ATP5F1 forms part of the Fo complex's peripheral stalk in mitochondrial ATP synthase, which contains nine subunits (a, b, c, d, e, f, g, F6, and 8) . The protein ensures structural stability by linking the catalytic F1 complex (α₃β₃γδε) to the membrane-embedded Fo proton channel .
ATP5F1 contributes to three core activities:
Anchors the F1-Fo stator, preventing rotation during ATP synthesis .
Interacts with ATP5D (δ subunit) to maintain Fo complex integrity .
Facilitates proton transport via Fo, enabling γ-subunit rotation in F1 for ATP synthesis .
Associates with ATP5J and ATP5H to modulate transmembrane proton conductance .
Mutations in ATP5F1 or its assembly factors correlate with severe pathologies:
Leigh Syndrome: Homozygous ATP5F1D variants disrupt Fo assembly, reducing cristae density and ATP output .
Encephalopathy: m.8993T>G mutations in mitochondrial DNA impair proton flow, causing >90% ATP deficit in neurons .
Alzheimer’s Disease: ATP5F1 dysregulation exacerbates amyloid-β toxicity by depleting synaptic ATP .
Batten Disease: Lysosomal accumulation of ATP5F1’s c-subunit aggregates correlates with neuronal ceroid lipofuscinosis .
Achilles Cell Line Data: ATP5F1 knockdown reduces fitness in 23% of cancer cell lines, highlighting metabolic dependencies .
ProSpec’s ATP5F1 Human Recombinant (22.6 kDa) serves as a tool for mechanistic studies:
Property | Details |
---|---|
Expression System | E. coli |
Tag | N-terminal His-tag |
Purity | >95% via proprietary chromatography |
Storage | -20°C in 20 mM Tris-HCl (pH 8.0), 0.4M urea, 10% glycerol |
This reagent has been utilized to study Fo complex assembly and screen ATP synthase inhibitors .
Cristae Morphology: ATP5F1 dimerization is critical for cristae formation; its loss triggers mitochondrial fragmentation .
Ischemic Injury: During hypoxia, ATP5F1 reverses to hydrolyze ATP, accelerating cellular energy collapse .
Gene Therapy: Drosophila models expressing human ATP5F1 variants reveal rescue potentials for δ-subunit deficiencies .
ATP5F1A (also known as ATP Synthase F1 Subunit Alpha) is a critical component of mitochondrial ATP synthase (Complex V), which catalyzes ATP synthesis using an electrochemical gradient of protons during oxidative phosphorylation. The ATP synthase complex consists of two linked multi-subunit complexes: the soluble catalytic core (F1) and the membrane-spanning component (Fo) comprising the proton channel .
The F1 catalytic portion contains 5 different subunits (alpha, beta, gamma, delta, and epsilon) assembled with a stoichiometry of 3 alpha, 3 beta, and a single representative of the other three subunits . ATP5F1A forms part of the catalytic core along with beta subunits, though interestingly, the alpha subunit itself does not bear the catalytic high-affinity ATP-binding sites .
During catalysis, ATP synthesis in F1 is coupled via a rotary mechanism of the central stalk subunits to proton translocation. The rotation of the central stalk against the surrounding alpha(3)beta(3) subunits leads to ATP synthesis in three separate catalytic sites on the beta subunits .
ATP5F1A has multiple isoforms and aliases that researchers need to be aware of when designing experiments. The major isoforms include:
Cardiac muscle isoform (previously known as ATP5A1) - predominantly expressed in cardiac tissue
Non-cardiac muscle isoform (previously known as ATP5AL2) - expressed in other tissues
To distinguish between these isoforms, researchers should employ the following methodological approaches:
RT-PCR with isoform-specific primers: Design primers that span unique exon junctions or sequence regions specific to each isoform
Western blotting with isoform-specific antibodies: Use antibodies raised against unique epitopes in each isoform
Mass spectrometry: Analyze tryptic peptides unique to each isoform
RNA-seq analysis: Look for differential expression patterns across tissues
When reporting results, researchers should clearly specify which isoform nomenclature they are using, as the field has undergone several naming conventions (ATP5A, ATP5F1A, ATP5A1, etc.) . Additionally, researchers should validate antibody specificity using appropriate positive and negative controls to ensure accurate isoform identification.
