ATP6V0A1 (UniProt ID: Q93050) encodes the 116 kDa a1-subunit of the V0 domain in V-ATPase, a multisubunit proton pump. This enzyme acidifies intracellular compartments (e.g., lysosomes, endosomes) and is essential for:
Synaptic vesicle acidification and neurotransmitter storage .
Cholesterol absorption in cancer cells, influencing immune evasion .
The ATP6V0A1 antibody targets epitopes within its 837-amino acid sequence, often validated in human, mouse, and rat samples .
Role in Colorectal Cancer (CRC):
ATP6V0A1 drives RABGEF1-dependent cholesterol absorption in CRC cells, activating TGF-β1/SMAD3 signaling to suppress memory CD8+ T cells .
High ATP6V0A1 expression correlates with decreased immune activity and poor survival in CRC patients (HR = 2.1, p < 0.0001) .
Therapeutic targeting with daclatasvir (an ATP6V0A1 inhibitor) restores CD8+ T cell activity and reduces tumor growth in murine models .
Developmental and Epileptic Encephalopathy:
ATP6V0A1 colocalizes with lysosomal markers (Lamp2) and V1-domain subunits (ATP6V1A) in HEK293 and N2A cells .
Misrouting to the plasma membrane in Cln1−/− mice disrupts AP-3 interaction, altering lysosomal function .
The ATP6V0A1 antibody is designed to recognize ATP6V0A1 proteins from human, mouse, and rat samples. This antibody is generated through a robust immunization process. A rabbit was immunized with recombinant human ATP6V0A1 protein (amino acids 80-260), eliciting a strong immune response and the production of antibodies against the target protein. The resulting antiserum was then subjected to protein G purification, yielding a highly pure ATP6V0A1 polyclonal antibody (purity exceeding 95%). This antibody has demonstrated excellent specificity in various applications including ELISA, Western blotting (WB), immunohistochemistry (IHC), and immunofluorescence (IF).
ATP6V0A1, a subunit of the vacuolar ATPase (V-ATPase) enzyme, plays a pivotal role in proton translocation across cellular membranes. It is indispensable for the proper functioning of the V-ATPase, which is responsible for maintaining the pH balance of acidic compartments within cells. ATP6V0A1's critical functions encompass protein degradation, membrane trafficking, and ion homeostasis. Notably, mutations in ATP6V0A1 have been associated with several disorders, including renal tubular acidosis and osteopetrosis.
ATP6V0A1 (ATPase, H+ transporting, lysosomal V0 subunit a1) is a critical component of vacuolar ATPase (V-ATPase), a multisubunit enzyme responsible for mediating acidification of eukaryotic intracellular organelles. It serves as part of the membrane-embedded V0 domain that facilitates proton translocation across cellular membranes. This protein is essential for maintaining pH balance in acidic compartments within cells and plays vital roles in multiple cellular processes including protein degradation, membrane trafficking, and ion homeostasis . Recent research has identified ATP6V0A1 as a novel immunosuppressive factor in colorectal cancer, where it promotes RABGEF1-dependent cholesterol absorption and subsequently initiates paracrine TGF-β1/SMAD3 signaling to deactivate memory CD8+ T cells . This mechanism represents a previously uncharacterized pathway by which tumor cells can evade immune responses.
ATP6V0A1 antibodies have been validated for multiple research applications with specific recommended dilutions:
Application | Validated Dilution (Proteintech 13828-1-AP) | Validated Dilution (CUSABIO CSB-PA846292LA01HU) |
---|---|---|
Western Blot (WB) | 1:1000-1:6000 | 1:500-1:5000 |
Immunohistochemistry (IHC) | 1:50-1:500 | 1:20-1:200 |
Immunoprecipitation (IP) | 0.5-4.0 μg for 1.0-3.0 mg protein lysate | Not specified |
Immunofluorescence (IF) | Published applications confirmed | 1:50-1:200 |
ELISA | Published applications confirmed | Confirmed for conjugated versions |
Both antibodies have been rigorously tested and validated in peer-reviewed publications across multiple techniques, with demonstrated reactivity in human, mouse, and rat samples . For optimal results, it is recommended to titrate the antibody concentration in each specific experimental system to account for variations in tissue type, sample preparation, and detection methods.
ATP6V0A1 antibodies have demonstrated reliable detection in specific tissues with important methodological considerations. Positive Western blot detection has been confirmed in mouse and rat brain tissues, suggesting particularly high expression or favorable detection conditions in neural tissues . For immunohistochemistry applications in brain tissues, antigen retrieval becomes a critical step. The recommended protocol utilizes TE buffer at pH 9.0, though citrate buffer at pH 6.0 serves as an acceptable alternative . This difference in buffer systems highlights the importance of optimizing antigen retrieval conditions when working with different tissue types. Researchers studying ATP6V0A1 in colorectal cancer contexts should note that while the protein is detectable in these tissues, optimization may be required due to potentially varying expression levels based on the tumor's lipid metabolism status and immune activity profile .
