Recombinant Rickettsia typhi ATP synthase subunit c (atpE) is a bioengineered protein derived from the atpE gene of Rickettsia typhi, a pathogen causing murine typhus. This protein represents the full-length subunit c of the bacterial F₀F₁-ATP synthase complex, which catalyzes ATP synthesis and proton transport across membranes . The recombinant version is expressed in E. coli with an N-terminal His tag for purification and structural studies .
The sequence begins with MDIVSLKFIGIGFMAIGMYGAALGVSNIFSSLLSAIARNPSAAENLQRMALIGAGLAEAMGLFAFVIAMLLIFS, highlighting conserved motifs critical for proton translocation and ATP synthesis .
Subunit c (atpE) forms part of the F₀ sector of the ATP synthase, facilitating proton transport across the inner mitochondrial membrane in eukaryotes. In Rickettsia typhi, this subunit likely supports the organism’s energy metabolism, enabling survival within host cells .
The recombinant protein is produced via E. coli expression systems, leveraging its soluble expression profile. Post-expression, purification involves:
Challenges include maintaining solubility during purification, though E. coli systems avoid the need for chaperonins required for other Rickettsia proteins like RPP .
Unlike surface antigens (e.g., rOmpA/B), atpE exhibits minimal diversification due to low selective pressure, indicating functional conservation across Rickettsia species . This contrasts with mammalian subunit c isoforms (P1/P2/P3), which differ in targeting peptides but share identical mature sequences .
Aspect | Observation |
---|---|
Proton Translocation | Subunit c oligomers form a proton channel, critical for ATP synthesis . |
Cofactor Dependence | No direct evidence reported; structural studies pending. |
Vaccine Development: While atpE is not a primary immune target , its structural analysis could inform novel therapeutic strategies.
Structural Biology: Crystallization studies to elucidate F₀ sector dynamics in Rickettsia ATP synthase.
Diagnostic Tools: Potential use in serological assays for Rickettsia typhi detection.
KEGG: rty:RT0106
STRING: 257363.RT0106
ATP synthase subunit c (atpE) in Rickettsia typhi is a critical component of the F-type ATP synthase complex, specifically within the FO membrane domain. This protein functions as part of the proton channel that catalyzes the production of ATP from ADP in the presence of a sodium or proton gradient across the membrane . The atpE protein forms the central rotor element (c-ring) of the ATP synthase complex, which is essential for the rotary mechanism during the catalytic process. In R. typhi, this 74-amino acid protein (UniProt ID: Q68XQ0) plays a vital role in energy metabolism during both active replication and potential dormancy states .
The R. typhi ATP synthase subunit c is a small, hydrophobic protein composed of 74 amino acids with the sequence: MDIVSLKFIGIGFMAIGMYGAALGVSNIFSSLLSAIARNPSAAENLQRMALIGAGLAEAMGLFAFVIAMLLIFS . Structurally, it contains membrane-spanning alpha helices that aggregate to form the c-ring, which constitutes the central rotor element of the F1F0 ATP synthase complex. The F0 domain typically consists of residues between positions 5-25 and 57-77 . The atpE protein is predominantly hydrophobic, allowing it to be inserted into the bacterial inner membrane where it participates in proton translocation. This protein's high level of conservation across species reflects its essential function in cellular bioenergetics.
Recombinant R. typhi atpE protein typically includes modifications to facilitate laboratory research, such as the addition of an N-terminal histidine tag for purification purposes . While the core amino acid sequence remains identical to the native protein, these modifications can alter certain biochemical properties:
When using recombinant atpE for functional studies, researchers should consider how these modifications might influence protein behavior compared to the native form.
When expressing R. typhi atpE in E. coli, researchers should use specialized vectors containing strong, inducible promoters, but maintain low induction levels to prevent toxicity from membrane protein overexpression. Codon optimization for E. coli expression is also recommended since Rickettsia has different codon usage patterns. Addition of fusion partners like thioredoxin or MBP can sometimes improve solubility beyond what is achieved with a simple His-tag .
Studying interactions between R. typhi atpE and other ATP synthase components requires specialized techniques that accommodate the hydrophobic nature of these proteins:
Co-expression strategies: Express atpE alongside other ATP synthase components in E. coli to promote proper complex formation. This approach has been successful for studying other bacterial ATP synthases.
Membrane reconstitution: Purified atpE can be reconstituted into liposomes with other ATP synthase components to study functional interactions. Monitoring proton translocation using pH-sensitive dyes or electrochemical methods provides insights into functional assembly.
