Recombinant Thermosynechococcus elongatus ATP synthase subunit c (atpE)

Shipped with Ice Packs
In Stock

Description

Overview of Recombinant Thermosynechococcus elongatus ATP Synthase Subunit c (atpE)

Recombinant Thermosynechococcus elongatus ATP synthase subunit c (atpE) is a bioengineered protein derived from the cyanobacterium Thermosynechococcus elongatus. It represents the F₀-sector subunit c of the F₀F₁-ATP synthase complex, critical for proton translocation and ATP synthesis in photosynthetic organisms. This recombinant protein is expressed in E. coli with an N-terminal histidine (His) tag for purification and structural characterization .

Production and Purification

The recombinant atpE is produced via heterologous expression in E. coli, leveraging the organism’s robust protein synthesis machinery. The His-tag facilitates affinity chromatography, yielding high-purity protein suitable for structural and functional studies .

Key Steps:

  1. Cloning: The atpE gene (Q8DLP7) is inserted into a plasmid vector.

  2. Expression: Induced in E. coli under optimized conditions.

  3. Purification: Nickel affinity chromatography (via His-tag) followed by gel filtration.

Notes: Avoid repeated freeze-thaw cycles; store at -20°C/-80°C .

Role in ATP Synthase Regulation

Subunit c interacts with regulatory proteins to modulate ATP synthase activity:

  • AtpΘ Interaction: AtpΘ, a cyanobacterial inhibitor, binds to subunit c and other F₀F₁ components to arrest ATP hydrolysis during darkness or low proton gradients .

  • γ Subunit Coordination: The β-hairpin structure in the γ subunit (rotor) influences subunit c’s proton-translocating efficiency, balancing ATP synthesis and hydrolysis .

Experimental Findings:

Study FocusKey ObservationsSource
AtpΘ BindingSubunit c (atpE) co-purifies with AtpΘ in pull-down assays; inhibits ATPase activity
γ Subunit β-Hairpinβ-hairpin deletion reduces ATP synthesis efficiency and increases ATP hydrolysis
ε Subunit SynergyC-terminal α-helix of ε subunit enhances inhibition of ATP hydrolysis via subunit c

Physiological Relevance in Cyanobacteria

In Thermosynechococcus elongatus, subunit c contributes to survival under environmental stress:

  • Proton Gradient Maintenance: Subunit c-driven proton translocation sustains membrane potential during darkness .

  • ATP Homeostasis: Coordination with AtpΘ prevents ATP depletion by blocking reverse ATP hydrolysis .

Mutational Studies:

  • atpE Knockouts: Not feasible in cyanobacteria, indicating essentiality for ATP synthase assembly .

  • γ Subunit Mutants: Loss of β-hairpin disrupts rotational coupling, reducing ATP synthesis .

Challenges and Future Directions

  1. Structural Elucidation: High-resolution crystallography of subunit c in complex with AtpΘ or γ subunit remains pending.

  2. Dynamic Regulation: Mechanisms underlying light-dependent AtpΘ expression and subunit c interaction require further study .

  3. Biotechnological Applications: Recombinant atpE may aid in developing biohybrid systems for renewable energy .

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 contact your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard blue ice packs. Dry ice shipping requires advance notification and incurs additional charges.
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 settle 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%, provided as a guideline for your reference.
Shelf Life
Shelf life depends on several factors including storage conditions, buffer composition, temperature, and the protein's inherent stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms 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 you require a specific tag, please inform us; we will prioritize its development.
Synonyms
atpE; atpH; tlr0431; 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
1-82
Protein Length
full length protein
Species
Thermosynechococcus elongatus (strain BP-1)
Target Names
atpE
Target Protein Sequence
MNPLIASASVLAAALAIGLASLGPGLAQGNASGQALEGIARQPEAEGKIRGTLLLSLAFM ESLTIYGLVIALVLLFANPFAS
Uniprot No.