Assessing the proper assembly of ATP5F1 subunits into functional Complex V requires multiple complementary approaches:
Protein Complex Analysis:
Blue Native PAGE (BN-PAGE): This technique separates intact protein complexes according to their molecular weight while preserving native protein-protein interactions .
2D BN/SDS-PAGE: Combines BN-PAGE with subsequent SDS-PAGE to identify individual subunits within complexes .
Immunoprecipitation with subunit-specific antibodies: Allows for the isolation of intact complexes and identification of interacting partners.
Functional Assessments:
In-gel activity assays: After BN-PAGE, gels can be incubated with ATP and lead nitrate to visualize ATP hydrolysis activity as lead phosphate precipitates.
Spectrophotometric enzyme activity assays: Measures ATP synthesis or hydrolysis rates in isolated mitochondria.
Oxygen consumption rate (OCR): Using platforms like Seahorse XF Analyzer to measure oligomycin-sensitive respiration, which correlates with ATP synthase function.
Structural Analysis:
Transmission electron microscopy (TEM): Can reveal abnormalities in mitochondrial cristae structure, which often correlate with ATP synthase assembly defects .
Immunofluorescence microscopy: Visualizes co-localization of ATP5F1 subunits within mitochondria.
In research studies, abnormal ATP synthase assembly has been observed in ATP5F1D mutations, where despite normal levels of the mutated subunit, other Complex V subunits (ATP5F1A, ATP5F1B, and ATP5PO) showed decreased abundance, indicating impaired complex assembly .
Measuring ATP synthase activity requires specialized techniques that assess both ATP production and the proton pumping function of the complex:
1. Spectrophotometric Coupled Enzyme Assays:
ATP hydrolysis can be measured by coupling ADP production to NADH oxidation through pyruvate kinase and lactate dehydrogenase
The decrease in NADH absorbance at 340 nm correlates with ATP hydrolysis rate
Oligomycin sensitivity distinguishes ATP synthase activity from other ATPases
2. Luminescence-Based ATP Measurement:
Luciferase-luciferin reactions quantify ATP production
Can be applied to isolated mitochondria, permeabilized cells, or tissue homogenates
Allows real-time ATP production monitoring
3. Oxygen Consumption Rate (OCR) Analysis:
Using Seahorse XF Analyzer to measure OCR in intact cells or isolated mitochondria
Sequential addition of oligomycin, FCCP, and rotenone/antimycin A allows calculation of:
ATP synthesis-linked respiration
Proton leak
Maximum respiratory capacity
Non-mitochondrial respiration
4. Membrane Potential Measurements:
Fluorescent probes (TMRM, JC-1) assess the proton gradient driving ATP synthesis
Flow cytometry or fluorescence microscopy quantifies the signal
Provides important context when assessing ATP synthase function
When designing experiments, researchers should include appropriate controls (oligomycin inhibition), normalize to mitochondrial content (using citrate synthase activity), and account for tissue/cell-specific differences in mitochondrial content.
