The ATP6V1D antibody targets the D subunit of the V1 domain of V-ATPase, a 28 kDa protein encoded by the ATP6V1D gene on human chromosome 14 . It is commonly used in immunoassays to study V-ATPase activity in diverse cell types, including neurons, kidney cells, and cancer cells.
Host/Isotype: Rabbit (polyclonal or recombinant monoclonal).
Reactivity: Cross-reacts with human, mouse, and rat samples.
Applications: Western blot (WB), immunoprecipitation (IP), immunohistochemistry (IHC), immunofluorescence (IF), and flow cytometry (FC).
The ATP6V1D antibody is employed in studies investigating cellular acidification, lysosomal function, and disease mechanisms:
a. Neurodevelopment and Behavior
A study (Ncoa7 locus) used this antibody to demonstrate that V-ATPase dysfunction disrupts neurodevelopment, leading to behavioral abnormalities .
b. Diabetic Nephropathy
In a kidney disease model, the antibody revealed enhanced V-ATPase activity in podocytes, linking lysosomal acidification to autophagy regulation .
c. Glioblastoma
Research showed that inhibiting autophagy via NEO214 induces glioblastoma cell death, with ATP6V1D serving as a marker for lysosomal disruption .
d. Endolysosomal System
Proteomic analysis highlighted ATP6V1D as a key component in cross-linked endolysosomal structures, suggesting roles in cargo trafficking .
Protocols for common applications include :
Western Blot: 1:10,000 dilution with heat-mediated antigen retrieval.
IHC: 1:50 dilution on paraffin-embedded tissues (e.g., human breast, kidney).
IP: 1:10 dilution for pulldown assays.
Flow Cytometry: 1:10 dilution for intracellular staining.
The antibody has been cited in six studies, including analyses of lysosomal proteomes , synaptic vesicle acidification , and ciliary biogenesis .
ATP6V1D (also known as ATP6M, VATD, or V-ATPase subunit D) is a 28-35 kDa subunit of the V-type proton ATPase complex. This complex plays essential roles in:
Lysosomal acidification through proton translocation
Endosomal trafficking and protein sorting
Macroautophagic/autophagic flux maintenance
Cellular homeostasis regulation
In particular, research has shown that ATP6V1D enhances hepatocellular carcinoma stemness and progression by maintaining autophagic flux through two mechanisms: promoting lysosomal acidification and enhancing the interaction between CHMP4B and IST1 to foster ESCRT-III complex assembly, thereby facilitating autophagosome-lysosome fusion .
ATP6V1D antibodies have been validated for multiple research applications with specific recommended protocols:
| Application | Validated Dilutions | Sample Types | Notes |
|---|---|---|---|
| Western Blot (WB) | 1:500-1:5000 | Human brain, mouse lung and skeletal muscle | 28-35 kDa band expected |
| Immunohistochemistry (IHC) | 1:50-1:200 | Human lung cancer tissue | Paraffin-embedded sections |
| Immunoprecipitation (IP) | 1:200-1:2000 | Mouse lung tissue | 4μg antibody for 4000μg lysate |
| Flow Cytometry (FC) | 0.2μg per 10⁶ cells | HeLa cells | Fixed with 90% MeOH, blocked with 3% BSA |
| ELISA | 1:500-1:2000 | Various | For quantitative detection |
The cross-species reactivity has been confirmed for human, mouse, and rat samples .
For rigorous experimental design with ATP6V1D antibodies:
Positive tissue controls: Human brain tissue and lung cancer samples have shown consistent ATP6V1D expression
Negative controls: Include isotype control (rabbit IgG) at equivalent concentrations
Knockdown validation: Compare expression in wild-type vs ATP6V1D-silenced samples
Loading controls: For Western blots, GAPDH has been validated as an appropriate normalization control
Blocking peptide controls: Pre-incubation with immunizing peptide (ATP6V1D recombinant protein) to confirm specificity
Cross-validation: Compare results using different antibody clones targeting distinct epitopes of ATP6V1D
Cross-validation is particularly important as ATP6V1D is part of a complex with multiple similar subunits .
Optimizing Western blot protocols for ATP6V1D detection:
Sample preparation: Use lysis buffers containing protease inhibitors to prevent degradation
Protein denaturation: Heat samples at 95°C for 5 minutes in sample buffer containing 5% β-mercaptoethanol
Gel selection: 10-12% polyacrylamide gels provide optimal resolution for 28-35 kDa proteins
Transfer conditions: Semi-dry transfer at 15V for 30 minutes or wet transfer at 100V for 1 hour
Membrane blocking: Block with 5% non-fat milk or 3% BSA in TBST for 1 hour at room temperature
Antibody incubation: Primary antibody (1:1000) overnight at 4°C, followed by appropriate HRP-conjugated secondary antibody
Signal detection: Enhanced chemiluminescence with exposure times adjusted based on expression levels
Recent studies achieved clear detection in human brain tissue at 1:500 dilution with minimal background .
