Recombinant Rickettsia africae ATP synthase subunit c (atpE)

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: We prioritize shipping the format we have in stock. However, if you have specific format requirements, please specify them in your order. We will fulfill your request whenever possible.
Lead Time
Delivery time may vary based on the purchasing method and location. For specific delivery times, please consult your local distributors.
Note: All our proteins are shipped with standard blue ice packs. If you require dry ice shipping, please inform us in advance. Additional fees will apply.
Notes
Repeated freezing and thawing is not recommended. Store working aliquots at 4°C for up to one week.
Reconstitution
We recommend centrifuging the vial briefly before opening to ensure the contents settle at the bottom. Reconstitute the protein in deionized sterile water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our default final concentration of glycerol is 50%. You can use this as a reference.
Shelf Life
The shelf life is influenced by several factors, including storage conditions, buffer components, storage temperature, and the protein's inherent stability.
Generally, the shelf life of liquid form is 6 months at -20°C/-80°C. The shelf life of lyophilized form is 12 months at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is necessary for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type will be determined during the manufacturing process.
The tag type is decided during production. If you have a specific tag type preference, please inform us, and we will prioritize developing the specified tag.
Synonyms
atpE; RAF_ORF0024; ATP synthase subunit c; ATP synthase F(0 sector subunit c; F-type ATPase subunit c; F-ATPase subunit c; Lipid-binding protein
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-74
Protein Length
full length protein
Species
Rickettsia africae (strain ESF-5)
Target Names
atpE
Target Protein Sequence
MDMVSLKFIGTGLMAIGMYGAALGVSNIFSSLLSSIARNPSATENLQRMALIGAGLTEAM GLFSFVIAMLLIFS
Uniprot No.

Target Background

Function
F(1)F(0) ATP synthase generates ATP from ADP in the presence of a proton or sodium gradient. F-type ATPases consist of two structural domains: F(1), containing the extramembraneous catalytic core, and F(0), containing the membrane proton channel. These domains are connected by a central stalk and a peripheral stalk. During catalysis, ATP synthesis in the catalytic domain of F(1) is coupled to proton translocation through a rotary mechanism of the central stalk subunits. The c-ring, a homomeric structure of 10-14 subunits, forms the central stalk rotor element, interacting with the F(1) delta and epsilon subunits. It plays a crucial role in transmembrane translocation.
Database Links
Protein Families
ATPase C chain family
Subcellular Location
Cell membrane; Multi-pass membrane protein.

Q&A

What is the structural and functional significance of ATP synthase subunit c (atpE) in Rickettsia africae?

ATP synthase subunit c (atpE) in Rickettsia africae is a critical component of the F0F1 ATP synthase complex. This protein functions as part of the membrane-embedded F0 sector, forming the c-ring structure that facilitates proton translocation across the membrane. The c-ring rotation couples the proton motive force to ATP synthesis in the F1 sector.

In obligate intracellular pathogens like Rickettsia africae, the ATP synthase complex is particularly important because these organisms have limited metabolic capabilities and rely heavily on oxidative phosphorylation for energy production. The atpE protein contains hydrophobic regions that anchor it within the membrane and a conserved carboxylate residue essential for proton binding and translocation .

How does atpE function in the context of Rickettsia's limited metabolic capabilities?

Despite lacking a complete glycolytic pathway, Rickettsia africae maintains a functional tricarboxylic acid (TCA) cycle and oxidative phosphorylation system in which atpE plays a crucial role. Experimental protocols to study this function typically involve:

  • Comparative metabolic flux analysis using isotope-labeled glutamate and measuring ATP production rates

  • Membrane potential measurements using fluorescent probes like JC-1 or TMRM

  • Oxygen consumption rate (OCR) measurements in infected cells versus controls

Rickettsia utilizes host-acquired glutamate as a primary energy source, which feeds into the TCA cycle. The resulting NADH and FADH2 fuel the electron transport chain, generating a proton gradient across the membrane that drives ATP synthesis via the F0F1 ATP synthase complex containing atpE . This process is essential for bacterial survival, as Rickettsia also possesses an ATP/ADP symporter (Tlc1) that exchanges host ATP for bacterial ADP, indicating the critical importance of energy metabolism for this pathogen .