Cross-linking studies: Chemical cross-linking followed by mass spectrometry analysis can identify specific residues involved in subunit interactions. This approach requires careful optimization of cross-linking conditions to avoid non-specific interactions.
Bacterial two-hybrid systems: Modified membrane-based two-hybrid systems can detect protein-protein interactions involving membrane proteins like atpE.
Cryo-EM analysis: Recent advances in cryo-electron microscopy have made it possible to visualize ATP synthase complexes at near-atomic resolution, providing structural insights into subunit interactions .
These approaches have complementary strengths and should be combined for comprehensive interaction studies.
R. typhi atpE presents a promising drug target due to several advantageous characteristics:
Essential function: As a critical component of ATP production machinery, targeting atpE can effectively inhibit bacterial energy metabolism .
Conservation and specificity: While atpE is conserved across bacterial species, sufficient structural differences exist between bacterial and mammalian ATP synthases to allow selective targeting.
Established precedent: ATP synthase inhibitors have proven effective against other pathogens. For example, bedaquiline targets mycobacterial ATP synthase with high specificity .
Alternative to current targets: AtpE provides an alternative path for antimicrobial development when resistance develops to drugs targeting other pathways, such as those involving the Sec secretion system .
Compared to other potential Rickettsia drug targets in the table below, atpE offers distinct advantages:
Drug development targeting atpE would likely focus on compounds that bind to the c-ring and disrupt proton translocation, similar to the mechanism of action for diarylquinolines against mycobacteria.
Purification of recombinant R. typhi atpE requires specialized protocols due to its hydrophobic nature:
Recommended Purification Protocol:
Membrane Fraction Isolation:
Harvest E. coli cells expressing His-tagged atpE
Disrupt cells via sonication or French press
Isolate membrane fraction through differential centrifugation
Solubilization:
Solubilize membrane fraction using detergents such as n-dodecyl-β-D-maltoside (DDM), LDAO, or Triton X-100
Critical step: maintain 5:1 detergent:protein ratio to prevent aggregation
Affinity Chromatography:
Bind solubilized protein to Ni-NTA resin
Wash with increasing imidazole concentrations (20-40 mM)
Elute with 250-500 mM imidazole buffer containing detergent
Size Exclusion Chromatography:
Further purify using gel filtration to separate monomeric protein from aggregates
Use buffer containing 0.05-0.1% detergent to maintain solubility
Reconstitution (if required):
Incorporate purified protein into liposomes for functional studies
Gradually remove detergent using Bio-Beads or dialysis
Proper storage is critical - lyophilized powder of purified atpE should be stored at -20°C/-80°C, with 6% trehalose as a cryoprotectant in Tris/PBS-based buffer (pH 8.0) . Working aliquots reconstituted in deionized water (0.1-1.0 mg/mL) should be supplemented with 5-50% glycerol and stored at 4°C for up to one week to avoid repeated freeze-thaw cycles .
Evaluating the functional activity of recombinant R. typhi atpE requires assessing both its ability to integrate into the ATP synthase complex and its proton translocation function:
Integration Assessment Methods:
Blue Native PAGE:
Determines if recombinant atpE can incorporate into ATP synthase complexes
Compare complexes from native Rickettsia and reconstituted systems
Proteoliposome Reconstitution Assays:
Reconstitute atpE with other ATP synthase components in liposomes
Measure ATP synthesis driven by artificially imposed proton gradients
Compare activity to known standards using luciferase-based ATP detection
Proton Translocation Assays:
Load proteoliposomes with pH-sensitive fluorescent dyes (ACMA or pyranine)
Monitor fluorescence changes upon energization
Calculate proton flux rates under various conditions
Complementation Studies:
Binding Studies with Known Inhibitors:
Use thermal shift assays or surface plasmon resonance to measure binding of known ATP synthase inhibitors
Compare binding profiles with characterized ATP synthase proteins
A comprehensive functional assessment should combine multiple methods to overcome the limitations of individual approaches.
Computational analysis provides crucial insights into R. typhi atpE structure and function:
Recommended Bioinformatics Pipeline:
Sequence Analysis:
Multiple sequence alignment using MUSCLE or T-Coffee to compare atpE across Rickettsia species
Conservation analysis with ConSurf to identify functionally important residues
Hydropathy plots using TMHMM or HMMTOP to predict transmembrane regions
Structure Prediction:
Functional Analysis:
Evolutionary Analysis:
Detection of selection pressure using PAML
Coevolution analysis to identify residue networks using PSICOV
These computational approaches should guide experimental design by identifying key residues for mutagenesis and potential binding sites for inhibitor development.
