Applications : IF/IHC
Sample type: Mouse, human Mouse and human kidney tissue
Review: H+-ATPase G1 subunit is expressed along the mouse and human distal nephron.
ATP6V1G1 (also known as ATP6G, ATP6G1, ATP6J, V-type proton ATPase subunit G 1) is a subunit of the V1 complex of vacuolar H+ ATPase (V-ATPase), a multisubunit enzyme that consists of a peripheral V1 complex that hydrolyzes ATP and a membrane integral V0 complex that translocates protons . This protein plays a crucial role in acidifying and maintaining the pH of intracellular compartments including lysosomes and endosomes . In certain cell types, it is also targeted to the plasma membrane where it acidifies the extracellular environment . ATP6V1G1 is involved in several critical cellular processes including protein sorting, zymogen activation, receptor-mediated endocytosis, and synaptic vesicle proton gradient generation . Under aerobic conditions, it participates in intracellular iron homeostasis by triggering the activity of Fe(2+) prolyl hydroxylase enzymes, leading to HIF1A hydroxylation and subsequent proteasomal degradation .
ATP6V1G1 antibodies are utilized in multiple experimental applications to investigate the expression and function of this protein. Based on available research data, these antibodies have been validated for Western blotting (WB), immunocytochemistry/immunofluorescence (ICC/IF), immunohistochemistry (IHC), and enzyme-linked immunosorbent assay (ELISA) . For Western blotting, ATP6V1G1 antibodies are typically used at dilutions ranging from 1:1000 to 1:5000 . For immunohistochemistry applications, dilutions between 1:20 and 1:200 are recommended . For immunofluorescence procedures, the suggested dilution range is 1:50 to 1:200 . These applications enable researchers to detect and quantify ATP6V1G1 expression in various experimental contexts, from cell lines to tissue samples.
When using ATP6V1G1 antibodies, it is essential to include appropriate positive and negative controls to validate experimental findings. For positive controls, researchers should consider using cell lines known to express high levels of ATP6V1G1, such as LN229 and T98G glioblastoma cells . These cell lines have been documented to express elevated levels of ATP6V1G1 compared to less aggressive lines like SW1088 . Protein extracts from GBM neurospheres also show high expression of ATP6V1G1 and can serve as positive controls . For negative or low-expression controls, researchers may use extracts from grade I brain tumors, where V1G1 expression has been reported to be undetectable in protein extracts . Additionally, when performing knockdown or overexpression studies, appropriate vector controls should be included to account for non-specific effects. Using β-actin as a loading control for Western blotting is recommended to normalize protein expression across samples .
For optimal detection of ATP6V1G1 using antibodies, careful sample preparation is crucial. For protein extraction from adherent cells, RIPA lysate containing cocktail protease phosphatase inhibitor and PMSF pyrolysis should be used . Protein concentration should be determined using the BCA assay prior to SDS-PAGE separation . For Western blotting, proteins should be transferred to a PVDF membrane, which is then blocked in TBST solution containing 5% skim milk for 1 hour before incubation with the primary ATP6V1G1 antibody (typically at a concentration of 1:500) overnight at 4°C . For immunofluorescence analysis, PFA fixation followed by Triton X-100 permeabilization has been successfully used with ATP6V1G1 antibodies at a concentration of 4 μg/ml . When studying phosphorylated forms influenced by ATP6V1G1, special phosphoprotein enrichment techniques like the TiO2 method after trypsin digestion may be necessary .
ATP6V1G1 has been implicated in the progression of various cancers, including glioblastoma (GBM) and hepatocellular carcinoma (HCC) . To investigate its role in cancer progression, researchers can employ ATP6V1G1 antibodies in a multi-faceted approach. Immunohistochemical analysis of patient tumor samples at different stages can reveal correlations between ATP6V1G1 expression and disease progression, as demonstrated in HCC where ATP6V1G1 expression was shown to be elevated in advanced stages . For mechanistic studies, researchers should consider combining ATP6V1G1 antibody detection with functional assays after genetic manipulation (overexpression or knockdown) of ATP6V1G1 in cancer cell lines.