What expression systems are most effective for producing functional recombinant Rickettsia africae atpE protein?

When expressing recombinant Rickettsia africae atpE protein, researchers should consider these methodological approaches based on the protein's hydrophobic nature:

  • E. coli expression systems:

    • BL21(DE3) strains with pET vectors incorporating a C-terminal His-tag

    • Codon optimization for E. coli is essential due to GC content differences (32.4% in R. africae vs. ~50% in E. coli)

    • Induction at lower temperatures (16-20°C) to minimize inclusion body formation

    • Addition of membrane-stabilizing agents (e.g., 5% glycerol) to the culture medium

  • Cell-free expression systems:

    • Particularly useful for membrane proteins like atpE

    • Incorporation of detergents or nanodiscs to stabilize the hydrophobic regions

  • Baculovirus expression systems:

    • Preferred for maintaining proper folding of membrane proteins

    • Higher yield of functional protein compared to bacterial systems

When purifying the expressed protein, a two-step purification protocol involving immobilized metal affinity chromatography followed by size exclusion chromatography in buffers containing appropriate detergents (e.g., n-dodecyl-β-D-maltoside) has proven most effective for maintaining protein stability and function .

How can researchers effectively use recombinant atpE protein to study rickettsia-host energetic interactions?

Investigating rickettsia-host energetic interactions requires sophisticated experimental approaches using recombinant atpE:

  • Reconstitution experiments:

    • Purified recombinant atpE can be reconstituted into liposomes with other ATP synthase components

    • Proton pumping can be measured using pH-sensitive fluorescent dyes

    • ATP synthesis rates can be quantified under varying conditions

  • Protein-protein interaction studies:

    • Use recombinant atpE as bait in pull-down assays to identify host factors that may interact with the ATP synthase complex

    • Employ chemical crosslinking followed by mass spectrometry to map interaction interfaces

    • Validate interactions using surface plasmon resonance or microscale thermophoresis

  • Metabolic flux analysis:

    • Compare energy metabolism in cells infected with wild-type Rickettsia versus atpE mutants

    • Use 13C-labeled substrates to track metabolic pathways

The biological significance of these investigations lies in understanding how Rickettsia africae, which lacks glycolysis, can efficiently harvest energy from the host. Studies have demonstrated that Rickettsia oxidizes glutamate to drive electron transport coupled to oxidative phosphorylation, with the generated ATP facilitating import of other essential nutrients like proline and lysine . This metabolic dependency represents a potential therapeutic target.

What are the most effective methods for analyzing the interaction between atpE and diphtheria toxin resistance protein (DtxR) in Rickettsia africae?

Analyzing atpE-DtxR interactions requires multiple complementary approaches:

  • Binding assays methodology:

    • Express both proteins with different tags (His and GST)

    • Perform reciprocal co-immunoprecipitation experiments

    • Use isothermal titration calorimetry to determine binding kinetics

    • Employ fluorescence resonance energy transfer (FRET) to visualize interactions in vitro

  • Structural analysis workflow:

    • Generate protein-protein docking models based on known structures

    • Verify interaction sites through site-directed mutagenesis

    • Attempt co-crystallization for X-ray crystallography

    • Use cryo-electron microscopy for structural determination of the complex

  • Functional validation:

    • Develop Rickettsia strains with mutations in the predicted interaction sites

    • Assess changes in proton translocation efficiency

    • Measure ATP production in the presence of wild-type vs. mutant proteins

This interaction is particularly significant because DtxR proteins typically function as metal-dependent transcriptional regulators, and their potential interaction with ATP synthase components suggests a novel regulatory mechanism linking metal homeostasis with energy production in these specialized intracellular pathogens .