Researchers frequently encounter several obstacles when working with recombinant R. typhi atpE:
For particularly challenging preparations, consider native-like nanodiscs or amphipols as alternatives to conventional detergent solubilization. These approaches often better preserve functional properties of membrane proteins like atpE.
Distinguishing specific from non-specific effects is critical in antimicrobial research targeting atpE:
Recommended Control Experiments:
Mutational Analysis:
Generate R. typhi atpE variants with mutations in predicted binding sites
Compare antimicrobial efficacy against wild-type and mutant proteins
Specific inhibitors show reduced activity against mutants with alterations at binding sites
Competitive Binding Assays:
Perform displacement studies with known ATP synthase ligands
Specific inhibitors compete for the same binding sites
Cross-species Comparisons:
Test compounds against ATP synthase from related Rickettsia species
Compare with more distant bacterial species and mammalian ATP synthase
Specific inhibitors show activity patterns that correlate with sequence conservation
Direct Binding Measurements:
Use isothermal titration calorimetry or microscale thermophoresis
Determine binding constants and thermodynamic parameters
Specific interactions typically exhibit saturable binding with nanomolar to micromolar affinity
Functional Rescue Experiments:
Overexpress atpE in bacterial cells
Specific inhibitors show reduced efficacy in overexpression systems
Test if exogenous ATP supplementation rescues growth inhibition
Time-kill Kinetics:
Compare kill curves with other ATP synthase inhibitors
Similar kinetic profiles suggest shared mechanisms
These approaches collectively provide strong evidence for specific targeting of atpE rather than generalized membrane disruption or other non-specific effects.
While traditionally considered a drug target, R. typhi atpE also holds potential in vaccine development:
T-cell Epitope Identification:
Cross-protection Potential:
Combination Vaccine Strategies:
Delivery Systems Development:
Test various platforms including DNA vaccines, viral vectors, and protein subunit approaches
Evaluate adjuvants that enhance T-cell responses
Compare immune responses to different forms of atpE presentation
Predictive Immunoinformatics:
This research could potentially address the current lack of commercially available anti-Rickettsia vaccines, providing protection against these highly pathogenic organisms .
Several cutting-edge technologies hold promise for furthering our understanding of R. typhi atpE:
Cryo-Electron Microscopy (Cryo-EM):
Recent advances enable visualization of membrane protein complexes at near-atomic resolution
Could reveal the exact arrangement of atpE within the ATP synthase complex
May identify species-specific structural features for drug targeting
AlphaFold2 and Related AI Systems:
Deep learning approaches for protein structure prediction
Can model protein complexes and interactions
Particularly valuable for membrane proteins like atpE that are challenging for experimental structure determination
Single-Molecule Techniques:
FRET and optical tweezers to study rotary dynamics of ATP synthase
Direct observation of conformational changes during catalysis
Real-time monitoring of inhibitor effects on rotation
Native Mass Spectrometry:
Analysis of intact membrane protein complexes
Determination of subunit stoichiometry and stability
Identification of lipid and small molecule interactions
Microfluidic Systems:
High-throughput screening of atpE inhibitors
Rapid assessment of antimicrobial effects on engineered bacterial systems
Combination with live cell imaging for real-time activity monitoring
CRISPR-Based Approaches:
Generation of conditional knockdowns to study atpE function
Creation of reporter systems for ATP synthase activity
Precise genome editing to introduce mutations for structure-function studies
Integration of these technologies would provide unprecedented insights into the molecular mechanisms of R. typhi atpE and facilitate rational drug design targeting this essential protein.
Understanding how R. typhi atpE function varies across conditions is crucial for developing effective interventions:
Key Research Questions for Conditional Function:
Host Cell Adaptation:
How does atpE expression and function change during intracellular growth in host cells?
Are there post-translational modifications that regulate activity in response to host conditions?
Does the c-ring stoichiometry remain constant or adapt to different energy requirements?
Stress Response Mechanisms:
How does atpE respond to nutrient limitation, oxidative stress, or antibiotic pressure?
Is atpE part of a regulated stress response network in Rickettsia?
Can ATP synthase reverse its function under certain conditions to maintain membrane potential?
Dormancy and Persistence:
Methodological Approaches:
Develop reporter systems to monitor ATP synthase activity in living cells
Employ quantitative proteomics to measure atpE abundance across conditions
Use metabolomics to correlate ATP synthase activity with metabolic states
This research direction is particularly important for understanding how to target R. typhi during different stages of infection, potentially leading to more effective therapeutic strategies.