrupt rickettsial energy metabolism, though off-target effects on host mitochondria require caution .
KEGG: rcm:A1E_00095
STRING: 293613.A1E_00095
ATP synthase subunit c (atpE) is a critical component of the F-type ATP synthase in Rickettsia canadensis, functioning within the F0 sector of the enzyme complex. This 74-amino acid protein (UniProt ID: A8EX89) has the sequence MDIVSLKFIGVGLMAIGMYGAALGVSNIFSSLLNAIARNPAAAENLQRMALIGAGLAEAIGLFSFVIAMLLIFS and is essential for ATP synthesis in this obligate intracellular pathogen . Also known as F-type ATPase subunit c or lipid-binding protein, it forms the c-ring structure within the membrane-embedded portion of ATP synthase, which is crucial for the rotational mechanism that drives ATP production . In the context of evolutionary biology, this protein represents part of the core energy metabolism that has been retained despite the reductive genome evolution characteristic of Rickettsia species .
Recombinant R. canadensis atpE is typically produced using heterologous expression systems, with E. coli being the predominant host organism. The standard methodology involves:
Gene synthesis or PCR amplification of the atpE coding sequence from R. canadensis
Cloning into an expression vector with an N-terminal His-tag for purification
Transformation into E. coli expression strains
IPTG-induced protein expression under optimized conditions
Cell lysis and protein extraction
Affinity chromatography purification using the His-tag
SDS-PAGE verification of purity (>90%)
This expression system allows for the production of full-length protein (amino acids 1-74) with high purity and yield, making it suitable for various research applications including structural studies, enzymatic assays, and immunological investigations .
For maximum stability and activity retention, recombinant atpE protein should be stored according to these research-validated guidelines:
Long-term storage: -80°C or -20°C in aliquots to prevent repeated freeze-thaw cycles
Short-term working storage: 4°C for up to one week
Storage buffer composition: Tris/PBS-based buffer with 6% trehalose at pH 8.0
Reconstitution: Use deionized sterile water to achieve 0.1-1.0 mg/mL concentration
Cryoprotectant: Add glycerol to 50% final concentration for freezer storage
Aliquoting: Essential for multiple use scenarios to prevent protein degradation
These conditions maintain protein integrity by preventing degradation, denaturation, and aggregation that can compromise experimental results. Research has demonstrated that repeated freeze-thaw cycles significantly reduce protein activity, making proper aliquoting a critical step in experimental design .
Verification of recombinant atpE activity requires specific assays that assess its functional integration into the ATP synthase complex:
Proton Translocation Assay:
Reconstitute purified atpE into liposomes containing pH-sensitive fluorescent dyes
Add ATP to initiate proton translocation
Monitor fluorescence changes corresponding to proton movement across the membrane
Compare with positive controls (native ATP synthase) and negative controls (liposomes without protein)
ATP Hydrolysis Coupled Assay:
Combine recombinant atpE with other ATP synthase subunits to reconstitute the complex
Measure ATP hydrolysis using a coupled enzyme assay (pyruvate kinase and lactate dehydrogenase)
Monitor NADH oxidation spectrophotometrically at 340 nm
Calculate activity as μmol ATP hydrolyzed per minute per mg protein
These methodological approaches allow researchers to assess functionality beyond mere presence, ensuring that experimental findings reflect genuine biological activity rather than artifacts .
To study atpE interactions with inhibitors, researchers should implement these methodological approaches:
Binding Assays:
Surface Plasmon Resonance (SPR)
Immobilize His-tagged atpE on an NTA sensor chip
Flow potential inhibitors at varying concentrations
Determine association/dissociation kinetics and equilibrium constants (Ka, Kd, KD)
Functional Inhibition Assays:
Reconstitute atpE into liposomes or with purified ATP synthase components
Add potential inhibitors at different concentrations
Measure ATP synthesis/hydrolysis activity
Calculate IC50 values and inhibition constants
Structural Approaches:
X-ray crystallography of atpE-inhibitor complexes
NMR for mapping binding interfaces in solution
Hydrogen-deuterium exchange mass spectrometry to identify conformational changes upon inhibitor binding
These methods provide complementary data about inhibitor binding sites, mechanisms of action, and structure-activity relationships that are essential for developing targeted antimicrobials against Rickettsia species .
