Recombinant Human V-type proton ATPase subunit e 1 (ATP6V0E1) is a synthetic protein engineered to study the functional and structural roles of the e1 subunit in vacuolar-type ATPase (V-ATPase) complexes. This subunit belongs to the V₀ domain of V-ATPase, a multisubunit proton pump critical for acidifying intracellular organelles like lysosomes, synaptic vesicles, and the Golgi apparatus . ATP6V0E1 is encoded by the ATP6V0E1 gene (chromosome 5q35.3) and is ubiquitously expressed in human tissues .
Production Process:
Recombinant ATP6V0E1 is synthesized via in vitro wheat germ expression systems, followed by affinity purification using GST tags . This method ensures high yield and stability for downstream applications like antibody development and biochemical assays .
ATP6V0E1 is part of the V₀ domain, which couples ATP hydrolysis (by the V₁ domain) to proton translocation across membranes. Key roles include:
Proton Pump Assembly: Stabilizes the V₀ complex and facilitates interaction with accessory proteins (e.g., ATP6AP1) .
pH-Dependent Regulation: Modulates V₁ domain dissociation during organelle acidification .
Tissue-Specific Functions:
Cancer Metabolism: ATP6V0E1 knockdown reduces pancreatic cancer cell viability, highlighting its role in lactate transport and survival .
Neurological Functions: Structural studies reveal ATP6V0E1’s interaction with synaptophysin, suggesting a role in synaptic vesicle acidification .
Diabetes Pathophysiology: ATP6V0E1 regulates lysosomal acidification in β-cells, impacting insulin secretion under lipotoxic stress .
ATP6V0E1 is a component of the vacuolar ATPase (V-ATPase), a multisubunit enzyme that mediates acidification of eukaryotic intracellular organelles. V-ATPase is composed of two domains:
V1 domain (cytosolic): Contains eight subunits (A-H) that catalyze ATP hydrolysis
V0 domain (transmembrane): Contains five different subunits (a, c, c', c", and d) involved in proton translocation
ATP6V0E1 encodes the e1 subunit of the V0 domain. The protein is relatively small at approximately 9.4 kDa with 81 amino acid residues in the canonical form in humans .
V-ATPase-dependent organelle acidification is necessary for crucial cellular processes including protein sorting, zymogen activation, receptor-mediated endocytosis, and synaptic vesicle proton gradient generation . The e subunit has been shown to be essential for normal V-ATPase function, with studies demonstrating that knockdown of the e subunit reduces V-ATPase efficiency .
ATP6V0E1 differs from other V-ATPase subunits in several key ways:
Size and location: It is one of the smaller subunits within the V0 domain (9.4 kDa)
Isoform variation: ATP6V0E1 has a paralog called ATP6V0E2, with distinct expression patterns
Functional specificity: While many V-ATPase subunits have tissue-specific expression, ATP6V0E1 is more ubiquitously expressed across many tissue types
For studying ATP6V0E1 specifically:
Recommended techniques:
RNA interference (siRNA): Effective for knockdown studies to assess functional impact, as demonstrated in neuroblastoma research
Immunofluorescence microscopy: For subcellular localization studies
Co-immunoprecipitation: To identify interaction partners
CRISPR-Cas9 gene editing: For generating knockout cell lines
Methodological considerations:
When performing knockdown experiments, include ATP6V0E2 controls to account for potential compensatory mechanisms
Use multiple antibodies targeting different epitopes for validation due to the small size of the protein
Consider V-ATPase activity assays (measuring proton transport or ATP hydrolysis) to assess functional impact of ATP6V0E1 manipulation
To accurately determine ATP6V0E1 expression patterns:
Transcriptional analysis approaches:
RT-qPCR: Design primers specific to ATP6V0E1 that do not amplify ATP6V0E2 or pseudogenes
RNA-Seq: For comprehensive transcriptome analysis and splice variant identification
In situ hybridization: For spatial expression analysis in tissue sections
Protein-level approaches:
Western blotting: Using validated ATP6V0E1-specific antibodies
Mass spectrometry: For unbiased proteomic profiling
Immunohistochemistry: For spatial localization in tissues
Important methodological considerations:
Validate antibody specificity against recombinant ATP6V0E1 protein
Include ATP6V0E2 controls to ensure isoform specificity
Consider post-translational modifications like glycosylation that may affect detection
Use subcellular fractionation to confirm organelle-specific localization
Studies have shown that ATP6V0E1 is ubiquitously expressed across many tissue types but may show variable expression levels. Proper controls and validation steps are essential for accurate expression profiling.
