ATP6V1G2 Antibody

Shipped with Ice Packs
In Stock

Description

Overview of ATP6V1G2 Antibody

ATP6V1G2 antibodies are polyclonal reagents (rabbit IgG) that target the human ATP6V1G2 protein, encoded by the ATP6V1G2 gene. This protein is part of the V1 domain of V-ATPase, a multisubunit enzyme responsible for acidifying intracellular compartments (e.g., lysosomes, synaptic vesicles) to enable processes like neurotransmitter packaging and immune cell function . Key characteristics include:

  • Molecular Weight: 14 kDa (observed and calculated) .

  • Immunogen: Typically a recombinant full-length protein or fusion protein .

  • Reactivity: Confirmed in human, mouse, and rat samples .

Table 1: Recommended Dilutions and Applications

ApplicationDilution RangeKey Findings/Validation Data
Western Blot1:500–1:5000Detected in HeLa, A549, and brain lysates .
Immunohistochemistry1:20–1:500Strong signal in mouse/rat brain tissues .
ELISANot specifiedUsed for quantitative protein analysis .

Key Research Findings

  • Compensatory Upregulation: Atp6v1g2 knockout mice showed no developmental defects, but exhibited increased expression of the G1 subunit (ATP6V1G1) in brain tissues, suggesting functional redundancy .

  • Neuronal Specificity: ATP6V1G2 is predominantly expressed in the brain, where it facilitates synaptic vesicle acidification for neurotransmitter storage .

  • Immune System Role: Macrophage activation upregulates ATP6V1G2, linking it to phagocytosis and inflammatory responses .

Clinical and Functional Insights

  • Disease Associations: Dysfunctional ATP6V1G2 is implicated in Noonan Syndrome, distal renal tubular acidosis, and autoimmune disorders .

  • Mechanistic Role:

    • Enables V-ATPase assembly by bridging V1 (cytosolic) and V0 (membrane) domains .

    • Critical for proton gradient generation in osteoclasts (bone remodeling) and kidney epithelial cells .

Validation and Quality Control

  • Specificity: Antibodies show no cross-reactivity with other V-ATPase subunits (e.g., G1 or G3) .

  • Storage: Stable at -20°C in PBS with 0.02% sodium azide .

  • Controls: Validated using KO mouse models and siRNA-mediated gene silencing .

Limitations and Considerations

  • Tissue Specificity: Limited reactivity in non-neuronal tissues (e.g., kidneys) .

  • Compensation Effects: G1 subunit upregulation in G2 KO models may complicate data interpretation .

Product Specs

Buffer
**Preservative:** 0.03% Proclin 300
**Constituents:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship your orders within 1-3 business days of receipt. Delivery timelines may vary depending on the chosen shipping method and destination. For precise delivery estimates, we recommend contacting your local distributor.
Synonyms
ATP6G antibody; ATP6G2 antibody; ATP6V1G2 antibody; ATPase H+ transporting lysosomal (vacuolar proton pump) subunit G antibody; ATPase H+ transporting lysosomal 13kDa V1 subunit G2 antibody; H(+) transporting two sector ATPase subunit G2 antibody; NG 38 antibody; NG38 antibody; OTTHUMP00000036058 antibody; OTTHUMP00000036060 antibody; V ATPase 13 kDa subunit 2 antibody; V ATPase G subunit 2 antibody; V ATPase subunit G 2 antibody; V-ATPase 13 kDa subunit 2 antibody; V-ATPase subunit G 2 antibody; V-type proton ATPase subunit G 2 antibody; Vacuolar ATP synthase subunit G 2 antibody; Vacuolar proton pump G subunit 2 antibody; Vacuolar proton pump subunit G 2 antibody; VATG2_HUMAN antibody; VMA 10 antibody; VMA10 antibody
Target Names
Uniprot No.