Target Background

Function

F1F0 ATP synthase synthesizes ATP from ADP using a proton or sodium gradient. This enzyme comprises two domains: the extramembranous catalytic F1 domain and the membrane-bound proton channel F0 domain, linked by a central and peripheral stalk. ATP synthesis in the F1 catalytic domain is coupled to proton translocation via a rotary mechanism involving the central stalk subunits. A key component of the F0 channel, subunit c plays a direct role in transmembrane translocation. A homomeric c-ring, consisting of 10-14 subunits, forms the central stalk rotor element with the F1 delta and epsilon subunits. The enzyme from Thermosynechococcus elongatus exhibits exceptional stability, retaining functionality after 6 hours at 55°C.

Database Links

KEGG: tel:tlr0431

STRING: 197221.tlr0431

Protein Families
ATPase C chain family
Subcellular Location
Cellular thylakoid membrane; Multi-pass membrane protein.

Q&A

What is the structure and function of ATP synthase subunit c (atpE) in Thermosynechococcus elongatus?

ATP synthase subunit c (atpE) is a membrane-embedded component of the Fo portion of the FoF1 ATP synthase complex. This protein forms a ring structure in the membrane that facilitates proton translocation across the thylakoid membrane, which drives the rotary mechanism needed for ATP synthesis. In cyanobacteria like Thermosynechococcus elongatus, this process is particularly important as ATP synthase complexes can utilize proton gradients generated by both photosynthesis and respiration .

The atpE protein typically contains two transmembrane alpha-helices connected by a hydrophilic loop. The c-ring structure in ATP synthase comprises multiple copies of this subunit arranged in a circular formation. This arrangement creates a pathway for protons, contributing to the proton motive force that drives ATP synthesis.

How does atpE function differ between light and dark conditions in cyanobacteria?

Under light conditions, the proton gradient driving ATP synthase activity in T. elongatus is primarily generated through photosynthetic electron transport. In darkness, respiratory complexes become the main contributors to the proton gradient. Unlike plant chloroplasts, cyanobacteria cannot use the same inhibitory mechanisms for ATP synthases during night periods because both respiratory and photosynthetic complexes share the same membrane system .

Regulatory proteins like AtpΘ (encoded by atpT) work alongside atpE to prevent ATP hydrolysis (the reverse reaction) under unfavorable conditions, which would otherwise deplete cellular ATP reserves. This regulation ensures energy conservation when light is unavailable. The expression of these regulatory factors is typically highest in darkness but minimal under optimal phototrophic growth conditions .

What experimental design approach is most appropriate for studying recombinant T. elongatus atpE function?

When designing experiments to study recombinant T. elongatus atpE function, researchers should employ true experimental research designs with proper controls and variable manipulation . A recommended approach includes:

  • Define variables clearly:

    • Independent variables: Expression conditions, mutation sites, or inhibitor concentrations

    • Dependent variables: ATP synthesis/hydrolysis rates, proton translocation efficiency, or binding affinity

    • Controlled variables: Temperature, pH, ion concentrations

  • Implement proper controls:

    • Negative controls: Inactive atpE variants or reactions without substrate

    • Positive controls: Wild-type atpE or well-characterized variants

  • Randomization and replication:

    • Perform experiments in random order to minimize systemic bias

    • Conduct sufficient biological and technical replicates to ensure statistical significance

  • Variable manipulation:

    • Systematically alter expression conditions, substrates, or potential inhibitors

    • Measure corresponding changes in activity or structural properties

How should researchers approach homology modeling of T. elongatus atpE structure?

Developing accurate structural models of T. elongatus atpE requires a systematic homology modeling approach:

  • Template selection:

    • Identify resolved structures of homologous ATP synthase subunit c proteins

    • Prioritize templates with high sequence identity (>30%) and resolution (<2.5Å)

    • Consider templates from related cyanobacterial species when available

  • Sequence alignment and model building:

    • Generate sequence alignments between T. elongatus atpE and template structures

    • Use modeling software like Modeller9.16 to construct the 3D model structure

    • Generate multiple models and select the best based on scoring functions

  • Energy minimization and refinement:

    • Subject the initial model to energy minimization to resolve steric clashes

    • Perform molecular dynamics (MD) simulations to refine the structure

    • Validate transmembrane regions and orientation within the lipid bilayer

  • Model validation:

    • Evaluate stereochemical quality using Ramachandran plots

    • Assess model quality using metrics like QMEAN or ProSA

    • Compare conservation of key functional residues with homologous proteins

This methodological approach provides a reliable structural foundation for further functional and inhibitor studies of T. elongatus atpE.