Recommended approaches for subcellular localization studies:
Fluorescently-tagged fusion proteins:
GFP-ATP6V0E1 fusion constructs for live-cell imaging
Consider both N-terminal and C-terminal tags to determine which preserves function
Validate with co-localization studies using organelle markers
Immunofluorescence microscopy:
Use validated ATP6V0E1 antibodies
Co-stain with organelle markers (LAMP1 for lysosomes, EEA1 for early endosomes)
Super-resolution microscopy for precise localization
Biochemical fractionation:
Differential centrifugation to isolate organelle fractions
Density gradient separation of organelles
Western blotting of fractions to detect ATP6V0E1
Proximity labeling methods:
BioID or TurboID fusion proteins to identify proximal proteins
APEX2 for electron microscopy visualization
Important considerations:
The small size of ATP6V0E1 (9.4 kDa) may make certain fusion proteins challenging
Trafficking studies should include inhibitors of specific trafficking pathways
Consider temporal dynamics using live-cell imaging
Verify functional incorporation into the V-ATPase complex
Research on other V-ATPase subunits has shown remarkable organelle specificity. For example, studies in Paramecium identified 17 genes encoding a-subunit isoforms, with representatives showing highly specific targeting to at least seven different compartments . This suggests ATP6V0E1 may also have specific targeting mechanisms worth investigating.
ATP6V0E1, as part of the V0 domain, is critical for V-ATPase function in lysosomal acidification, which directly impacts autophagy:
Role in lysosomal acidification:
ATP6V0E1 is essential for normal V-ATPase function and proton translocation
Knockdown studies suggest reduced ATP6V0E1 decreases V-ATPase efficiency and impacts lysosomal pH
Impact on autophagy:
V-ATPase activity is required for autophagosome-lysosome fusion and degradation of autophagic cargo
Research has shown that targeting V-ATPase subunits can activate autophagy through mTORC1 inhibition
Methods to quantify lysosomal acidification:
pH-sensitive fluorescent probes:
LysoSensor probes that exhibit pH-dependent changes in fluorescence intensity
LysoTracker dyes that accumulate in acidic compartments
Ratiometric measurements using pH-sensitive fluorophores like pHrodo
Direct measurement of lysosomal pH:
FITC-dextran pulse-chase followed by ratio imaging
Organelle-targeted pH-sensitive GFP variants
Methods to assess autophagy:
LC3 processing and localization:
Western blotting for LC3-I to LC3-II conversion
GFP-LC3 puncta formation by microscopy
Tandem mRFP-GFP-LC3 to distinguish autophagosomes from autolysosomes
Autophagy flux assays:
Treatment with bafilomycin A1 (V-ATPase inhibitor) to block autophagy completion
Monitoring degradation of autophagy substrates (p62/SQSTM1)
Transmission electron microscopy:
Visualization of autophagic structures
Quantification of autophagic vacuoles
Research has shown that covalent targeting of the ATP6V1A subunit of V-ATPase activates autophagy via mTORC1 inhibition and increases lysosomal acidification . Similar approaches could be applied to study ATP6V0E1's specific role.