The calculated molecular weight of ATP6V0A1 (96 kDa based on 837 amino acids) differs from its observed molecular weight of approximately 116 kDa on SDS-PAGE . This 20 kDa discrepancy likely results from post-translational modifications, particularly glycosylation patterns that are common in membrane proteins. When troubleshooting western blots showing unexpected band patterns:
Verify antibody specificity through knockout/knockdown validation controls if available (publications have confirmed KD/KO applications) .
Consider enzymatic deglycosylation treatments (PNGase F or Endo H) prior to SDS-PAGE to determine if glycosylation accounts for the size difference.
Explore tissue-specific variations, as expression patterns may differ between brain tissues (where validation is robust) and other sample types.
If investigating ATP6V0A1 in cancer contexts, note that malignant transformation may alter post-translational modification patterns, potentially resulting in subtle changes to the observed molecular weight .
This molecular weight consideration becomes particularly important when analyzing ATP6V0A1 in different disease contexts, where altered glycosylation might correlate with pathological states.
Immunoprecipitation (IP) of ATP6V0A1 requires careful optimization due to its membrane-embedded nature and association with other V-ATPase subunits. Based on validated protocols:
Lysis buffer composition is critical - use buffer containing 1% NP-40 or Triton X-100, 150 mM NaCl, 50 mM Tris-HCl (pH 7.4), and protease inhibitors.
For brain tissue samples (where IP has been confirmed successful):
When investigating ATP6V0A1 interactions with potential binding partners (like RABGEF1 in colorectal cancer cells), consider:
Crosslinking prior to lysis to stabilize transient interactions
Sequential IP approaches to capture multi-protein complexes
Modified wash conditions that maintain specific interactions while removing background
For co-immunoprecipitation validations, reciprocal IP with antibodies against known V-ATPase complex components provides important confirmation of specificity.
These optimized IP approaches are particularly valuable when investigating novel interaction partners of ATP6V0A1, such as those involved in the recently identified cholesterol absorption pathway in colorectal cancer .
ATP6V0A1's involvement in both normal cellular physiology and cancer pathology necessitates careful experimental design. Recent findings demonstrate that tumor cell-intrinsic ATP6V0A1 suppresses anti-tumor immune responses, particularly in colorectal cancer contexts with high lipid metabolism . When designing experiments to investigate this dual functionality:
Include appropriate control systems:
Consider lipid metabolism context:
Implement parallel in vitro and in vivo approaches:
Evaluate downstream pathways methodically:
Assess RABGEF1-dependent cholesterol absorption
Measure paracrine TGF-β1/SMAD3 signaling
Quantify memory CD8+ T cell activation status
These methodological considerations ensure robust experimental design that can distinguish between ATP6V0A1's normal physiological functions and its pathological roles in cancer contexts.
When investigating ATP6V0A1 in disease contexts, antibody selection requires careful consideration of several factors:
Epitope location considerations:
Cross-reactivity validation:
Application-specific optimization:
For investigations of ATP6V0A1 in colorectal cancer immune evasion mechanisms, antibodies that successfully detect the protein in both western blot and immunohistochemistry applications are preferable
In complex tissue environments, higher antibody concentrations within the recommended range may be necessary
Control strategies:
These optimization approaches ensure reliable detection of ATP6V0A1 across diverse experimental contexts, particularly important when investigating novel functions like its role in cancer immunosuppression.
Recent research has uncovered ATP6V0A1's critical involvement in colorectal cancer immune evasion through cholesterol metabolism . When designing experiments to further investigate this pathway:
Cholesterol absorption assessment techniques:
Utilize fluorescently labeled cholesterol analogs to quantify uptake rates
Measure intracellular cholesterol accumulation through filipin staining
Assess membrane cholesterol through methyl-β-cyclodextrin extraction and quantification
RABGEF1-ATP6V0A1 interaction analysis:
Implement proximity ligation assays to visualize protein interactions in situ
Utilize FRET-based approaches to assess dynamic interactions
Confirm interactions through co-immunoprecipitation with appropriate controls
T cell functional assessment:
Design co-culture systems with ATP6V0A1-manipulated cancer cells and isolated memory CD8+ T cells
Quantify T cell activation through multiple parameters (cytokine production, cytotoxicity, activation markers)
Implement flow cytometry to assess memory CD8+ T cell functional status
In vivo verification approaches:
Compare tumor growth kinetics in immunocompetent versus immunodeficient mouse models
Analyze tumor-infiltrating lymphocyte populations through flow cytometry and spatial transcriptomics
Assess responses to immune checkpoint inhibition in ATP6V0A1-high versus ATP6V0A1-low tumors
These methodological considerations enable comprehensive investigation of ATP6V0A1's newly identified role in establishing an immunosuppressive tumor microenvironment through cholesterol metabolism manipulation.