Target Background

Function
The ATP6V1G2 Antibody targets the catalytic subunit of the peripheral V1 complex of vacuolar ATPase (V-ATPase). V-ATPase plays a crucial role in acidifying various intracellular compartments within eukaryotic cells.
Database Links

HGNC: 862

OMIM: 606853

KEGG: hsa:534

STRING: 9606.ENSP00000302194

UniGene: Hs.249227

Protein Families
V-ATPase G subunit family
Subcellular Location
Melanosome. Note=Highly enriched in late-stage melanosomes.
Tissue Specificity
Brain.

Q&A

What is ATP6V1G2 and what cellular functions does it perform?

ATP6V1G2 (ATPase, H+ transporting, lysosomal 13kDa, V1 subunit G2) is a subunit of the V1 complex of vacuolar(H+)-ATPase (V-ATPase), a multisubunit enzyme that plays a crucial role in cellular pH regulation. The protein functions as part of a peripheral complex (V1) that hydrolyzes ATP and works in concert with a membrane integral complex (V0) that translocates protons. V-ATPase is responsible for acidifying and maintaining the pH of intracellular compartments, and in some cell types, it is targeted to the plasma membrane where it acidifies the extracellular environment . ATP6V1G2 has a calculated molecular weight of 14 kDa and consists of 118 amino acids . It is primarily expressed in brain tissue, with positive Western Blot detection in rat brain tissue and positive IHC detection in mouse brain tissue .

Most commercially available ATP6V1G2 antibodies show reactivity with human, mouse, and rat samples . Specifically, positive Western Blot detection has been documented in rat brain tissue, while positive IHC detection has been observed in mouse brain tissue . When performing immunohistochemistry, it is suggested to use antigen retrieval with TE buffer pH 9.0, although citrate buffer pH 6.0 may be used as an alternative . For human samples, the antibody detects endogenous levels of the protein .

What are the optimal storage conditions for ATP6V1G2 antibodies?

For maximum stability and activity retention, ATP6V1G2 antibodies should be stored at -20°C . Under these conditions, most antibodies remain stable for up to one year after shipment . It is generally recommended to avoid repeated freeze-thaw cycles as this can compromise antibody performance . Most commercial ATP6V1G2 antibodies are supplied in liquid form, typically in PBS containing 50% glycerol and 0.02% sodium azide with a pH of approximately 7.3 . For small-volume antibodies (around 20 μl), some manufacturers include 0.1% BSA in the formulation .

How can ATP6V1G2 antibodies be used in glioma research and what findings have emerged?

Recent studies have identified ATP6V1G2 as part of a five-gene signature derived from the V-ATPase complex that is associated with glioma patient prognosis . Using bioinformatics analyses, researchers discovered that ATP6V1G2, along with ATP6V1C2, TCIRG1, ATP6AP1, and ATP6AP2, can be used to sub-classify glioma patients into different prognostic clusters .

Single-cell RNA-seq data revealed that ATP6AP1, ATP6AP2, ATP6V1G2, and TCIRG1 might serve as cell-type potential markers . High protein expression levels of ATP6V1G2, ATP6AP1, and ATP6AP2 were found in normal brain tissues as documented in the CPTAC database . The aberrant expression of these genes appears to be regulated by copy number variation and DNA promoter methylation, and is associated with alterations in the immune microenvironment .

When conducting research on ATP6V1G2 in the context of gliomas, it is advisable to employ multiple detection methods, including IHC and Western blot, to validate expression patterns across different patient samples. This multi-modal approach helps to establish the reliability of findings regarding the role of ATP6V1G2 in glioma pathogenesis and prognosis.

What methodological considerations should be taken into account when performing Western Blot for ATP6V1G2?

When performing Western Blot to detect ATP6V1G2, several methodological considerations are crucial for obtaining reliable and reproducible results:

  • Sample Preparation: Brain tissue samples (particularly from rat or mouse) have shown positive results for ATP6V1G2 detection . Proper homogenization and protein extraction protocols are essential.