ATP6V0E1, as part of the V-ATPase complex, plays important roles in various membrane trafficking processes:
Role in endocytosis:
V-ATPase acidifies early endosomes, which is crucial for receptor-ligand dissociation
Proper sorting in the endocytic pathway depends on pH gradients established by V-ATPase
ATP6V0E1 knockdown may impair endosomal trafficking due to altered acidification
Role in exocytosis:
V-ATPase acidifies secretory vesicles, necessary for proper protein sorting and processing
In neurons, V-ATPase generates the proton gradient that drives neurotransmitter loading into synaptic vesicles
Experimental approaches to study these relationships:
Receptor-mediated endocytosis assays:
Fluorescently-labeled transferrin uptake and recycling
EGFR internalization and degradation kinetics
Quantitative analysis of endocytic rate using flow cytometry
Exocytosis measurement techniques:
Total internal reflection fluorescence (TIRF) microscopy of labeled secretory vesicles
Capacitance measurements (for electrophysiological detection of exocytosis)
Secreted protein quantification (ELISA or Western blotting)
Live cell imaging approaches:
pH-sensitive cargo to track acidification during trafficking
Dual-color imaging of ATP6V0E1 and endocytic/exocytic markers
Super-resolution microscopy to visualize V-ATPase distribution on vesicles
Research has shown that ATP6V0E1 knockdown in neuroblastoma cells reduced cell viability but was not sufficient to induce neural cell differentiation, suggesting complex roles beyond simple pH regulation . Additionally, some V-ATPase subunits have been implicated in membrane fusion events independent of their role in acidification, which may be worth investigating for ATP6V0E1.
While ATP6V0E1-specific mutations have not been extensively characterized, research on V-ATPase subunits has revealed important disease associations:
Disease associations of V-ATPase subunits:
ATP6V0E1-specific research findings:
In neuroblastoma, ATP6V0E1 is directly targeted by microRNA-506-3p, which functions as a tumor suppressor
Knockdown of ATP6V0E1 reduced neuroblastoma cell proliferation and viability
ATP6V0E1 appears essential for cancer cell survival, suggesting its potential relevance in cancer biology
Recommended experimental models:
Cellular models:
Patient-derived fibroblasts or iPSCs
CRISPR-engineered cell lines with ATP6V0E1 mutations
Conditional knockdown systems (inducible shRNA)
Animal models:
Conditional knockout mice (since complete knockout may be lethal)
Zebrafish models for high-throughput phenotypic analysis
Drosophila models for genetic interaction studies
Organoid models:
Brain organoids for neurological disease modeling
Tumor organoids for cancer studies
Important considerations:
Due to the essential nature of V-ATPase function, complete loss of ATP6V0E1 may be lethal, necessitating conditional approaches
Compensatory mechanisms involving ATP6V0E2 should be considered
Tissue-specific phenotypes should be evaluated when designing disease models
Research on other V-ATPase subunits has demonstrated that even subtle functional changes can lead to tissue-specific disease manifestations, suggesting that careful phenotypic analysis across multiple systems is necessary.
While ATP6V0E1-specific therapeutics are not yet well-established, several approaches for targeting V-ATPase components show promise:
Current therapeutic approaches for V-ATPase targets:
Small molecule inhibitors:
RNA-based therapeutics:
Structure-based drug design:
Targeting specific interfaces between V-ATPase subunits
Allosteric modulators of V-ATPase assembly/disassembly
Optimized methods for ATP6V0E1-focused drug screening:
High-throughput screening approaches:
Cell-based assays measuring lysosomal pH (LysoSensor-based fluorescence)
ATP hydrolysis assays using purified V-ATPase complexes
Thermal shift assays to identify compounds binding directly to ATP6V0E1
Target validation methods:
Functional readouts for screening:
Emerging directions:
Research on colorectal cancer has shown that ATP6V0A1-dependent cholesterol absorption triggers immunosuppressive signaling, suggesting similar mechanisms might exist for ATP6V0E1
Targeting specific V-ATPase subunit isoforms may allow more precise modulation of compartment-specific functions
Developing compounds that modify V-ATPase assembly/disassembly rather than blocking activity completely may offer therapeutic advantages
Research has demonstrated that the V-ATPase regulates mTORC1 via the Rag GTPases, and that covalent targeting of ATP6V1A can decouple the V-ATPase from the Rags, leading to mTORC1 inhibition and autophagy activation . Similar target-specific approaches could be developed for ATP6V0E1.