For high-resolution ATP6V1D localization:
Fixation: 4% paraformaldehyde for 15-30 minutes (frozen sections) or formalin-fixed paraffin embedding
Antigen retrieval: Heat-induced epitope retrieval in citrate buffer (pH 6.0) for 15-20 minutes
Permeabilization: 0.5% Triton X-100 for 10 minutes at room temperature
Blocking: 3% BSA for 30 minutes to reduce non-specific binding
Primary antibody: Incubate with ATP6V1D antibody (1:100) overnight at 4°C
Secondary detection: Appropriate species-specific secondary antibody (1:1000)
Co-staining: Combine with lysosomal markers (LAMP1) to assess subcellular localization
Mounting: Use anti-fade mounting medium containing DAPI for nuclear visualization
This approach has successfully revealed ATP6V1D distribution in human lung cancer tissue with high specificity .
For meaningful co-localization studies:
Compatible antibody pairs: Select ATP6V1D antibody and co-staining markers from different host species
Sequential staining: For antibodies from the same species, use sequential immunostaining with blocking steps
Fluorophore selection: Choose fluorophores with minimal spectral overlap
Confocal microscopy settings: Optimize laser power, detector gain, and pinhole settings for each channel
Quantification methods: Use Pearson's correlation coefficient or Manders' overlap coefficient for quantitative assessment
Validation controls: Include single-stained samples to rule out bleed-through
Biological controls: Compare ATP6V1D localization under different physiological or pathological conditions
Recent studies demonstrated successful co-localization of ATP6V1D with lysosomal markers using rabbit polyclonal anti-ATP6V1B2 antibody and mouse monoclonal anti-Lamp1 antibody .
To dissect ATP6V1D's function in autophagy:
Genetic manipulation approaches:
siRNA/shRNA-mediated knockdown (transient or stable)
CRISPR-Cas9 knockout or knockin of mutant variants
Overexpression of wild-type or mutant ATP6V1D
Autophagy flux assessment:
Monitor LC3-II/LC3-I conversion by Western blot
Track p62/SQSTM1 accumulation or degradation
Use tandem fluorescent-tagged LC3 (mRFP-GFP-LC3) to distinguish autophagosome formation from fusion
Measure long-lived protein degradation rates
Lysosomal function evaluation:
LysoTracker staining to assess lysosomal acidification
Cathepsin activity assays to measure lysosomal enzyme function
Lysosomal membrane permeabilization assessment
Autophagosome-lysosome fusion analysis:
Co-localization of LC3 and LAMP1
Electron microscopy for ultrastructural analysis
Live-cell imaging of fusion events
Pharmacological interventions:
Bafilomycin A1 for V-ATPase inhibition (positive control)
Chloroquine or NH₄Cl to neutralize lysosomal pH
3-methyladenine (3-MA) to inhibit autophagosome formation
Recent studies have shown that ATP6V1D not only promotes lysosomal acidification but also enhances the interaction between CHMP4B and IST1 to foster ESCRT-III complex assembly, facilitating autophagosome-lysosome fusion .
For investigating ATP6V1D in cancer stem cell biology:
Expression correlation analysis:
Assess correlation between ATP6V1D and established stemness markers (PROM1, CD24, EPCAM, CD13, CD44, THY1)
Analyze patient-derived samples with varying degrees of stemness
Functional stemness assays:
Sphere formation assays following ATP6V1D modulation
Colony formation efficiency
Limiting dilution tumor initiation assays in vivo
Serial transplantation experiments
Signaling pathway investigation:
Examine effects on Wnt/β-catenin, Notch, and Hedgehog pathways
Analyze transcription factors regulating stemness (OCT4, SOX2, NANOG)
Therapeutic implications:
Test low-dose bafilomycin A1 effects on cancer stemness
Evaluate combination approaches with conventional chemotherapeutics
Pearson correlation analysis has shown that ATP6V1D expression positively correlates with cancer stem cell markers including PROM1, CD24, EPCAM, CD13, CD44, and THY1, suggesting its potential role in maintaining cancer stem cell properties .