Detecting protein-protein interactions within the ATP synthase complex requires techniques that preserve native interactions:
1. Co-immunoprecipitation (Co-IP):
Use antibodies against specific ATP5F1 subunits to precipitate the entire complex
Western blot analysis of precipitated samples reveals interacting partners
Requires careful buffer optimization to maintain interactions
Works well for stable interactions but may miss transient ones
2. Proximity Labeling Approaches:
BioID: Fusion of a promiscuous biotin ligase to an ATP5F1 subunit labels proximal proteins
APEX2: Peroxidase-based proximity labeling with higher temporal resolution
These methods capture both stable and transient interactions in living cells
Mass spectrometry identifies labeled proteins
3. Förster Resonance Energy Transfer (FRET):
Fluorescently tag pairs of ATP5F1 subunits (donor and acceptor)
Energy transfer occurs only when proteins are in close proximity (1-10 nm)
Can be measured by acceptor photobleaching or fluorescence lifetime imaging
Allows for real-time monitoring of interactions in living cells
4. Crosslinking Mass Spectrometry (XL-MS):
Chemical crosslinkers create covalent bonds between interacting proteins
MS/MS analysis identifies crosslinked peptides
Provides detailed spatial information about interaction interfaces
Particularly valuable for mapping the rotary mechanism of ATP synthase
5. Cryo-Electron Microscopy:
Visualizes the entire ATP synthase complex at near-atomic resolution
Can reveal structural details of subunit interactions
When combined with site-directed mutagenesis, can validate interaction models
Research has shown that pathogenic variants in ATP5F1D (p.Pro82Leu and p.Val106Gly) do not affect protein levels but disrupt binding to other F1 subunits, demonstrating how these techniques can reveal mechanistic insights into disease-causing mutations .
Isolating functional ATP5F1 subunits presents several technical challenges that researchers must address:
1. Optimized Solubilization Strategies:
Gentle detergents: Digitonin (0.5-1%) or n-dodecyl-β-D-maltoside (0.5-1%) preserve native structures
Gradual solubilization at low temperatures (4°C) minimizes denaturation
Buffer optimization (pH 7.2-7.8, 150-250 mM NaCl) maintains stability
Addition of ATP or ADP (1-2 mM) can stabilize conformations
2. Advanced Purification Approaches:
Affinity chromatography using:
Histidine-tagged recombinant subunits
ATP-agarose for nucleotide-binding subunits
Specific antibody columns
Blue Native electrophoresis followed by electroelution for intact complexes
Size exclusion chromatography to separate fully assembled complexes from subcomplexes
3. Expression Systems Considerations:
Mammalian expression systems (HEK293T, CHO) maintain proper folding and modifications
Co-expression of multiple subunits improves stability and assembly
Inducible expression systems with temperature control prevent toxicity
Addition of specific chaperones can improve yield of properly folded proteins
4. Stability Enhancement Approaches:
Addition of stabilizing agents:
10-15% glycerol
1-5 mM DTT or TCEP to maintain reduced cysteines
Protease inhibitor cocktails
Chemical crosslinking for structural studies
Nanodiscs or amphipols to maintain membrane protein stability
5. Functional Validation Methods:
ATPase activity assays to confirm enzymatic function
Reconstitution into liposomes to assess proton pumping
Structural analysis by circular dichroism and thermal shift assays
When purifying ATP5F1D specifically, researchers have observed that disease-associated variants (p.Pro82Leu and p.Val106Gly) affect interaction with other F1 subunits but not protein stability, highlighting the importance of assessing both protein levels and functional interactions .
Mutations in ATP5F1 genes can lead to severe mitochondrial diseases through several pathogenic mechanisms:
1. Impaired ATP Synthase Assembly:
Biallelic missense variants in ATP5F1D (p.Pro82Leu and p.Val106Gly) have been shown to cause a Mendelian mitochondrial disease characterized by episodic metabolic decompensation, lethargy, metabolic acidosis, 3-methylglutaconic aciduria, and hyperammonemia . Functional studies demonstrated that these mutations do not affect ATP5F1D protein levels but disrupt its ability to interact with other F1 subunits, leading to reduced complex V assembly .
2. Structural Mitochondrial Abnormalities:
Transmission electron microscopy of fibroblasts from patients with ATP5F1D mutations revealed dramatically decreased cristae number . In induced pluripotent stem cell (iPSC)-derived cardiomyocytes, both mitochondrial size and cristae number were reduced . These structural abnormalities directly impact the capacity for oxidative phosphorylation.