A comprehensive approach would include:
Western blotting to quantify ATP6V1G1 protein levels in different cancer cell lines and patient samples
Immunofluorescence to determine subcellular localization in cancer cells
Co-immunoprecipitation with ATP6V1G1 antibodies to identify interaction partners
Phosphoproteomic analysis to identify downstream signaling pathways affected by ATP6V1G1 expression
Studies have shown that ATP6V1G1 expression influences the phosphorylation status of several proteins involved in cancer-related pathways, including p-RPS6(Ser235) and p-SQSTM1(Ser272) (upregulated) and p-PDPK1(Ser241) and p-EEF2(Ser57) (downregulated) . These proteins are involved in critical processes such as the mTOR and PI3K/Akt signaling pathways, autophagy, and protein synthesis, all of which contribute to cancer cell growth and survival .
Establish stable cell lines with ATP6V1G1 overexpression or knockdown using lentiviral transfection
Extract total proteins using SDS lysate with protease and phosphatase inhibitors
Perform protein reduction, alkylation, and trypsin digestion
Label peptides with TMT6 labeling reagent for quantitative analysis
Enrich phosphorylated peptides using the TiO2 method
Conduct LC-MS/MS analysis to identify phosphorylated proteins
Validate key findings using western blotting with specific phospho-antibodies
For western blot validation, researchers should use antibodies specific to phosphorylated forms of proteins of interest, such as p-RPS6(Ser235), p-SQSTM1(Ser272), p-PDPK1(Ser241), and p-EEF2(Ser57) . It is essential to normalize results against total protein levels and use appropriate loading controls. RT-PCR should be employed to confirm successful genetic manipulation at the transcript level before proceeding with protein analysis .
ATP6V1G1 has been found to be highly expressed in cancer stem cell-enriched neurospheres isolated from glioblastoma patients, suggesting its potential role in cancer stemness . To investigate this aspect, researchers can employ ATP6V1G1 antibodies in several experimental approaches:
Compare ATP6V1G1 expression between cancer stem cell populations and differentiated tumor cells using western blotting and immunofluorescence
Analyze patient-derived neurospheres and their corresponding differentiated cultures for ATP6V1G1 expression
Perform functional assays after manipulating ATP6V1G1 expression in cancer stem cells to assess self-renewal, differentiation, and tumorigenicity
Studies have demonstrated that ATP6V1G1 gene and protein expression significantly decrease when GBM neurospheres are differentiated into adherent cell monolayers, indicating its potential role in maintaining stemness . For therapy resistance studies, researchers can investigate the correlation between ATP6V1G1 expression and response to standard treatments. The phosphorylation of SQSTM1, which is regulated by ATP6V1G1, has been shown to be closely associated with cisplatin resistance in tumor cells , suggesting a potential mechanism through which ATP6V1G1 might influence therapy resistance.
When incorporating ATP6V1G1 antibodies into multiplexed immunoassays, researchers may encounter several technical challenges that require careful consideration:
Cross-reactivity: ATP6V1G1 has several related isoforms (ATP6V1G2, ATP6V1G3) and researchers must ensure the antibody specifically detects ATP6V1G1 without cross-reacting with these paralogues .
Antibody compatibility: In multiplexed fluorescence immunoassays, antibodies must be compatible in terms of species origin to avoid cross-reactivity between secondary antibodies. ATP6V1G1 antibodies are commonly available in rabbit polyclonal format , which may limit combinations with other rabbit-derived antibodies.
Signal interference: When studying phosphorylated proteins regulated by ATP6V1G1, phospho-specific antibodies may have different optimal conditions for antigen retrieval and detection compared to ATP6V1G1 antibodies.
Epitope accessibility: As ATP6V1G1 functions as part of a multisubunit complex, epitope masking may occur depending on complex formation, potentially affecting antibody binding.