What methods are most effective for analyzing the proton translocation function of recombinant T. elongatus atpE?

Analyzing proton translocation through recombinant T. elongatus atpE requires specialized techniques:

  • Reconstitution in liposomes:

    • Purify recombinant atpE protein using detergent solubilization

    • Reconstitute into liposomes with defined lipid composition

    • Verify proper orientation using protease protection assays

  • Proton flux measurements:

    • Monitor pH changes using pH-sensitive fluorescent dyes (e.g., ACMA, pyranine)

    • Establish proton gradients using valinomycin/K+ or acid-base transitions

    • Quantify proton flux rates under different conditions

  • Patch-clamp electrophysiology:

    • Form giant liposomes or proteoliposomes

    • Measure ion currents across membranes containing reconstituted atpE

    • Analyze conductance properties under varying voltage and ionic conditions

  • Site-directed mutagenesis:

    • Introduce mutations at key residues predicted to participate in proton translocation

    • Compare proton translocation efficiency between wild-type and mutant proteins

    • Correlate functional changes with structural perturbations

These methodological approaches provide direct functional assessments of atpE's role in proton translocation, the fundamental process driving ATP synthesis.

How can researchers effectively study interactions between recombinant T. elongatus atpE and potential inhibitors?

Studying interactions between T. elongatus atpE and potential inhibitors requires a systematic approach:

  • Virtual screening and docking:

    • Search compound databases (e.g., Zinc, PubChem) for potential atpE binders

    • Perform molecular docking to identify compounds with favorable binding energies

    • Filter compounds based on physicochemical properties (Lipinski's rule of five)

  • Binding assays:

    • Develop fluorescence-based binding assays using labeled inhibitors

    • Perform isothermal titration calorimetry (ITC) to determine binding thermodynamics

    • Use surface plasmon resonance (SPR) to measure binding kinetics

  • Functional inhibition assays:

    • Measure ATP synthesis/hydrolysis rates in the presence of potential inhibitors

    • Determine IC50 values and inhibition constants

    • Characterize inhibition mechanisms (competitive, non-competitive, uncompetitive)

  • Structure-activity relationship (SAR) studies:

    • Test structural analogs of promising inhibitors

    • Correlate structural features with inhibition potency

    • Guide rational design of improved inhibitors

  • Validation in biological systems:

    • Test effects on ATP synthesis in membrane vesicles or whole cells

    • Evaluate specificity by testing effects on other ATP-dependent processes

    • Assess potential off-target effects through proteomic approaches

This comprehensive approach enables identification and validation of specific atpE inhibitors with potential applications in fundamental research and possibly therapeutic development.

What experimental approaches can reveal the transcriptional regulation of T. elongatus atpE?

To study transcriptional regulation of T. elongatus atpE, researchers should consider these methodological approaches:

  • Promoter analysis:

    • Identify promoter regions through bioinformatic analysis

    • Clone promoter regions into reporter gene constructs

    • Measure reporter activity under different conditions (light/dark cycles, nutrient availability)

  • Transcription factor identification:

    • Perform DNA co-immunoprecipitation followed by mass spectrometry

    • Identify binding of transcriptional regulators to the promoter region

    • In cyanobacteria, factors like cyAbrB1 and cyAbrB2 may be involved, as they regulate similar ATP synthase components

  • Chromatin immunoprecipitation (ChIP):

    • Perform ChIP experiments with antibodies against suspected transcription factors

    • Quantify enrichment of atpE promoter regions

    • Map binding sites through ChIP-seq analysis

  • Transcriptional response analysis:

    • Monitor atpE mRNA levels under different conditions using RT-qPCR

    • Compare with other ATP synthase components to identify coordinated regulation

    • Create transcriptional profiles across different growth phases and environmental conditions

These approaches can reveal the complex regulatory networks controlling atpE expression in response to changing environmental conditions.

How can researchers investigate post-transcriptional regulation of T. elongatus atpE?