3. Bioenergetic Dysfunction:
Patient-derived cells carrying ATP5F1D mutations exhibited impaired maximal respiration, particularly in response to metabolic substrates like palmitate . This functional respiratory deficit translates to insufficient ATP production, especially during periods of increased energy demand.
4. Tissue-Specific Manifestations:
The clinical presentation of ATP5F1 mutations often shows tissue specificity, with energy-demanding tissues like the brain, heart, and skeletal muscle being most affected. This reflects the different energy requirements and mitochondrial content across tissues.
A particularly effective experimental approach for validating pathogenicity is the use of model organisms. Knockdown of the ATP5F1D homolog (ATPsynδ) in Drosophila caused a near-complete loss of the fly head, and this severe phenotype could be rescued by wild-type human ATP5F1D but not by the mutant versions, confirming their pathogenicity .
The relationship between ATP5F1 expression and cancer involves complex metabolic reprogramming:
1. Altered Expression Patterns:
Studies have systematically analyzed the expression of mitochondrial ATP synthase (Complex V) subunits in clear cell renal cell carcinoma (ccRCC) . ATP5F1A and other complex V subunits show altered expression in various cancers, potentially serving as biomarkers for diagnosis or prognosis .
2. Metabolic Reprogramming Mechanisms:
Cancer cells often exhibit the Warburg effect - a shift from oxidative phosphorylation to aerobic glycolysis despite oxygen availability. Changes in ATP synthase subunit expression may either:
Drive this metabolic shift by limiting oxidative phosphorylation
Occur as a consequence of metabolic adaptation to the tumor microenvironment
Represent a compensatory response to altered energy demands
3. Beyond Energy Production:
ATP synthase components have roles beyond ATP production that may influence cancer biology:
Involvement in apoptosis regulation (particularly through interactions with mitochondrial permeability transition pore)
Potential roles in cell signaling pathways
Contributions to mitochondrial morphology and dynamics
4. Research Approaches:
To study ATP5F1 in cancer contexts, researchers employ:
Expression analysis comparing tumor vs. adjacent normal tissue
Correlation of expression levels with clinical parameters and outcomes
Knockdown/overexpression studies in cancer cell lines
Metabolic flux analysis to assess bioenergetic consequences
5. Therapeutic Implications:
Understanding ATP5F1 dysregulation in cancer could lead to:
Novel diagnostic or prognostic biomarkers
Potential therapeutic targets
Strategies to target cancer-specific metabolic vulnerabilities
The systematic analysis of ATP synthase subunit expression in ccRCC represents an important step toward developing better understanding of this complex's role in cancer biology .
Post-translational modifications (PTMs) of ATP5F1 subunits represent a critical regulatory mechanism:
1. Types of PTMs Affecting ATP5F1 Subunits:
Modification | Sites | Regulatory Enzymes | Function |
---|---|---|---|
Phosphorylation | Ser/Thr/Tyr residues | PKA, AMPK, Src-family kinases | Activity regulation, assembly control |
Acetylation | Lysine residues | Sirtuins (especially SIRT3) | Metabolic sensing, activity modulation |
Oxidative modifications | Cysteine, tyrosine residues | Non-enzymatic (ROS-mediated) | Stress response, often detrimental |
SUMOylation | Lysine residues | SUMO E1/E2/E3 enzymes | Stability, quality control |
2. Functional Consequences of PTMs:
Altered catalytic efficiency of ATP synthesis
Modified assembly/disassembly kinetics of the complex
Changed stability of individual subunits
Altered interactions between subunits
Modified responses to regulatory signals
3. Methods for Studying ATP5F1 PTMs:
Mass Spectrometry Approaches:
Enrichment strategies for specific PTMs (phosphopeptides, acetylated peptides)
Parallel reaction monitoring for targeted analysis
SILAC or TMT labeling for quantitative comparisons
PTM crosstalk analysis using multi-protease digestion
Functional Validation:
Site-directed mutagenesis of modified residues
Expression of phosphomimetic/phospho-dead mutants
In vitro treatment with modifying/demodifying enzymes
4. Disease Implications:
PTM dysregulation can contribute to pathology through:
Hyperphosphorylation or hyperacetylation altering ATP synthase function
Oxidative stress leading to irreversible modifications
Disrupted PTM regulatory networks affecting mitochondrial adaptation
Studying these modifications provides insights into how ATP synthase function is fine-tuned in health and dysregulated in disease, potentially revealing novel therapeutic targets for mitochondrial dysfunction.