The regulation of V-ATPase through assembly/disassembly is a sophisticated process that ATP6V0E1 likely contributes to:
V-ATPase assembly/disassembly regulation:
V-ATPase is regulated by reversible disassembly into autoinhibited V1-ATPase and V0 proton channel subcomplexes
The TLDc protein Oxr1p has been shown to induce V-ATPase disassembly in vitro and is essential for efficient disassembly in cells
ATP hydrolysis is needed for release of Oxr1p so that free V1 can adopt the autoinhibited conformation
Methods to study ATP6V0E1's role in V-ATPase assembly:
Biochemical approaches:
Co-immunoprecipitation with other V-ATPase subunits
Blue native PAGE to analyze intact complexes
Chemical crosslinking followed by mass spectrometry
In vitro reconstitution assays with purified components
Structural biology methods:
Cryo-electron microscopy of assembled complexes
X-ray crystallography of subdomains
Hydrogen-deuterium exchange mass spectrometry to map interaction sites
Dynamic interaction studies:
FRET/BRET pairs between ATP6V0E1 and other subunits
Split-GFP complementation assays
Single-molecule tracking in live cells
Important considerations:
The assembly state of V-ATPase changes in response to nutrient conditions
Different cellular compartments may have different assembly regulation mechanisms
ATP6V0E1 may interact with chaperones or assembly factors before incorporation into the complete complex
Research on yeast V-ATPase has shown that the protein Oxr1p is essential for efficient V-ATPase disassembly in vivo, with its absence resulting in ~40% more V1 on vacuoles compared to wild-type . This suggests that regulating assembly/disassembly is a critical control point for V-ATPase function, where ATP6V0E1 likely plays an important role.
Current challenges in ATP6V0E1 research:
Technical limitations:
Small size of ATP6V0E1 (9.4 kDa) making it difficult to detect
Potential functional redundancy with ATP6V0E2
Limited availability of highly specific antibodies
Challenges in reconstituting functional V-ATPase complexes in vitro
Biological complexity:
Compartment-specific roles of V-ATPase complexes
Cell type-specific functions and regulation
Integration with other pH regulatory mechanisms
Distinguishing between direct effects of ATP6V0E1 versus indirect effects of altered pH
Emerging techniques and solutions:
Advanced imaging approaches:
Expansion microscopy for improved resolution of small proteins
Live-cell super-resolution microscopy (PALM/STORM)
Cryo-electron tomography of intact cellular compartments
Correlative light and electron microscopy (CLEM)
Genomic and proteomic technologies:
Proximity labeling (BioID, APEX) to map compartment-specific interactomes
Single-cell transcriptomics to identify cell type-specific expression patterns
Targeted proteomics for accurate quantification of ATP6V0E1 and interacting proteins
CRISPR screening to identify genetic interactions
Functional analysis innovations:
Optogenetic control of ATP6V0E1 activity or localization
Genetically encoded pH sensors targeted to specific compartments
Organelle-specific isolation techniques for biochemical analysis
Microfluidic approaches for real-time measurement of ion transport
Research frontiers to explore:
Specialized cell types of particular interest:
Neurons (role in synaptic vesicle acidification)
Immune cells (antigen processing, phagocytosis)
Cancer cells (metabolic adaptation, drug resistance)
Renal tubular cells (acid-base homeostasis)
Emerging biological contexts:
Integration with metabolic pathways
Roles in signaling beyond pH regulation
Connections to membrane contact sites between organelles
Contributions to organelle identity and maturation
Research in Paramecium has revealed 17 genes encoding a-subunit isoforms with highly specific targeting to different compartments, demonstrating remarkable specialization within the V-ATPase family . This suggests that ATP6V0E1 may have similarly specialized functions that remain to be fully elucidated using these emerging techniques.