The emerging literature reveals context-dependent functions of ATP6V0A1 that may appear contradictory without proper interpretation. When analyzing apparently discrepant results:
Consider cellular context specificity:
ATP6V0A1's primary role in normal physiology involves acidification of intracellular organelles
In colorectal cancer, it adopts additional functions in cholesterol metabolism and immune suppression
These divergent functions likely reflect repurposing of normal cellular machinery in the tumor context
Evaluate interaction partners systematically:
Assess regulatory mechanism differences:
Consider evolutionary context:
This interpretive framework helps reconcile seemingly contradictory observations about ATP6V0A1 functions across different experimental systems and disease contexts.
The identification of ATP6V0A1 as a driver of immune suppression in colorectal cancer presents opportunities for therapeutic targeting . Researchers investigating this potential should consider:
Target validation methodologies:
Implement conditional knockout systems in established tumor models
Utilize domain-specific mutations to identify critical functional regions
Develop selective inhibitors through structure-guided design approaches
Therapeutic assessment parameters:
Monitor changes in tumor immune microenvironment following ATP6V0A1 inhibition
Assess combination potential with existing immunotherapies
Evaluate effects on tumor growth versus normal tissue homeostasis
Biomarker development approaches:
Correlate ATP6V0A1 expression with response to immunotherapy in patient cohorts
Develop assays to measure ATP6V0A1-dependent cholesterol metabolism activity
Identify patient stratification parameters based on lipid metabolism status
Delivery system considerations:
Explore tumor-targeted siRNA/shRNA delivery systems
Investigate small molecule inhibitors with favorable pharmacokinetic properties
Consider antibody-drug conjugates targeting cells with high ATP6V0A1 expression
These methodological approaches provide a framework for translating the basic science discovery of ATP6V0A1's role in cancer immune evasion into potential therapeutic strategies.
ATP6V0A1 functions as part of the multi-subunit V-ATPase complex, necessitating specialized approaches to study subunit interactions:
Structural biology approaches:
Cryo-EM analysis of V-ATPase complexes with and without ATP6V0A1
Crosslinking mass spectrometry to map intersubunit contact points
Hydrogen-deuterium exchange mass spectrometry to identify dynamic interaction surfaces
Functional reconstitution strategies:
Develop in vitro reconstitution systems with purified components
Utilize liposome-based proton pumping assays to assess functional consequences of subunit interactions
Implement targeted mutations at predicted interaction interfaces
Live-cell imaging methodologies:
Apply bimolecular fluorescence complementation to visualize protein-protein interactions
Utilize FRET-based sensors to monitor dynamic assembly/disassembly
Implement single-molecule tracking to assess complex formation kinetics
Computational prediction validation:
Generate interaction models based on related ATPase structures
Validate through targeted mutagenesis of predicted interface residues
Correlate functional consequences with structural alterations
These approaches enable detailed characterization of how ATP6V0A1 interfaces with other V-ATPase subunits in different cellular contexts, potentially revealing how these interactions are modified in disease states.
Recent research has implicated several V-ATPase components in human disease, including ATP6V1A in encephalopathy and ATP6V1C1/ATP6V1B2 in multisystem phenotypic disorders . When investigating potential connections:
Functional redundancy assessment:
Determine whether ATP6V0A1 functions can be compensated by other family members
Implement simultaneous knockdown/knockout experiments to identify synthetic interactions
Assess tissue-specific expression patterns that might explain differential disease manifestations
Mutation impact comparison:
Compare effects of disease-associated variants across family members
Assess whether mutations cluster in analogous functional domains
Evaluate differential effects on proton pumping versus potential moonlighting functions
Pathway integration analysis:
Investigate whether ATP6V0A1's role in cholesterol metabolism is shared by other family members
Determine if immunosuppressive functions extend to mutations in ATP6V1A, ATP6V1C1, or ATP6V1B2
Map shared versus distinct interactomes across family members
Model system coordination:
Develop comparable cellular and animal models for different V-ATPase component disorders
Implement consistent phenotyping approaches across models
Utilize rescue experiments to assess functional overlap
These considerations facilitate comparison across the emerging spectrum of V-ATPase-associated disorders, potentially revealing common mechanisms and therapeutic opportunities applicable across multiple conditions.