  • Blocking and Antibody Incubation:

    • Primary antibody dilution: 1:500-1:5000 depending on the specific antibody used

    • Incubation times and temperatures should be optimized according to the manufacturer's protocol

  • Detection: ATP6V1G2 should be observed at approximately 14 kDa on Western blots

  • Controls: Due to the specificity of ATP6V1G2 expression in certain tissues, positive controls (such as brain tissue lysates) should be included to validate the experimental procedure

  • Troubleshooting: If non-specific binding occurs, optimization of blocking conditions or further purification of the sample may be necessary

Most manufacturers provide specific Western Blot protocols optimized for their ATP6V1G2 antibodies, which should be followed for best results .

What are the challenges in ATP6V1G2 detection across different experimental systems and how can they be addressed?

Detecting ATP6V1G2 across various experimental systems presents several challenges that researchers should be aware of:

  • Tissue-Specific Expression: ATP6V1G2 shows highest expression in brain tissue, which may make detection in other tissues challenging . When working with non-neural tissues, more sensitive detection methods may be required.

  • Cross-Reactivity: Some ATP6V1G2 antibodies may cross-react with other V-ATPase G subunit isoforms (G1 or G3). Validation of antibody specificity using knockout or knockdown systems is recommended for critical experiments.

  • Antigen Retrieval for IHC: For immunohistochemistry applications, proper antigen retrieval is crucial. TE buffer at pH 9.0 is generally recommended, though citrate buffer at pH 6.0 may also be used as an alternative . Optimization of antigen retrieval conditions may be necessary depending on the specific tissue and fixation method.

  • Variable Results Across Species: While most ATP6V1G2 antibodies show reactivity with human, mouse, and rat samples , there may be species-specific variations in epitope recognition and binding efficiency. When working with samples from species not explicitly tested by the manufacturer, additional validation steps are recommended.

  • Dilution Optimization: The recommended dilution ranges can vary significantly (e.g., 1:20-1:500 for IHC ), and the optimal dilution is often sample-dependent . Titration experiments should be conducted to determine the optimal antibody concentration for each specific application and sample type.

To address these challenges, researchers should:

  • Conduct thorough validation experiments with appropriate positive and negative controls

  • Consider using multiple antibodies targeting different epitopes of ATP6V1G2

  • Optimize protocols for specific sample types and experimental conditions

  • Perform complementary experiments (e.g., mRNA detection) to corroborate protein expression data

How do the different commercially available ATP6V1G2 antibodies compare in terms of epitope recognition and experimental performance?

Several ATP6V1G2 antibodies are commercially available, each with specific characteristics that may influence their performance in different experimental contexts:

ManufacturerCatalog #Immunogen/EpitopeApplicationsHost/TypeSpecial Features
Proteintech25316-1-APATP6V1G2 fusion protein Ag18007WB, IHC, ELISARabbit/PolyclonalValidated in 3 publications
St John's LabsSTJ1916171-50 aa of human proteinWB, ELISARabbit/PolyclonalAvailable with various conjugates (HRP, FITC, Biotin)
CUSABIOCSB-PA002409LA01HUNot specified in search resultsWB, IHC, IFRabbit/PolyclonalAvailable with multiple conjugates
Sigma-AldrichHPA068667NLSAEVEQATRRQVQGMQSSQQRNRERVLAQLLGMVCICC-IF, IHCRabbit/PolyclonalEnhanced validation with orthogonal RNAseq

When selecting an ATP6V1G2 antibody for specific applications, researchers should consider:

  • Epitope Recognition: Antibodies targeting different regions of ATP6V1G2 may perform differently depending on protein folding, post-translational modifications, or protein-protein interactions that could mask certain epitopes.

  • Validation Method: Some antibodies, like Sigma's HPA068667, have undergone enhanced validation through orthogonal methods such as RNAseq , which may provide additional confidence in specificity.

  • Published Validations: Antibodies that have been used in peer-reviewed publications, such as Proteintech's 25316-1-AP , have demonstrated utility in research settings.