CRISPR-Cas9 technology offers powerful approaches for studying ATP5F1 subunits, but requires careful optimization:
1. Strategic Design Considerations:
Guide RNA Design:
Target specificity is critical due to homology between ATP5F1 subunits
Off-target analysis using tools like CRISPOR or Cas-OFFinder
Design multiple gRNAs per target and validate efficiency
Consider chromatin accessibility at target sites
Editing Strategies:
Complete knockout may be lethal - consider conditional approaches
Knockin of specific mutations (e.g., ATP5F1D p.Pro82Leu) to model patient variants
Epitope tagging for visualization and purification
Fluorescent protein fusions for live imaging
2. Delivery Optimization:
Delivery Method | Advantages | Limitations | Best Applications |
---|---|---|---|
Plasmid transfection | Simple, inexpensive | Lower efficiency, longer Cas9 expression | Stable cell line generation |
RNP complexes | Higher efficiency, reduced off-targets | Transient editing | Primary cells, reduced off-target concerns |
Lentiviral delivery | Works in difficult-to-transfect cells | Packaging size limitations | Neurons, iPSCs |
AAV delivery | In vivo applications | Limited packaging capacity | Animal models |
3. Advanced CRISPR Applications:
Base editing: For precise nucleotide changes without double-strand breaks
Prime editing: For precise insertions, deletions, and all base-to-base conversions
CRISPRi/CRISPRa: For modulation of gene expression without DNA modification
CRISPR screening: For identifying genetic interactions with ATP5F1 subunits
4. Validation Approaches:
Western blotting to confirm protein knockout/modification
Functional assays (OCR, ATP production)
Rescue experiments with wild-type cDNA to confirm specificity
5. Cell Model Selection:
Patient-derived fibroblasts for studying natural mutations
iPSCs for differentiation into affected cell types (e.g., cardiomyocytes, neurons)
Cell types with high mitochondrial content show more pronounced phenotypes
CRISPR-based studies have been instrumental in confirming pathogenicity of ATP5F1D variants, as demonstrated by rescue experiments in model organisms .
Computational prediction of ATP5F1 variant effects requires multiple complementary approaches:
1. Sequence-Based Methods:
Conservation Analysis: Highly conserved residues are more likely functionally important
Variant Effect Predictors:
SIFT: Predicts effects based on sequence homology
PolyPhen-2: Combines sequence conservation with structural features
CADD: Integrates multiple annotations into a single pathogenicity score
Ensemble methods (REVEL, VEST) often outperform individual predictors
2. Structure-Based Methods:
Protein Stability Analysis:
FoldX: Calculates changes in folding free energy (ΔΔG)
Rosetta: Models structural perturbations
DUET: Integrates multiple stability predictors
Molecular Dynamics Simulations:
Simulates atomic movements over time
Reveals potential disruptions to subunit interactions and conformational changes
Particularly valuable for the rotary mechanism of ATP synthase
3. ATP Synthase-Specific Approaches:
Interface Analysis: Examining subunit interaction interfaces is critical, as demonstrated by ATP5F1D mutations that disrupt binding to other F1 subunits
Functional Domain Mapping: Identifying variants in regions critical for:
Nucleotide binding
Catalytic activity
Rotary mechanism
Subunit assembly
4. Validation Against Known Disease Mutations:
The p.Pro82Leu and p.Val106Gly variants in ATP5F1D provide valuable benchmarks for computational prediction methods . These variants don't affect protein stability but disrupt complex assembly, highlighting the importance of considering protein-protein interactions in predictions.