To overcome these challenges, researchers should:
Perform careful antibody validation using positive and negative controls
Use antibodies raised in different host species when designing multiplexed assays
Consider sequential rather than simultaneous staining for challenging combinations
Employ spectral unmixing techniques to resolve overlapping fluorescence signals
Validate findings with alternative methods such as mass spectrometry-based approaches
ATP6V1G1, as a component of V-ATPase, plays a critical role in maintaining the acidic environment of lysosomes necessary for proper degradation of cellular components through autophagy . To investigate this function, researchers can employ ATP6V1G1 antibodies in conjunction with autophagy markers and functional assays:
Co-localization studies: Use immunofluorescence with ATP6V1G1 antibodies alongside lysosomal markers (LAMP1, LAMP2) and autophagy markers (LC3, SQSTM1/p62) to assess the association of ATP6V1G1 with these structures.
Autophagic flux assays: Manipulate ATP6V1G1 expression and monitor changes in autophagy markers by western blotting. The relationship between ATP6V1G1 and SQSTM1 phosphorylation is particularly relevant, as SQSTM1 is a key protein in autophagy formation and its phosphorylation status affects autophagy activity .
Lysosomal function assessment: Use lysosomal pH-sensitive dyes to determine if ATP6V1G1 manipulation affects lysosomal acidification. Previous research has shown that altering ATP6V1G1 expression (either overexpression or silencing) slowed acid kinetics in HeLa cells, suggesting impaired lysosomal function .
Proteolytic activity assays: Measure the activity of lysosomal enzymes that depend on acidic pH after ATP6V1G1 manipulation to assess functional consequences on lysosomal degradation.
Research has shown that ATP6V1G1 regulates liver lipid metabolism by maintaining normal acidification function of lysosomes, suggesting its potential role in non-alcoholic fatty liver disease . This connection between ATP6V1G1, lysosomal function, and lipid metabolism represents an important area for further investigation.
Studying the interactions between ATP6V1G1 and other V-ATPase subunits is crucial for understanding the assembly and function of the V-ATPase complex. Several antibody-based approaches can be employed:
Co-immunoprecipitation (Co-IP): Use ATP6V1G1 antibodies to pull down the protein and its interacting partners, followed by western blotting with antibodies against other V-ATPase subunits. This approach can identify direct or indirect interactions within the complex.
Proximity ligation assay (PLA): This technique can detect protein-protein interactions in situ by generating a fluorescent signal when two proteins are in close proximity. It requires antibodies against ATP6V1G1 and potential interacting partners raised in different species.
Cross-linking immunoprecipitation: Use chemical cross-linkers to stabilize protein-protein interactions before immunoprecipitation with ATP6V1G1 antibodies, followed by mass spectrometry to identify interacting proteins.
Immunofluorescence co-localization: Perform multi-color immunofluorescence using ATP6V1G1 antibodies alongside antibodies against other V-ATPase subunits to assess their spatial co-localization in cells.
ATP6V1G1 is known to be part of the peripheral V1 complex that hydrolyzes ATP , and understanding its interactions with other subunits can provide insights into how V-ATPase assembly and activity are regulated in different cellular contexts, particularly in pathological conditions where ATP6V1G1 expression is altered.
Proper validation of ATP6V1G1 genetic manipulation models is essential for ensuring experimental reliability when studying its functions. ATP6V1G1 antibodies play a crucial role in this validation process:
Western blotting: The primary method to confirm successful manipulation of ATP6V1G1 at the protein level. For knockdown validation, researchers should observe a significant reduction in ATP6V1G1 protein levels compared to control samples. For overexpression studies, increased protein levels should be detected. Quantification of band intensities normalized to loading controls (e.g., β-actin) is essential .
Immunofluorescence: Provides visual confirmation of ATP6V1G1 knockdown or overexpression at the cellular level, allowing assessment of expression levels in individual cells and subcellular localization patterns .
Functional validation: Beyond confirming expression changes, researchers should validate functional consequences of ATP6V1G1 manipulation:
Measure V-ATPase activity using acidification assays
Assess the phosphorylation status of proteins known to be regulated by ATP6V1G1, such as RPS6, SQSTM1, PDPK1, and EEF2
Evaluate cellular phenotypes associated with ATP6V1G1 expression changes, such as proliferation, migration, and resistance to apoptosis
Controls: Proper controls are essential:
Research has shown successful construction of HepG2 and Huh7 HCC cell lines with stable overexpression of ATP6V1G1 through lentiviral transfection, validated by both RT-PCR and western blot .