  • Required Applications: Not all antibodies are validated for all applications. For example, if immunofluorescence is needed, CUSABIO's antibody (1:50-1:200 dilution) or Sigma's HPA068667 (0.25-2 μg/mL) would be appropriate choices.

For critical experiments, it is advisable to test multiple antibodies in parallel to identify the one that performs optimally in the specific experimental context.

What are common troubleshooting strategies for weak or absent ATP6V1G2 signal in Western Blot?

When facing weak or absent ATP6V1G2 signal in Western Blot experiments, consider implementing the following troubleshooting strategies:

  • Sample Preparation:

    • Ensure you are using appropriate tissue samples; ATP6V1G2 is highly expressed in brain tissue, particularly rat brain for WB detection

    • Use fresh samples or properly stored frozen samples to minimize protein degradation

    • Include protease inhibitors in your lysis buffer to prevent protein degradation

    • Increase the protein concentration loaded onto the gel (up to 50-100 μg per lane may be necessary)

  • Antibody Optimization:

    • Try a more concentrated antibody dilution (e.g., 1:500 instead of 1:1000)

    • Extend primary antibody incubation time (overnight at 4°C may improve signal)

    • Ensure the antibody is stored properly and has not expired or degraded

  • Detection System:

    • Switch to a more sensitive detection system (e.g., enhanced chemiluminescence or ECL-plus)

    • Extend exposure time when imaging the blot

    • Consider using a signal amplification system

  • Transfer Efficiency:

    • Optimize protein transfer conditions, particularly for the 14 kDa size range where ATP6V1G2 is expected

    • Consider using PVDF membrane instead of nitrocellulose for better protein retention

    • Verify transfer efficiency with reversible protein stains like Ponceau S

  • Buffer and Blocking Optimization:

    • Test different blocking agents (e.g., BSA vs. milk)

    • Reduce the concentration of detergents in washing buffers

    • Ensure optimal pH of buffers used throughout the protocol

If the signal remains problematic after these adjustments, consider validating your experimental system with a positive control sample known to express ATP6V1G2, such as rat brain tissue lysate .

How should researchers address non-specific binding when using ATP6V1G2 antibodies in immunohistochemistry?

Non-specific binding in immunohistochemistry with ATP6V1G2 antibodies can lead to misleading results. To address this issue, researchers should implement the following strategies:

  • Optimize Blocking Conditions:

    • Extend blocking time (1-2 hours at room temperature)

    • Test different blocking agents (BSA, normal serum, commercial blocking solutions)

    • Use blocking serum from the same species as the secondary antibody

  • Antibody Dilution and Incubation:

    • Test a range of primary antibody dilutions, starting with the manufacturer's recommendation (1:50-1:500 for IHC)

    • Reduce incubation temperature (4°C instead of room temperature)

    • Include 0.1-0.3% Triton X-100 in antibody diluent to improve penetration and reduce non-specific membrane binding

  • Antigen Retrieval Optimization:

    • For ATP6V1G2, both TE buffer pH 9.0 and citrate buffer pH 6.0 have been suggested for antigen retrieval

    • Optimize antigen retrieval time and temperature

    • Consider testing different retrieval methods (heat-induced vs. enzymatic)

  • Controls:

    • Include a negative control by omitting the primary antibody

    • Use tissue known not to express ATP6V1G2 as a negative control

    • Include positive control tissue (mouse brain tissue is recommended)

    • Consider using peptide competition assays to verify antibody specificity

  • Washing Protocols:

    • Increase the number and duration of washes between steps

    • Use buffers with appropriate salt concentration to reduce non-specific ionic interactions

  • Secondary Antibody Selection:

    • Use highly cross-adsorbed secondary antibodies to minimize cross-species reactivity

    • Optimize secondary antibody dilution

By systematically addressing these aspects of the IHC protocol, researchers can significantly reduce non-specific binding and improve the specificity of ATP6V1G2 detection in tissue samples.

What role does ATP6V1G2 play in the context of neurodegenerative diseases and can antibodies be used to study this relationship?