Challenges in ATP6V0E1 recombinant production:
Small protein size (9.4 kDa)
Membrane protein requiring proper folding environment
Potential need for other V-ATPase subunits for stability
Recommended expression systems:
Expression System | Advantages | Limitations | Special Considerations |
---|---|---|---|
E. coli | High yield, low cost | Limited post-translational modifications | Use membrane protein-optimized strains (C41/C43); consider fusion tags |
Insect cells | Mammalian-like processing, good for membrane proteins | Moderate cost, longer production time | Baculovirus expression with Sf9 or High Five cells |
Mammalian cells | Native processing and folding | Higher cost, lower yield | HEK293 or CHO cells with inducible expression systems |
Cell-free systems | Rapid, direct access to reaction conditions | Limited for membrane proteins | Supplement with lipids or nanodiscs for membrane proteins |
Optimization strategies:
Fusion tags and constructs:
Solubility-enhancing tags (MBP, SUMO, Trx)
Affinity tags (His, GST, FLAG) for purification
Fluorescent protein fusions for functional validation
Consider both N- and C-terminal tag positions
Membrane protein considerations:
Inclusion of detergents (DDM, CHAPS, Triton X-100)
Reconstitution in lipid nanodiscs or liposomes
Co-expression with partner subunits from V0 domain
Bicelle or amphipol formulations for structural studies
Purification approach:
Two-step affinity purification (e.g., His tag followed by second affinity tag)
Size exclusion chromatography for final polishing
On-column refolding for proteins expressed in inclusion bodies
Blue native PAGE to verify complex assembly
Validation methods:
Circular dichroism spectroscopy for secondary structure assessment
Thermal shift assays for stability analysis
Limited proteolysis to verify proper folding
Functional reconstitution assays measuring proton transport
Functional integrity assessment techniques:
Biochemical activity assays:
ATP hydrolysis measurements (colorimetric phosphate release assays)
Proton transport assays using pH-sensitive fluorescent dyes in reconstituted vesicles
ATPase coupled enzyme assays (e.g., pyruvate kinase/lactate dehydrogenase coupled system)
Structural integrity verification:
Limited proteolysis resistance compared to unfolded protein
Antibody recognition of conformational epitopes
Thermal stability measurement using differential scanning fluorimetry
Native mass spectrometry to assess complex formation
Interaction assays:
Surface plasmon resonance (SPR) to measure binding kinetics with other V-ATPase subunits
Microscale thermophoresis (MST) for protein-protein interaction studies
Isotherm titration calorimetry (ITC) for thermodynamic binding parameters
Pull-down assays with other purified V-ATPase components
V-ATPase complex assembly assessment:
Biochemical approaches:
Blue native PAGE to visualize intact complexes
Gradient ultracentrifugation to separate assembled complexes from individual components
Chemical crosslinking followed by SDS-PAGE or mass spectrometry
Co-immunoprecipitation with antibodies against other V-ATPase subunits
Structural analysis methods:
Negative stain electron microscopy for complex visualization
Cryo-electron microscopy for higher resolution structural analysis
Small-angle X-ray scattering (SAXS) for solution structure analysis
Hydrogen-deuterium exchange mass spectrometry to map interaction interfaces
Functional reconstitution:
Proteoliposome reconstitution with purified components
Activity measurements of reconstituted V-ATPase
Comparison of activities with and without ATP6V0E1
Patch-clamp electrophysiology of reconstituted complexes
Research on V-ATPase regulation has shown that ATP hydrolysis is crucial for the proper disassembly of the complex . Therefore, testing both assembly and disassembly dynamics in reconstituted systems would provide valuable insights into ATP6V0E1's role in V-ATPase function.