Tissue microarray analysis: Perform immunohistochemistry using ATP6V1G1 antibodies on patient tumor samples of different grades to establish correlation between expression levels and tumor grade. Studies have shown that ATP6V1G1 expression is higher in high-grade human glioma tissues compared to grade II tumors .
Cancer stem cell investigation: Analyze ATP6V1G1 expression in GBM neurospheres (cancer stem cell-enriched) compared to their differentiated counterparts using both western blotting and immunofluorescence. Research has demonstrated elevated gene and protein expression of ATP6V1G1 in neurospheres, which significantly decreases upon differentiation .
Functional studies: After manipulating ATP6V1G1 expression in GBM cell lines, assess changes in:
Proliferation and colony formation
Migration and invasion
Apoptosis resistance
Response to standard GBM treatments
Microenvironment interactions: Investigate how ATP6V1G1 expression influences the tumor microenvironment, as research suggests it may interact with other factors to reprogram the surrounding non-tumor microenvironment to a pro-tumor state .
In vivo models: Validate findings using orthotopic GBM models with ATP6V1G1 knockdown or overexpression, followed by immunohistochemical analysis of tumor sections.
Hepatocellular carcinoma (HCC) represents another cancer type where ATP6V1G1 plays a significant role in disease progression . Researchers can employ ATP6V1G1 antibodies in various methodological approaches to investigate this involvement:
Expression analysis in patient samples: Perform immunohistochemistry on HCC tissue microarrays to correlate ATP6V1G1 expression with clinicopathological features and patient outcomes. Public data has revealed that ATP6V1G1 expression is dramatically increased in HCC compared to normal tissues and is elevated in advanced stages of HCC .
Phosphoproteomic profiling: After manipulating ATP6V1G1 expression in HCC cell lines, use phospho-specific antibodies to validate changes in phosphorylation levels of key proteins:
Pathway analysis: Investigate the impact of ATP6V1G1 on critical signaling pathways in HCC:
Functional validation: Assess the effects of ATP6V1G1 manipulation on:
HCC cell proliferation and colony formation
Migration and invasion capabilities
Resistance to apoptosis
Response to standard HCC treatments
Mechanistic studies: Use co-immunoprecipitation with ATP6V1G1 antibodies to identify protein interaction partners that may mediate its effects on phosphorylation-related pathways in HCC.
ATP6V1G1 has been implicated in several neurological disorders, including amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) . ATP6V1G1 antibodies can be valuable tools for investigating these connections:
Expression analysis in patient samples: Perform immunohistochemistry and western blotting on post-mortem brain tissue from patients with ALS, FTD, and other neurodegenerative disorders to assess ATP6V1G1 expression levels compared to controls.
Mutation studies: Investigate how UBQLN2 mutations, which have been found to cause ATP6V1G1 overexpression leading to ALS and FTD , affect ATP6V1G1 protein levels and localization using specific antibodies.
Cell models: In neuronal cell cultures with manipulated ATP6V1G1 expression:
Assess lysosomal function and autophagy using co-immunofluorescence with lysosomal markers
Evaluate effects on proteostasis and protein aggregation
Investigate mitochondrial function and oxidative stress
Animal models: In transgenic mouse models of neurodegenerative diseases:
Perform immunohistochemistry to analyze ATP6V1G1 expression patterns in different brain regions
Assess correlation between ATP6V1G1 levels and disease progression
Investigate rescue effects of normalizing ATP6V1G1 expression
Development studies: Analyze ATP6V1G1 expression in embryonic neural tissues, as research has shown it is abundant in rat embryonic hippocampal neuronal cells (E18 RHN) , suggesting a role in neural development.