While the search results don't directly address ATP6V1G2's role in neurodegenerative diseases, several important inferences can be made based on the protein's function and expression pattern:

ATP6V1G2 is a subunit of V-ATPase, which is responsible for acidification of intracellular compartments . In neurons, proper pH regulation is critical for multiple processes including synaptic vesicle loading, endosomal trafficking, and protein degradation pathways. Dysfunction in these processes has been implicated in various neurodegenerative conditions.

Research strategies to investigate ATP6V1G2's role in neurodegeneration could include:

  • Expression Analysis: Using ATP6V1G2 antibodies for IHC or Western blotting to compare expression levels between normal brain tissue and tissues affected by neurodegenerative diseases

  • Co-localization Studies: Performing dual-labeling immunofluorescence with ATP6V1G2 antibodies (recommended dilution 1:50-1:200) and markers of neurodegeneration to examine spatial relationships

  • Functional Studies: Employing ATP6V1G2 antibodies in conjunction with pH-sensitive fluorescent probes to assess whether V-ATPase function is altered in disease models

  • Interaction Studies: Using ATP6V1G2 antibodies for co-immunoprecipitation experiments to identify novel protein interactions that might be relevant to disease pathways

Given that ATP6V1G2 shows high expression in brain tissue , and its function in V-ATPase is critical for neuronal homeostasis, future research using specific antibodies could provide valuable insights into how disruptions in vesicular acidification contribute to neurodegenerative pathogenesis.

How can ATP6V1G2 antibodies be utilized in the study of glioma progression and potential therapeutic targets?

ATP6V1G2 has been identified as part of a five-gene signature from the V-ATPase complex that is associated with glioma patient prognosis . Researchers can leverage ATP6V1G2 antibodies in several strategic ways to advance understanding of glioma pathogenesis and identify potential therapeutic targets:

  • Prognostic Biomarker Validation:

    • Use IHC with ATP6V1G2 antibodies (dilution 1:50-1:500) to analyze expression patterns across glioma tissue microarrays

    • Correlate expression levels with patient survival data to validate the prognostic value of ATP6V1G2

    • Combine with other markers from the five-gene signature (ATP6V1C2, TCIRG1, ATP6AP1, and ATP6AP2) for comprehensive profiling

  • Tumor Microenvironment Analysis:

    • Implement multiplexed immunofluorescence with ATP6V1G2 antibodies to study its expression in relation to immune cell infiltration

    • This approach aligns with findings that the five-gene signature correlates with immune checkpoint expression and predicts response to immune checkpoint blockade (ICB) treatment

  • Mechanistic Studies:

    • Utilize ATP6V1G2 antibodies for Western blot analysis (dilution 1:500-1:5000) in glioma cell lines with various manipulations of the V-ATPase pathway

    • Investigate how ATP6V1G2 expression affects cellular processes relevant to tumor progression such as invasion, proliferation, and resistance to apoptosis

  • Therapeutic Target Validation:

    • Employ ATP6V1G2 antibodies to monitor protein expression changes in response to V-ATPase inhibitors

    • Develop screening assays using ATP6V1G2 antibodies to identify compounds that modulate its expression or function

  • Treatment Response Monitoring:

    • Assess changes in ATP6V1G2 expression in patient samples before and after treatment using IHC or Western blot

    • Correlate expression changes with treatment outcomes to identify predictive biomarkers

The risk score model incorporating ATP6V1G2 and other V-ATPase genes has shown correlation with treatment resistance and immune checkpoint blockade response , suggesting that antibody-based studies of these proteins could significantly contribute to developing more effective targeted therapies for glioma patients.

What are the methodological considerations for analyzing ATP6V1G2 in single-cell studies and spatial transcriptomics?