Post-transcriptional regulation of atpE can be investigated through these methodological approaches:

  • mRNA stability analysis:

    • Measure atpE mRNA half-life using transcription inhibitors

    • Compare stability under different conditions (light/dark, nutrient availability)

    • Identify sequence elements affecting stability through mutation analysis

    • For cyanobacteria, mRNA stability has been identified as a major regulatory process governing expression of ATP synthase components

  • RNA-binding protein identification:

    • Perform RNA immunoprecipitation followed by mass spectrometry

    • Identify proteins that bind to atpE mRNA

    • Map binding sites through techniques like CLIP-seq

  • Translational efficiency measurement:

    • Construct reporter genes fused with atpE 5'UTR

    • Measure translation rates under different conditions

    • Identify regulatory elements within the 5'UTR region

    • For ATP synthase components in cyanobacteria, the histone-like protein HU can bind to the 5'UTR, potentially affecting translation

  • Ribosome profiling:

    • Perform ribosome profiling to measure ribosome occupancy on atpE mRNA

    • Compare translational efficiency under different conditions

    • Identify potential translational pausing sites

These methodological approaches provide a comprehensive understanding of the post-transcriptional mechanisms that fine-tune atpE expression in response to cellular energy demands.

What strategies can overcome challenges in expressing and purifying functional recombinant T. elongatus atpE?

Membrane proteins like atpE present unique challenges in recombinant expression and purification. Researchers can overcome these through:

  • Expression system optimization:

    • Test multiple expression systems (E. coli, yeast, insect cells)

    • Evaluate different promoters, fusion tags, and signal sequences

    • Consider specialized E. coli strains designed for membrane protein expression

    • Optimize growth temperature, induction conditions, and media composition

  • Solubilization strategies:

    • Screen multiple detergents (DDM, LMNG, digitonin) for optimal extraction

    • Test detergent:protein ratios and solubilization times

    • Consider native nanodiscs or styrene-maleic acid copolymer lipid particles (SMALPs)

    • Maintain proper lipid environment throughout purification

  • Purification optimization:

    • Implement two-step affinity purification using tags on both N- and C-termini

    • Add stabilizing agents (glycerol, specific lipids) to all buffers

    • Minimize exposure to detergents through rapid purification protocols

    • Consider on-column detergent exchange or reconstitution

  • Functional verification:

    • Develop activity assays compatible with detergent-solubilized protein

    • Verify proper folding through circular dichroism or limited proteolysis

    • Confirm oligomeric state through size-exclusion chromatography

    • Validate proton translocation function in reconstituted systems

These methodological strategies address the specific challenges associated with membrane protein biochemistry, improving the likelihood of obtaining functional recombinant T. elongatus atpE.

How can researchers resolve inconsistent results in ATP synthase activity assays?

When facing inconsistent results in ATP synthase activity assays involving recombinant T. elongatus atpE, consider these troubleshooting approaches:

  • Sample quality assessment:

    • Verify protein purity through SDS-PAGE and mass spectrometry

    • Confirm proper folding and oligomerization state

    • Check protein stability under assay conditions using thermal shift assays

    • Ensure consistent protein:lipid ratios in reconstituted systems

  • Assay condition optimization:

    • Systematically vary pH, temperature, and ionic strength

    • Test different lipid compositions for reconstitution

    • Optimize ATP, ADP, and Pi concentrations

    • Control for pre-existing ion gradients that may affect results

  • Technical considerations:

    • Implement rigorous controls for each experiment

    • Standardize reagent preparation and storage

    • Use multiple detection methods to cross-validate results

    • Ensure instruments are properly calibrated

  • Experimental design improvements:

    • Apply systematic experimental design principles

    • Randomize experimental order to minimize systematic bias

    • Increase technical and biological replicates

    • Use statistical methods appropriate for the data type

  • Environmental variables:

    • Control light exposure during preparation and assays

    • Monitor and eliminate oxidative damage to the protein

    • Standardize all buffer components and their quality

    • Consider effects of mechanical stress during preparation

By systematically addressing these factors, researchers can identify sources of variability and establish reliable, reproducible assay conditions for studying T. elongatus atpE function.

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.