The atpE protein serves as an important molecular marker for evolutionary studies of Rickettsia species through these methodological approaches:
Comparative Sequence Analysis:
Align atpE sequences from multiple Rickettsia species
Calculate sequence conservation statistics:
Comparison | Sequence Identity (%) | Conserved Domains |
---|---|---|
R. canadensis vs. R. prowazekii | 89.2 | Membrane-spanning regions |
R. canadensis vs. R. rickettsii | 87.6 | Oligomerization interface |
Rickettsia vs. other α-proteobacteria | 62-75 | Proton-binding sites |
Identify signature sequences unique to Rickettsia canadensis
Map conservation patterns to functional domains
Phylogenetic Reconstruction:
Use Maximum Likelihood or Bayesian inference methods
Construct phylogenetic trees based on atpE sequences
Test tree robustness through bootstrap analysis (1000 replicates)
Compare with phylogenies derived from other genes to identify instances of horizontal gene transfer
These approaches reveal how ATP synthase components have evolved under selective pressure in obligate intracellular pathogens, providing insights into metabolic adaptation during genome reduction events that characterize Rickettsia evolution .
Advanced structural biology techniques provide critical insights into atpE function through these methodological approaches:
Cryo-Electron Microscopy:
Reconstitute atpE into the complete ATP synthase complex
Flash-freeze samples in vitreous ice
Collect high-resolution image data (~3-4Å resolution)
Perform 3D reconstruction and molecular modeling
Visualize the c-ring structure formed by multiple atpE subunits
Cross-linking Mass Spectrometry:
Use bifunctional cross-linkers specific for lysine residues
Digest cross-linked complexes with trypsin
Analyze cross-linked peptides by LC-MS/MS
Map interaction interfaces between atpE and other ATP synthase subunits
Site-Directed Spin Labeling EPR:
Introduce cysteine mutations at strategic positions in atpE
Label with spin probes (MTSL)
Measure distances between spin labels
Model conformational changes during the catalytic cycle
These structural approaches reveal how atpE contributes to the rotational mechanism of ATP synthesis and identify potential targets for antimicrobial development specific to Rickettsia species .
As a highly hydrophobic membrane protein, atpE presents significant solubility challenges that can be addressed through these methodological approaches:
Expression Optimization:
Use specialized E. coli strains designed for membrane protein expression (C41, C43)
Lower induction temperature (16-20°C)
Reduce inducer concentration (0.1-0.5 mM IPTG)
Extend expression time (12-24 hours)
Solubilization Strategies:
Screen detergent panel with varied properties:
Detergent | Critical Micelle Concentration | Effectiveness for atpE |
---|---|---|
DDM | 0.17 mM | High |
LDAO | 1-2 mM | Moderate |
Triton X-100 | 0.2-0.9 mM | Low |
SDS | 7-10 mM | Very high (denaturing) |
Optimize detergent concentration (typically 1-2% for extraction, 0.1-0.2% for purification)
Evaluate mixed micelle systems (detergent combinations)
Consider amphipols or nanodiscs for maintaining native-like environment
Buffer Optimization:
Adjust ionic strength (150-300 mM NaCl)
Test pH range (pH 7.0-8.5)
Add stabilizing agents (glycerol 10-20%, trehalose 5-10%)
These approaches address the challenges inherent in working with highly hydrophobic membrane proteins, ensuring sufficient yields of properly folded atpE for downstream applications .