Single-cell analysis and spatial transcriptomics represent cutting-edge approaches that could provide unique insights into ATP6V1G2 function. When implementing these techniques, researchers should consider the following methodological aspects:

  • Single-Cell Protein Detection:

    • For single-cell immunofluorescence, ATP6V1G2 antibodies should be used at optimized dilutions (1:50-1:200)

    • Signal amplification methods may be necessary due to the relatively low abundance of ATP6V1G2 in individual cells

    • Validation using multiple antibodies targeting different epitopes is recommended to confirm specificity at the single-cell level

  • Integration with Single-Cell RNA-seq Data:

    • Research has shown that ATP6V1G2 might serve as a cell-type potential marker based on single-cell RNA-seq analysis

    • When correlating protein expression (detected by antibodies) with mRNA levels, researchers should be aware of potential discrepancies due to post-transcriptional regulation

    • Consider using split-pool approaches that allow simultaneous detection of proteins (using antibodies) and mRNA

  • Spatial Transcriptomics Applications:

    • For in situ detection, antibodies should be rigorously validated for specificity in the spatial context

    • When using ATP6V1G2 antibodies in multiplexed spatial proteomics, careful panel design is necessary to avoid spectral overlap with other markers

    • Controls for autofluorescence, particularly in brain tissue, are critical for accurate interpretation

  • Tissue Preparation and Fixation:

    • Optimization of fixation protocols to preserve both protein epitopes and RNA integrity is essential

    • For brain tissue, where ATP6V1G2 is highly expressed , special attention should be paid to fixation times and conditions

  • Data Analysis Considerations:

    • When analyzing spatial patterns of ATP6V1G2 expression, appropriate statistical methods for spatial data should be employed

    • Integration of protein expression data with transcriptomic and genomic data requires sophisticated computational approaches

Given that single-cell RNA-seq data has already implicated ATP6V1G2 as a potential cell-type marker , further studies combining antibody-based detection with single-cell and spatial approaches could provide valuable insights into its cell-type specific functions in normal and pathological contexts.

How can researchers combine ATP6V1G2 antibody-based methods with genomic approaches to gain comprehensive insights?

Integrating ATP6V1G2 antibody-based detection methods with genomic approaches can provide multi-dimensional insights into this protein's role in normal physiology and disease. Researchers should consider the following integrated approaches:

  • Correlation of Protein Expression with Genetic Alterations:

    • Use ATP6V1G2 antibodies for protein detection via Western blot (dilution 1:500-1:5000) or IHC (dilution 1:50-1:500) in samples with known genetic profiles

    • Research has shown that copy number variation may regulate ATP6V1G2 expression ; correlating antibody-detected protein levels with genomic data can validate these findings

    • Analyze how mutations or polymorphisms in ATP6V1G2 or related genes affect protein expression and localization

  • Epigenetic Regulation Studies:

    • Combine chromatin immunoprecipitation (ChIP) of transcription factors or histone modifications with ATP6V1G2 antibody-based protein detection

    • DNA promoter methylation has been implicated in regulating ATP6V1G2 expression ; researchers can correlate methylation profiles with protein levels detected by antibodies

  • Functional Genomics Integration:

    • Use ATP6V1G2 antibodies to assess protein changes following CRISPR/Cas9 genome editing of ATP6V1G2 or other V-ATPase components

    • Integrate RNA interference studies with antibody-based detection to validate knockdown efficiency at the protein level

    • Implement ATP6V1G2 antibodies in high-content screening assays to identify genomic regulators of ATP6V1G2 expression

  • Multi-omics Data Integration:

    • Correlate ATP6V1G2 protein levels (detected via antibodies) with transcriptomic, metabolomic, and proteomic datasets

    • This approach is particularly relevant in glioma research, where ATP6V1G2 is part of a prognostic gene signature

    • Develop computational methods to integrate antibody-based protein quantification with other -omics data layers

  • Long-term Expression Studies:

    • Use ATP6V1G2 antibodies to monitor protein expression changes over time in longitudinal studies

    • Correlate these changes with evolving genomic profiles, particularly in cancer progression models

By implementing these integrated approaches, researchers can develop a more comprehensive understanding of how genetic and epigenetic factors influence ATP6V1G2 expression and function, particularly in the context of diseases such as glioma where it has demonstrated prognostic significance .