Genetic modification approaches:
Gene knockout technologies:
CRISPR-Cas9 for complete gene knockout
Conditional knockout systems (Cre-loxP, FLP-FRT)
Inducible degradation systems (auxin-inducible, dTAG)
Gene knockdown methods:
siRNA for transient knockdown
shRNA for stable knockdown
Antisense oligonucleotides for splice modulation
CRISPRi for transcriptional repression
Modification strategies:
Knock-in of point mutations to study specific residues
Tagging at endogenous loci (FLAG, HA, fluorescent proteins)
Domain swapping with ATP6V0E2 to identify functional regions
Reporter gene fusion for expression monitoring
Phenotypic assessment approaches:
Cellular phenotypes:
Organelle pH measurement with ratiometric probes
Lysosomal function assays (enzyme activity, cargo degradation)
Endocytosis and exocytosis rates
Autophagy flux analysis
Cell viability and proliferation
Molecular phenotypes:
Transcriptomic analysis to identify compensatory responses
Proteomic profiling of V-ATPase complex components
Metabolomic analysis for changes in pH-dependent pathways
Lipidomic analysis for membrane composition changes
Tissue and organism phenotypes:
Tissue-specific conditional knockout phenotyping
Developmental analysis in model organisms
Histological assessment for morphological changes
Physiological measurements (e.g., renal acid secretion)
Important considerations:
Complete knockout may be lethal, so inducible or cell type-specific approaches may be necessary
Compensatory upregulation of ATP6V0E2 should be monitored
Phenotypes may manifest differently across cell types and tissues
Combined approaches (e.g., knockdown plus rescue experiments) provide stronger evidence
Research in neuroblastoma cells demonstrated that siRNA knockdown of ATP6V0E1 reduced cell proliferation and viability, suggesting essential functions for cell survival . This highlights the importance of carefully titrated genetic modification approaches and comprehensive phenotypic analysis.
Experimental design strategies:
Comparative subunit analysis:
Parallel knockout/knockdown of different V-ATPase subunits
Comparison with general V-ATPase inhibitors (bafilomycin A1, concanamycin A)
Analysis of ATP6V0E1 vs. ATP6V0E2 (paralog) knockdown effects
Domain-swap chimeras between ATP6V0E1 and other subunits
Targeted rescue experiments:
Rescue of ATP6V0E1 knockout with wildtype vs. mutant versions
Rescue with ATP6V0E2 to test functional redundancy
Rescue with orthologous ATP6V0E1 from other species
Rescue with minimal domains to identify functional regions
Structure-function analysis:
Point mutations of conserved residues vs. isoform-specific residues
Identification of post-translational modification sites specific to ATP6V0E1
Analysis of protein-protein interaction interfaces
Compartment-specific targeting signals
Technical approaches for distinguishing specific effects:
Temporal analysis:
Acute vs. chronic depletion to separate direct vs. compensatory effects
Time-resolved phenotypic analysis after perturbation
Pulse-chase experiments for dynamic processes
Spatial analysis:
Organelle-specific pH measurements
Subcellular fractionation followed by biochemical analysis
High-resolution imaging of specific compartments
Targeted mass spectrometry of isolated organelles
Context-dependent analysis:
Cell type-specific effects
Stress conditions that may reveal specialized functions
Developmental stage-specific phenotypes
Disease-relevant contexts
Case study examples:
Research on V-ATPase a-subunit isoforms in Paramecium revealed 17 genes with highly specific targeting to at least seven different compartments, confirming functional specialization
Studies on ATP6V0A1 showed that it contributes to endolysosomal acidification and its knockdown leads to specific effects on autophagy and mitochondrial function
Research on dominant ATP6V1C1/ATP6V1B2 variants demonstrated gain-of-function effects leading to increased lysosomal acidification and disrupted lysosomal morphology
These approaches help distinguish between general effects of disrupting the V-ATPase complex versus specific roles of individual subunits like ATP6V0E1 in particular cellular contexts.