What techniques are most reliable for characterizing the conformational changes in atpE during the catalytic cycle of ATP synthesis?

The conformational dynamics of atpE during ATP synthesis can be characterized using:

  • Single-molecule FRET:

    • Introduce fluorescent labels at specific sites in recombinant atpE

    • Monitor distance changes between labels during proton translocation

    • Develop a workflow using total internal reflection fluorescence microscopy

  • Hydrogen-deuterium exchange mass spectrometry (HDX-MS):

    • Compare hydrogen-deuterium exchange rates in different catalytic states

    • Identify regions with altered solvent accessibility

    • Quantify the timing of conformational changes

  • Electron paramagnetic resonance (EPR) spectroscopy:

    • Introduce spin labels at strategic positions in atpE

    • Measure distances between labels in different functional states

    • Correlate structural changes with functional parameters

  • Molecular dynamics simulations:

    • Build atomic models of atpE within the c-ring

    • Simulate proton binding and release events

    • Predict energy barriers and rate-limiting steps

These techniques have revealed that the c-ring rotation in ATP synthase occurs in discrete steps, with each step corresponding to proton binding/release events. In Rickettsia africae, which relies heavily on oxidative phosphorylation for energy production, understanding these conformational dynamics is crucial for identifying potential species-specific inhibitors that could serve as novel antimicrobials .

What are the best approaches for studying the role of atpE in doxycycline resistance mechanisms in Rickettsia africae?

To investigate atpE's role in doxycycline resistance in Rickettsia africae, implement this systematic research approach:

  • Mutational analysis protocol:

    • Generate site-directed mutations in recombinant atpE at residues predicted to interact with doxycycline

    • Express and purify mutant proteins

    • Compare binding affinities using isothermal titration calorimetry

    • Develop a structural model of the interaction

  • Functional assays:

    • Reconstitute wild-type and mutant atpE proteins into liposomes

    • Measure proton translocation in the presence of varying doxycycline concentrations

    • Quantify ATP synthesis rates under similar conditions

  • In vivo validation:

    • Generate Rickettsia strains with the identified mutations

    • Determine minimum inhibitory concentrations of doxycycline

    • Assess fitness costs of resistance mutations

    • Monitor growth kinetics in the presence of subinhibitory antibiotic concentrations

This research is particularly important because doxycycline remains a first-line treatment for rickettsioses, and understanding resistance mechanisms is crucial. Recent studies suggest that mutations in ATP synthase components might contribute to reduced doxycycline susceptibility by altering membrane potential or drug efflux capabilities .

How can researchers effectively design experiments to investigate the assembly of the complete ATP synthase complex using recombinant atpE from Rickettsia africae?

Investigating ATP synthase assembly requires a comprehensive experimental design:

  • Sequential reconstitution methodology:

    • Express and purify all ATP synthase components individually

    • Add components in different orders to determine the assembly pathway

    • Monitor complex formation using native gel electrophoresis

    • Verify functionality at each step by measuring ATP synthesis or hydrolysis

  • Interaction mapping protocol:

    • Perform crosslinking mass spectrometry to identify contact sites between subunits

    • Generate subcomplex structures using cryo-electron microscopy

    • Develop an assembly map with defined intermediate states

  • In vivo fluorescence approaches:

    • Create fluorescent protein fusions to track assembly in living cells

    • Use fluorescence recovery after photobleaching (FRAP) to measure assembly kinetics

    • Implement super-resolution microscopy to visualize subcellular localization

  • Data analysis and integration:

    Assembly StageComponentsDetection MethodRate-Limiting StepValidation Approach
    c-ring formationatpE monomersNative PAGE, EMOligomerizationCrosslinking, FRET
    F0 assemblyc-ring + a, b subunitsBlue native PAGEa-subunit incorporationProtease protection
    F0F1 complexF0 + F1 sectorsATP synthesis assayF1 docking to F0Inhibitor studies

This research is particularly relevant for Rickettsia because, as an obligate intracellular pathogen with limited metabolic capabilities, proper assembly of the ATP synthase complex is essential for survival and pathogenesis .