When applying computational approaches to novel ATP5F1 variants, researchers should:
Use multiple complementary methods rather than relying on a single predictor
Consider the specific structural and functional context of the variant
Validate predictions with experimental approaches when possible
Be cautious about variants in regions with limited structural information
Multi-omics integration provides comprehensive insights into ATP5F1 dysfunction:
1. Multi-Omics Data Collection:
Omics Layer | Techniques | Insights for ATP5F1 Research |
---|---|---|
Genomics | WGS/WES, gene panels | Identification of pathogenic variants |
Transcriptomics | RNA-Seq, scRNA-Seq | Expression patterns, alternative splicing |
Proteomics | BN-PAGE-MS, PTM analysis | Complex assembly, post-translational modifications |
Metabolomics | LC-MS/MS, NMR | ATP/ADP ratios, TCA cycle intermediates |
Functional omics | OCR, membrane potential | Real-time functional assessment |
2. Integration Strategies:
Network-Based Approaches:
Construct multi-layered networks connecting genetic variants to functional outcomes
Identify key nodes that connect different data types
Tools: Cytoscape with OmicsIntegrator, NetworkAnalyst
Machine Learning Integration:
Dimensionality reduction using MOFA (Multi-Omics Factor Analysis)
Patient stratification based on integrated profiles
Predictive modeling of disease progression
Pathway-Centered Analysis:
Map all omics data onto oxidative phosphorylation pathways
Identify regulatory relationships between omics layers
Use ATP5F1 as focal point for integrative analysis
In the case of ATP5F1D mutations causing mitochondrial disease , a multi-omics approach revealed:
Genomics: Identified causal variants (p.Pro82Leu and p.Val106Gly)
Proteomics: Showed normal ATP5F1D levels but decreased other complex V subunits
Structural biology: Revealed decreased cristae
Functional assays: Demonstrated impaired respiration
Model organisms: Confirmed pathogenicity through rescue experiments
This comprehensive approach provided mechanistic understanding from gene to phenotype, demonstrating how multiple data types collectively explain the disease process.
4. Implementation Considerations:
Begin with hypothesis-driven core questions about ATP5F1 function
Carefully plan experimental design to enable integration
Consider time-course data to capture dynamic processes
Include appropriate controls at each omics layer
Validate key findings using orthogonal methods
Multi-omics approaches are particularly valuable for understanding complex mitochondrial diseases where ATP5F1 dysfunction may be just one component of broader bioenergetic dysregulation.
The mitochondrial ATP synthase complex is composed of two main components: the F1 catalytic core and the Fo proton channel. The Fo complex is embedded in the inner mitochondrial membrane and is responsible for proton translocation. The F1 complex protrudes into the mitochondrial matrix and is responsible for ATP synthesis. The ATP5F1 subunit is part of the Fo complex and plays a critical role in the transport of protons across the membrane, which drives the rotation of the F1 complex and the subsequent synthesis of ATP .
The human recombinant version of Synthase Transporting Mitochondrial Fo Complex B1 is produced using recombinant DNA technology. This involves inserting the gene encoding the ATP5F1 protein into a suitable expression system, such as bacteria or yeast, to produce the protein in large quantities. The recombinant protein is then purified for use in research and therapeutic applications .
The recombinant ATP5F1 protein is widely used in biochemical and biophysical studies to understand the mechanisms of ATP synthesis and mitochondrial function. It is also used in drug discovery and development, particularly in the search for compounds that can modulate mitochondrial function and treat mitochondrial diseases. Additionally, the protein is used in the study of various metabolic disorders and neurodegenerative diseases, where mitochondrial dysfunction is a key factor .