What controls and validation experiments are essential when using ATP6V1G2 antibodies in critical research applications?

When using ATP6V1G2 antibodies for critical research applications, rigorous controls and validation experiments are essential to ensure reliable and reproducible results. Researchers should implement the following validation strategies:

  • Antibody Specificity Validation:

    • Genetic Controls: Test the antibody in ATP6V1G2 knockout or knockdown models to confirm specificity

    • Peptide Competition Assays: Pre-incubate the antibody with the immunizing peptide to verify that this blocks specific binding

    • Multiple Antibody Validation: Use antibodies from different sources or targeting different epitopes to confirm consistent patterns

    • Recombinant Protein Controls: Test reactivity against purified recombinant ATP6V1G2 protein

  • Technical Controls for Specific Applications:

    • Western Blot:

      • Positive Control: Include rat brain tissue lysate, which is known to express ATP6V1G2

      • Molecular Weight Verification: Confirm detection at the expected 14 kDa size

      • Loading Controls: Include appropriate housekeeping proteins

    • Immunohistochemistry:

      • Positive Control: Use mouse brain tissue, which shows positive IHC detection

      • Negative Control: Omit primary antibody while maintaining all other steps

      • Isotype Control: Use non-specific antibody of the same isotype and concentration

      • Antigen Retrieval Controls: Compare different retrieval methods (TE buffer pH 9.0 vs. citrate buffer pH 6.0)

    • Immunofluorescence:

      • Autofluorescence Control: Include unstained sample to assess background

      • Secondary Antibody Control: Omit primary antibody to assess non-specific secondary binding

      • Counterstaining: Use nuclear counterstains to aid in cellular localization

  • Cross-Validation with Orthogonal Methods:

    • mRNA Expression: Correlate protein detection with RT-PCR or RNA-seq data for ATP6V1G2

    • Mass Spectrometry: Confirm protein identity in immunoprecipitated samples

    • Functional Assays: Correlate antibody detection with functional assessments of V-ATPase activity

  • Reproducibility Assessment:

    • Batch Testing: Test multiple antibody lots to ensure consistent performance

    • Inter-laboratory Validation: When possible, verify findings across different research groups

    • Protocol Robustness: Assess sensitivity to variations in experimental conditions

  • Publication Standards:

    • Document all validation experiments in publications

    • Report antibody catalog numbers, lot numbers, and dilutions used

    • Provide images of all controls alongside experimental results

By implementing these comprehensive validation strategies, researchers can ensure that findings based on ATP6V1G2 antibody detection are reliable and contribute meaningfully to the scientific understanding of this protein's role in normal physiology and disease states.

How might ATP6V1G2 antibodies be used to investigate the role of V-ATPase in immune responses and cancer immunotherapy?

Recent research has revealed connections between the V-ATPase complex, which includes ATP6V1G2, and immune responses in cancer contexts. ATP6V1G2 antibodies could be instrumental in exploring these relationships in the following ways:

  • Investigating Immune Checkpoint Correlations:

    • Research has demonstrated a positive correlation between a risk score model (including ATP6V1G2) and immune checkpoint expression in gliomas

    • Researchers could employ ATP6V1G2 antibodies for IHC (dilution 1:50-1:500) or IF (dilution 1:50-1:200) to analyze co-expression patterns with immune checkpoint molecules like PD-1, PD-L1, and CTLA-4

    • Dual immunofluorescence staining could reveal spatial relationships between ATP6V1G2-expressing cells and immune cell populations

  • Characterizing Immune Cell Infiltration:

    • Studies have shown higher rates of inhibitory immune cell infiltration in tumors with high expression of V-ATPase genes

    • ATP6V1G2 antibodies could be used in multiplexed immunofluorescence panels to simultaneously detect ATP6V1G2 and immune cell markers

    • This approach could help delineate the spatial organization of immune cells relative to ATP6V1G2-expressing tumor cells

  • Assessing Immune Checkpoint Blockade Response:

    • Research has indicated that tumors with high ATP6V1G2 expression (as part of a five-gene signature) showed higher rates of "non-response" to immune checkpoint blockade treatment

    • ATP6V1G2 antibodies could be employed in longitudinal studies to monitor protein expression changes before and after immunotherapy

    • Changes in ATP6V1G2 localization or expression level might serve as biomarkers for treatment response

  • Exploring Mechanistic Connections:

    • V-ATPases regulate pH in endosomal compartments, which is critical for antigen processing and presentation

    • Researchers could use ATP6V1G2 antibodies to investigate whether alterations in this protein affect antigen-presenting cell function

    • Co-immunoprecipitation experiments with ATP6V1G2 antibodies might reveal novel protein interactions relevant to immune signaling pathways

  • Developing Combination Therapies:

    • Based on the observed relationship between ATP6V1G2 (as part of the V-ATPase complex) and immune checkpoint expression , researchers could investigate whether targeting V-ATPase activity might enhance immunotherapy efficacy

    • ATP6V1G2 antibodies would be essential tools for monitoring the effects of such combination approaches on protein expression and localization

By leveraging ATP6V1G2 antibodies in these research directions, investigators can gain deeper insights into how V-ATPase components influence the tumor immune microenvironment and potentially develop more effective immunotherapeutic strategies for cancers like glioma.

What methodological innovations might enhance the utility of ATP6V1G2 antibodies in future research?

Several methodological innovations could significantly enhance the utility of ATP6V1G2 antibodies in research settings:

  • Advanced Imaging Technologies:

    • Super-resolution Microscopy: Applying techniques like STORM or PALM with ATP6V1G2 antibodies could reveal subcellular localization at nanometer resolution

    • Expansion Microscopy: Physical expansion of samples could improve visualization of ATP6V1G2 in complex cellular structures

    • Live-cell Imaging: Development of non-disruptive labeling methods using ATP6V1G2 antibody fragments or nanobodies could enable dynamic studies of protein trafficking

  • Multiplexed Detection Systems:

    • Mass Cytometry (CyTOF): Adapting ATP6V1G2 antibodies for metal-tagged detection could allow simultaneous assessment of dozens of proteins

    • Cyclic Immunofluorescence: Sequential staining and imaging with ATP6V1G2 antibodies alongside numerous other markers could provide comprehensive spatial proteomic data

    • Spatial Transcriptomics Integration: Combining ATP6V1G2 antibody detection with in situ RNA analysis could correlate protein and transcript levels at subcellular resolution

  • Enhanced Antibody Engineering:

    • Recombinant Antibody Fragments: Developing smaller binding fragments (Fab, scFv) against ATP6V1G2 could improve tissue penetration and reduce background

    • Bi-specific Antibodies: Creating antibodies that simultaneously target ATP6V1G2 and another protein of interest could facilitate co-localization studies

    • pH-sensitive Antibody Conjugates: Given ATP6V1G2's role in pH regulation, developing antibody conjugates that report on local pH while binding to the target could provide functional information

  • Quantitative Approaches:

    • Automated Image Analysis: Developing machine learning algorithms specifically trained to analyze ATP6V1G2 immunostaining patterns could improve quantification accuracy and reproducibility

    • Single-molecule Detection: Techniques like single-molecule pull-down could enable precise quantification of ATP6V1G2 protein complexes

    • Standardized Quantification: Development of calibrated standards for absolute quantification of ATP6V1G2 in various applications

  • Functional Readouts:

    • Activity-based Probes: Developing probes that report on V-ATPase activity, used in conjunction with ATP6V1G2 antibodies, could correlate protein presence with function

    • Conformation-specific Antibodies: Engineering antibodies that recognize specific conformational states of ATP6V1G2 could provide insights into protein activation states

Implementation of these methodological innovations would significantly enhance our ability to study ATP6V1G2's role in normal physiology and disease contexts, particularly in complex tissues like the brain where this protein is highly expressed .

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.