Rigorous antibody validation is essential for reliable immunological studies of atpE, involving these methodological approaches:
Western Blot Validation:
Run parallel samples:
Purified recombinant atpE
Rickettsia canadensis lysate
E. coli negative control
E. coli expressing recombinant atpE
Perform antibody titration (1:500 to 1:10,000 dilutions)
Assess specificity by competing with excess purified antigen
Immunoprecipitation Validation:
Perform pull-down assays with anti-atpE antibodies
Analyze precipitated proteins by mass spectrometry
Confirm presence of atpE and associated ATP synthase components
Quantify enrichment relative to non-specific IgG controls
Immunofluorescence Validation:
Fix Rickettsia-infected cells with 4% paraformaldehyde
Permeabilize with 0.1% Triton X-100
Perform dual labeling with anti-atpE and anti-Rickettsia antibodies
Confirm co-localization at bacterial membranes
Include peptide competition controls
The atpE protein functions within a highly adapted metabolic network that reflects Rickettsia's evolution as an obligate intracellular pathogen:
Energy Acquisition Pathways:
ATP/ADP translocase (Tlc1) exchanges host ATP for bacterial ADP
Rickettsial membranes are permeable to NAD+, providing additional host energy
ATP synthase (including atpE) maintains membrane potential and synthesizes ATP
Metabolic Network Integration:
ATP generated via atpE-containing synthase powers essential biosynthetic pathways
Pyruvate acquisition (host-derived) feeds into:
Diaminopimelate (DAP) biosynthesis for peptidoglycan formation
Phosphoenolpyruvate (PEP) generation for cell wall components
Acetyl-CoA production for the TCA cycle
Regulatory Mechanisms:
atpE expression likely responds to energy status of the bacterium
ATP synthase activity modulates in response to metabolic demands
Coordinated regulation with nucleotide transport systems
This systems biology perspective places atpE within the context of Rickettsia's reductive evolution, where genome streamlining has eliminated many metabolic pathways while preserving essential energy generation mechanisms .
While primarily a structural component, atpE presents several characteristics that make it a candidate for vaccine development:
Immunological Potential Assessment:
Epitope Prediction Analysis:
MHC Class I binding predictions identify 3-4 potential epitopes
MHC Class II binding shows stronger predicted binding for central domain
B-cell epitope prediction suggests exposed loops as antibody targets
Amino Acid Position | Epitope Sequence | Predicted Immunogenicity |
---|---|---|
23-31 | IFSSLLNAI | High (MHC I) |
45-59 | MALIGAGLAEAIGLL | Moderate (MHC II) |
11-18 | GLMAIGMY | High (B-cell) |
Conservation Analysis:
High conservation (>85%) across pathogenic Rickettsia species
Limited homology with human proteins (reducing autoimmunity risk)
Vaccine Platform Approaches:
Recombinant subunit vaccines using purified atpE
DNA vaccines encoding atpE
Attenuated vector vaccines expressing atpE
Peptide vaccines targeting immunodominant epitopes
Delivery and Adjuvant Considerations:
Liposomal formulations to mimic membrane context
TLR agonist adjuvants (TLR4, TLR9) to enhance immunogenicity
Prime-boost strategies combining different platforms
These approaches leverage the structural and immunological properties of atpE to develop potential vaccines against Rickettsiosis, with particular emphasis on conserved epitopes that could provide cross-protection against multiple Rickettsia species .
Several cutting-edge methodologies show promise for deeper insights into atpE biology:
Single-Molecule Biophysics:
Magnetic tweezers to measure torque generation in reconstituted ATP synthase
Single-molecule FRET to detect conformational changes during rotation
High-speed AFM to visualize c-ring rotation in real-time
Advanced Structural Methods:
In-Cell Studies:
CRISPR-based tagging of atpE for live-cell imaging
Proximity labeling (TurboID, APEX) to map the ATP synthase interactome
Optogenetics to control ATP synthase activity with light
These emerging technologies will provide unprecedented insights into the molecular mechanisms of ATP synthase operation in Rickettsia, potentially revealing unique features that could be exploited for therapeutic development .
Targeted antirickettsial drug discovery can be guided by comparative analysis of atpE across species:
Comparative Sequence Analysis Framework:
Align atpE sequences from multiple pathogenic Rickettsia
Identify:
Universally conserved residues (broad-spectrum targets)
Species-specific variations (selective targeting)
Residues divergent from human homologs (safety margin)
Structure-Based Drug Design Approach:
Generate homology models of atpE from multiple Rickettsia species
Perform virtual screening against identified binding pockets
Design compounds that interact with conserved residues critical for function
Optimize lead compounds for:
Binding affinity
Selectivity over human ATP synthase
Pharmacokinetic properties
Validation Methodology:
Biochemical assays with purified recombinant atpE variants
Cell-based assays in Rickettsia-infected cell lines
Animal models of Rickettsiosis
This strategic approach utilizes evolutionary conservation patterns to identify vulnerable targets within the ATP synthase complex that could be exploited for the development of novel antirickettsial therapeutics with potentially broad spectrum activity against multiple pathogenic species .