The detection of phosphorylated proteins regulated by ATP6V1G1 requires careful methodological considerations to ensure reliable and reproducible results. Based on available research, the following protocol has proven effective:
Sample preparation:
Extract total proteins from cells using RIPA lysate containing cocktail protease phosphatase inhibitor and PMSF
Determine protein concentration using the BCA assay
For phosphoproteomic analysis, take 200μg protein for reduction and alkylation, followed by trypsin digestion overnight at 37°C
Phosphopeptide enrichment:
Western blot validation:
Separate proteins by SDS-PAGE and transfer to PVDF membrane
Block membranes in TBST with 5% skim milk for 1 hour
Incubate with specific primary antibodies overnight at 4°C:
anti-p-RPS6 (Ser235) at 1:1000 dilution
anti-p-SQSTM1 (Ser272) at 1:1000 dilution
anti-p-EEF2 (Ser57) at 1:1000 dilution
anti-p-PDPK1 (Ser241) at 1:1000 dilution
anti-β-actin at 1:1000 dilution (loading control)
Incubate with secondary antibodies for 1 hour at room temperature
Quantification and analysis:
Perform densitometric analysis of western blot bands
Normalize phosphoprotein levels to total protein levels when possible
Compare phosphorylation profiles between ATP6V1G1 overexpression/knockdown and control groups
This protocol has successfully identified 163 differentially expressed phosphorylated proteins with 228 altered phosphorylation sites in ATP6V1G1 overexpression studies .
Optimizing immunofluorescence protocols for ATP6V1G1 detection requires consideration of cell type-specific factors and experimental goals. Based on published methodologies, the following optimization strategies are recommended:
Fixation and permeabilization:
For most cell types, PFA fixation (4%) followed by Triton X-100 permeabilization has proven effective for ATP6V1G1 detection
For neurons or delicate primary cells, milder permeabilization with 0.1% Triton X-100 or 0.1% saponin may preserve cellular structures better
For membrane localization studies, avoid methanol fixation which can disrupt membrane structures
Antibody concentration optimization:
Cell type-specific considerations:
For cancer stem cells (neurospheres): optimize for suspension culture using low-speed centrifugation on poly-L-lysine coated slides
For highly confluent cultures: ensure adequate permeabilization time to allow antibody penetration
For co-staining with V-ATPase complex markers: carefully select compatible antibodies raised in different host species
Signal amplification strategies:
For low expression contexts: consider tyramide signal amplification
For high background: increase blocking time or use alternative blocking reagents (5% BSA, normal serum)
For specific subcellular localization: combine with organelle markers (lysosomes, endosomes) for co-localization studies
Validation controls:
Designing effective ATP6V1G1 knockdown experiments for functional studies requires careful planning to ensure specificity, efficiency, and proper phenotypic analysis. Based on research approaches, consider the following:
Knockdown method selection:
siRNA transfection: suitable for short-term studies (48-72 hours)
shRNA via lentiviral transduction: preferable for stable, long-term knockdown studies
CRISPR-Cas9: for complete knockout studies where viable
Target sequence design:
Design multiple target sequences to control for off-target effects
Ensure specificity for ATP6V1G1 by avoiding sequences with homology to ATP6V1G2 or ATP6V1G3
Target conserved regions if working with non-human models
Validation of knockdown efficiency:
Validate at mRNA level using RT-PCR
Confirm protein reduction using western blotting with ATP6V1G1 antibody
Quantify knockdown efficiency through densitometric analysis
Functional assays following knockdown:
V-ATPase activity: measure acidification using pH-sensitive dyes
Cell phenotype: assess proliferation, migration, invasion, and apoptosis
Pathway analysis: examine effects on phosphorylation of key proteins (RPS6, SQSTM1, PDPK1, EEF2)
Lysosomal function: evaluate autophagy flux and degradative capacity
Control considerations:
Include non-targeting siRNA/shRNA controls
Consider rescue experiments by reintroducing ATP6V1G1 to confirm specificity
Include positive controls such as knockdown of known V-ATPase subunits
Cell type-specific considerations:
It's important to note that complete ATP6V1G1 knockdown may have profound effects on cell viability due to its crucial role in cellular pH homeostasis, so titrating knockdown levels or using inducible systems may be necessary for some experimental contexts.