Systems biology approaches for ATP6V0E1:
Network analysis methods:
Protein-protein interaction mapping (Y2H, AP-MS, BioID)
Genetic interaction screens (CRISPR screens, synthetic lethality)
Transcriptomic analysis after ATP6V0E1 perturbation
Pathway enrichment analysis integrating multi-omics data
Mathematical modeling approaches:
Kinetic modeling of V-ATPase assembly/disassembly dynamics
pH regulation network models including V-ATPase function
Integration of ATP6V0E1 into cellular metabolism models
Agent-based models of organelle pH maintenance
Multi-omics integration:
Combined analysis of transcriptomics, proteomics, and metabolomics data
Correlation of ATP6V0E1 expression with other genes across cell types
Mapping ATP6V0E1 to known pH-responsive pathways
Network visualization tools for data integration
Relevant biological pathways to investigate:
mTORC1 signaling pathway:
Autophagy-lysosome system:
V-ATPase activity is critical for autophagy progression
Investigate ATP6V0E1's role in autophagic flux
Study connections to lysosomal biogenesis pathways (CLEAR network)
Endocytic trafficking:
Map ATP6V0E1 to Rab GTPase networks
Analyze effects on receptor recycling vs. degradation pathways
Investigate connections to ESCRT machinery
Metabolic networks:
Study how ATP6V0E1 integrates with cellular energy metabolism
Investigate connections to lipid metabolism (especially cholesterol)
Analyze pH-dependent metabolic enzymes affected by ATP6V0E1 function
Research has shown that V-ATPase subunits interact with key regulators like RABGEF1, which affects endosome maturation and is crucial for processes like cholesterol absorption . Similarly, covalent targeting of ATP6V1A revealed connections between the V-ATPase and mTORC1 signaling . These findings highlight the importance of investigating ATP6V0E1 within broader cellular networks.
Computational prediction approaches:
Structural bioinformatics:
Homology modeling of ATP6V0E1 structure
Molecular docking to predict protein-protein interactions
Molecular dynamics simulations to study conformational changes
Identification of functional domains and motifs
Sequence-based predictions:
Evolutionary conservation analysis to identify critical residues
Co-evolution analysis to predict interaction partners
Post-translational modification site prediction
Subcellular localization signal prediction
Advanced machine learning methods:
Deep learning for protein function prediction
Network-based function prediction algorithms
Text mining of scientific literature for association discovery
Integration of multi-omics data for functional prediction
Validation approaches:
Structure-guided experimental design:
Site-directed mutagenesis based on computational predictions
Domain deletion/swapping guided by structural models
Design of peptide inhibitors targeting predicted interfaces
Structure-based drug design for ATP6V0E1 modulation
Network-based validation:
Testing predicted genetic interactions via CRISPR screens
Validation of physical interactions using co-IP or proximity labeling
Testing predicted pathway connections with targeted inhibitors
Perturbation studies of computationally identified hubs
Integrative data analysis:
Correlation analysis across large-scale datasets
Patient data mining for clinical correlations
Meta-analysis of ATP6V0E1-related studies
Multivariate statistical approaches to identify patterns
Emerging computational tools:
AlphaFold2 and RoseTTAFold for accurate protein structure prediction
Molecular dynamics platforms (GROMACS, NAMD) for studying dynamic processes
Network analysis tools (Cytoscape, STRING) for visualizing interactions
Omics data integration platforms (Galaxy, Bioconductor) for multi-level analysis
Pathway enrichment tools (GSEA, Metascape) for biological context
Research on V-ATPase has benefited from computational approaches, with structural studies revealing mechanisms of assembly/disassembly regulation and interaction networks identifying connections to cellular processes like cholesterol absorption and immune signaling . Similar approaches applied to ATP6V0E1 could reveal new functions and therapeutic targets.