What are the major technical challenges in purifying active recombinant Rickettsia africae atpE protein, and how can they be overcome?

Purifying active atpE presents several technical challenges with specific methodological solutions:

  • Challenge: Membrane protein solubility

    • Solution: Implement a systematic detergent screening approach:

      • Test a panel of 8-12 detergents (ionic, non-ionic, and zwitterionic)

      • Optimize detergent concentration through small-scale extractions

      • Consider styrene maleic acid lipid particles (SMALPs) as an alternative to detergents

      • Use fluorescence-detection size exclusion chromatography to assess protein stability

  • Challenge: Low expression yields

    • Solution: Optimize expression conditions through:

      • Fusion to solubility-enhancing tags (MBP, SUMO)

      • Reduced induction temperature (16-20°C)

      • Co-expression with chaperones (GroEL/GroES)

      • Use of specialized E. coli strains (C41/C43) designed for membrane protein expression

  • Challenge: Maintaining native conformation

    • Solution: Develop stabilization strategies:

      • Include lipids during purification (E. coli polar lipids or synthetic mixtures)

      • Add specific substrates or inhibitors during purification

      • Implement on-column refolding protocols if necessary

  • Challenge: Functional verification

    • Solution: Establish activity assays:

      • Reconstitution into proteoliposomes for proton translocation measurements

      • Incorporation into nanodiscs for structural studies

      • Circular dichroism to verify secondary structure content

When purified correctly, recombinant atpE should maintain its alpha-helical structure (typically >70% alpha-helical) and demonstrate the ability to oligomerize into ring-like structures when viewed by negative-stain electron microscopy .

What analytical techniques are most suitable for characterizing the lipid interactions of recombinant Rickettsia africae atpE protein?

For characterizing atpE-lipid interactions, implement these analytical approaches:

  • Lipid binding assays:

    • Develop a liposome flotation assay protocol using sucrose gradients

    • Perform isothermal titration calorimetry with different lipid compositions

    • Use microscale thermophoresis to measure binding affinities

    • Implement monolayer surface pressure measurements

  • Structural techniques:

    • Perform hydrogen-deuterium exchange mass spectrometry to identify lipid-protected regions

    • Use solid-state NMR to determine specific lipid-protein contacts

    • Employ electron paramagnetic resonance with spin-labeled lipids to measure distances

  • Functional correlation:

    • Reconstitute atpE into liposomes of varying lipid composition

    • Measure proton conductance as a function of lipid environment

    • Determine how lipid composition affects c-ring assembly and stability

This research is particularly relevant because Rickettsia africae, as an obligate intracellular pathogen, must adapt to the specific lipid environment of its host cell. Studies have shown that the lipid composition surrounding ATP synthase can significantly impact its rotational dynamics and energy coupling efficiency . Additionally, because atpE is also known as "lipid-binding protein," understanding these interactions may reveal novel aspects of Rickettsia biology .

How can researchers effectively interpret circular dichroism (CD) spectroscopy data for recombinant Rickettsia africae atpE to assess structural integrity?

Effective interpretation of CD spectroscopy data for atpE requires:

  • Experimental design considerations:

    • Collect spectra at multiple protein concentrations (0.1-1 mg/mL)

    • Use quartz cuvettes with 0.1-1 mm path lengths

    • Perform measurements in multiple buffer conditions (varying pH, salt concentrations)

    • Include appropriate blanks and controls

  • Data processing workflow:

    • Subtract buffer baseline

    • Convert raw ellipticity to mean residue ellipticity

    • Apply smoothing algorithms if necessary (Savitzky-Golay)

    • Perform spectrum deconvolution using reference datasets

  • Interpretation framework:

    Secondary StructureSpectral FeaturesExpected % in atpESignificance of Deviation
    α-helixNegative bands at 208 and 222 nm70-80%Potential misfolding or aggregation
    β-sheetNegative band at 218 nm5-10%Possible domain unfolding
    Random coilNegative band below 200 nm10-15%Denaturation or flexibility
  • Thermal stability analysis:

    • Collect CD spectra at 5°C increments from 20-90°C

    • Plot ellipticity at 222 nm versus temperature

    • Calculate melting temperature (Tm) using sigmoid fitting

    • Compare Tm values across different conditions

Properly folded atpE should show predominantly α-helical content (70-80%), consistent with its role as a membrane-spanning subunit that forms the c-ring structure. Significant deviations from this pattern may indicate problems with protein folding or stability that could affect functional studies .

What are the best approaches for analyzing proteomic data to identify post-translational modifications of atpE in Rickettsia africae?

To effectively analyze post-translational modifications (PTMs) of atpE, implement this comprehensive proteomics workflow:

  • Sample preparation protocol:

    • Perform parallel enrichment strategies (immunoprecipitation, phosphopeptide enrichment)

    • Digest using multiple proteases (trypsin, chymotrypsin, AspN) to ensure complete coverage

    • Include both top-down (intact protein) and bottom-up (peptide) approaches

    • Incorporate stable isotope labeling for quantitative analysis

  • Mass spectrometry strategy:

    • Use high-resolution MS/MS (Orbitrap or QTOF)

    • Implement electron transfer dissociation (ETD) for labile modifications

    • Perform parallel reaction monitoring for targeted analysis of suspected modification sites

    • Develop inclusion lists for low-abundance modified peptides

  • Data analysis pipeline:

    • Search against multiple databases using multiple search engines

    • Implement appropriate false discovery rate controls

    • Validate PTM identifications using synthetic peptide standards

    • Develop site localization scoring

  • Biological validation:

    • Generate site-specific antibodies against identified PTMs

    • Create site-directed mutants (modification-mimicking and non-modifiable)

    • Assess functional consequences through activity assays

This research is particularly important because PTMs may regulate ATP synthase activity in response to changing host conditions. In Rickettsia, which has a limited metabolic repertoire, such regulation could be crucial for survival during different phases of infection .

How do the structural and functional properties of Rickettsia africae atpE compare with those of other bacterial pathogens, and what methodologies best highlight these differences?

To systematically compare atpE across bacterial pathogens, implement these comparative approaches:

  • Sequence analysis methodology:

    • Perform multiple sequence alignment of atpE from diverse bacterial species

    • Calculate conservation scores for each position

    • Identify Rickettsia-specific sequence motifs

    • Map conservation onto structural models

  • Structural comparison workflow:

    • Generate homology models based on available structures

    • Perform molecular dynamics simulations to identify flexible regions

    • Calculate electrostatic surface potentials

    • Analyze c-ring stoichiometry differences

  • Functional comparison:

    • Measure proton/ATP ratios across species

    • Compare inhibitor sensitivity profiles

    • Assess pH dependence of activity

    • Evaluate thermal stability differences

  • Evolutionary analysis:

    Bacterial Groupc-ring SizeKey AdaptationsFunctional ImplicationsResearch Applications
    Rickettsia10-12 subunitsAdaptation to host environmentEfficiency at low energyDrug target specificity
    E. coli10 subunitsBroader metabolic capacityVersatilityComparative benchmarking
    Mycobacteria9 subunitsHigher coupling ratioEnergy conservationAlternative drug targets
    Mitochondria8 subunitsHighly regulatedIntegration with hostEvolutionary studies

This comparative approach reveals that Rickettsia africae atpE, as part of an obligate intracellular pathogen, has evolved specific adaptations for functioning within the host cell environment. These include sequence modifications that may affect proton affinity, regulatory sites that respond to host conditions, and structural features that optimize function under the unique metabolic constraints of obligate intracellular life .

What experimental approaches are most effective for studying the evolutionary conservation of atpE across Rickettsia species and its implications for pathogenicity?

To investigate evolutionary conservation of atpE and its relationship to pathogenicity, implement this research strategy:

  • Phylogenetic analysis framework:

    • Collect atpE sequences from all available Rickettsia genomes

    • Construct maximum likelihood phylogenetic trees

    • Calculate selection pressures (dN/dS ratios) across the sequence

    • Identify sites under positive or purifying selection

  • Structure-function correlation:

    • Map variable regions onto structural models

    • Express recombinant atpE from multiple Rickettsia species

    • Compare biochemical properties (stability, activity)

    • Perform domain swapping experiments between species

  • Host adaptation analysis:

    • Compare atpE sequences from Rickettsia with different host ranges

    • Identify correlation between sequence features and host specificity

    • Develop statistical models to predict host adaptation

  • Pathogenicity correlation:

    • Group Rickettsia species by pathogenicity level (high, moderate, low)

    • Identify atpE sequence features that correlate with virulence

    • Validate through site-directed mutagenesis

    • Test hypotheses in cell culture infection models

What emerging technologies hold the most promise for advancing our understanding of atpE function in Rickettsia africae, and what methodological approaches should researchers prioritize?

Emerging technologies with high potential for atpE research include:

  • Cryo-electron tomography:

    • Visualize ATP synthase in situ within Rickettsia cells

    • Determine native arrangement and interactions

    • Develop sample preparation protocols for bacterial cells

    • Implement subtomogram averaging to enhance resolution

  • Single-molecule techniques:

    • Apply magnetic tweezers to measure c-ring rotation

    • Develop fluorescence-based sensors for proton translocation

    • Implement high-speed atomic force microscopy to observe conformational changes

    • Correlate structural dynamics with functional states

  • Gene editing approaches:

    • Apply CRISPR interference for conditional knockdown

    • Develop site-specific integration systems for Rickettsia

    • Create reporter fusions to monitor expression in real-time

    • Engineer conditional expression systems

  • Computational methods:

    • Implement multiscale modeling combining quantum mechanics and molecular dynamics

    • Apply machine learning to predict structure-function relationships

    • Develop systems biology models integrating metabolic networks

    • Simulate proton translocation at atomistic resolution

Researchers should prioritize methodologies that bridge structural and functional information, particularly those that can be applied to living bacteria rather than isolated proteins. The combination of in situ structural biology with functional measurements represents the most promising approach for understanding how atpE contributes to Rickettsia survival and pathogenesis in the complex host environment .

How can researchers effectively design experiments to investigate the potential of atpE as a drug target for treating Rickettsia infections?

To investigate atpE as a drug target, implement this comprehensive drug discovery workflow:

  • Target validation strategy:

    • Demonstrate essentiality through gene silencing approaches

    • Perform competitive inhibition studies with known ATP synthase inhibitors

    • Develop cell-based assays measuring bacterial ATP production

    • Create conditional mutants to define the target validation window

  • Structure-based drug design methodology:

    • Generate high-resolution structures of Rickettsia atpE

    • Identify druggable pockets through computational analysis

    • Perform virtual screening of compound libraries

    • Validate hits through binding and functional assays

  • Fragment-based drug discovery approach:

    • Screen fragment libraries using thermal shift assays

    • Establish structure-activity relationships

    • Implement fragment growing and linking strategies

    • Optimize lead compounds for specificity

  • Lead optimization workflow:

    PropertyAssay MethodologySuccess CriteriaPotential Challenges
    Binding affinityIsothermal titration calorimetryKd < 100 nMMembrane protein stability
    SelectivityCounter-screening against human ATP synthase>100x selectivityConserved active site
    Cell penetrationIntracellular accumulation assays>10x extracellular concentrationPenetrating bacterial membranes
    In vitro efficacyInfected cell modelsEC50 < 1 μMHost cell toxicity

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.