To characterize ATP6V1D's interactome:
Co-immunoprecipitation (co-IP):
Use anti-Flag antibodies for tagged ATP6V1D constructs
Apply stringent washing conditions to reduce false positives
Verify interactions by reciprocal co-IP
Proximity-dependent labeling:
BioID or TurboID fusion proteins to identify proximal proteins
APEX2-based proximity labeling for temporal dynamics
FRET/FLIM analysis:
Fluorescently tag ATP6V1D and potential interaction partners
Measure energy transfer as indicator of protein proximity
Mass spectrometry-based approaches:
Label-free quantitative proteomics
SILAC labeling for quantitative comparison
Crosslinking mass spectrometry for structural information
Structural biology methods:
X-ray crystallography of ATP6V1D complexes
Cryo-EM analysis of V-ATPase assemblies
Hydrogen-deuterium exchange mass spectrometry
Recent research demonstrated successful co-IP of ATP6V1D with CHMP4B and IST1, revealing its role in enhancing ESCRT-III complex assembly for autophagosome-lysosome fusion .
| Challenge | Possible Causes | Recommended Solutions |
|---|---|---|
| Weak Western blot signal | Insufficient protein, suboptimal antibody concentration | Increase protein loading (30-50μg), optimize antibody concentration (1:500), extend ECL exposure |
| Multiple bands | Protein degradation, isoform detection, non-specific binding | Use fresh samples with protease inhibitors, validate with knockout controls, optimize blocking and washing |
| High background in IHC | Insufficient blocking, excessive antibody concentration | Increase blocking time (2h), dilute antibody further (1:200), add 0.1% Tween-20 to wash buffers |
| Poor reproducibility | Lot-to-lot antibody variation, inconsistent protocols | Standardize all protocol steps, maintain consistent antibody lots for entire study |
| Failed co-IP | Weak/transient interactions, harsh lysis conditions | Use gentler lysis buffers, crosslinking prior to lysis, optimize antibody-to-lysate ratio (4μg antibody per 4000μg lysate) |
| Inconsistent flow cytometry results | Variable fixation conditions, insufficient permeabilization | Standardize fixation (90% MeOH), ensure adequate permeabilization, block with 3% BSA for 30 minutes |
These troubleshooting approaches are based on validated protocols established with commercial ATP6V1D antibodies .
When encountering contradictory results:
Biological context consideration:
Cell/tissue-specific functions may vary dramatically
Disease stage may influence ATP6V1D behavior (particularly in cancer progression)
Acute vs. chronic modulation may have opposite effects
Technical assessment:
Antibody specificity confirmation through multiple methods
Knockdown/knockout efficiency verification
Cross-validation with multiple experimental approaches
Functional redundancy analysis:
Assess compensatory upregulation of other V-ATPase subunits
Evaluate potential adaptive mechanisms following ATP6V1D modulation
Data integration strategies:
Weigh evidence based on methodological rigor
Consider time-dependent effects in experimental design
Integrate findings across multiple model systems
Mechanistic reconciliation:
ATP6V1D may have V-ATPase-dependent and independent functions
Different protein interactions may predominate in different contexts
Post-translational modifications may alter protein function
Research has shown that ATP6V1D has dual mechanisms - promoting lysosomal acidification and enhancing autophagosome-lysosome fusion through ESCRT-III complex assembly - which may explain seemingly conflicting observations in different experimental systems .
Emerging therapeutic strategies include:
V-ATPase inhibition:
Low-dose bafilomycin A1 showed promise for HCC treatment
Concanamycin A derivatives with improved pharmacokinetics
Subunit-specific inhibitors to reduce systemic toxicity
Genetic modulation approaches:
siRNA/shRNA targeting ATP6V1D for transient knockdown
Antisense oligonucleotides for selective inhibition
PROTAC-based degradation of ATP6V1D protein
Combination therapies:
V-ATPase inhibitors with autophagy modulators
Synergistic effects with conventional chemotherapeutics
Targeting both ATP6V1D and its interaction partners (CHMP4B/IST1)
Biomarker development:
ATP6V1D expression as prognostic indicator in HCC
Correlation with treatment response prediction
Patient stratification for personalized therapy
Recent research demonstrated that low-dose bafilomycin A1 targeting the V-ATPase complex shows promise as a potential therapeutic strategy for hepatocellular carcinoma, highlighting ATP6V1D as an emerging therapeutic target .
ATP6V1D antibodies enable investigation of:
Neurodevelopmental disorders:
ATP6V1 family proteins (ATP6V1C1/ATP6V1B2) are implicated in neurodevelopmental phenotypes resembling DOORS syndrome
ATP6V1D may have parallel functions in neuronal development and function
Lysosomal storage diseases:
Detection of altered V-ATPase subunit expression or localization
Assessment of compensatory mechanisms in primary lysosomal disorders
Evaluation of lysosomal morphology and distribution changes
Autophagy-related pathologies:
Investigation of defective autophagic flux in neurodegenerative diseases
Analysis of ATP6V1D expression in models of impaired autophagy
Therapeutic modulation of V-ATPase activity to restore autophagic function
Cilium biogenesis and ciliopathies:
Assessment of ATP6V1D's role in cilium formation
Investigation of overlap between V-ATPase function and ciliopathies
Recent studies have shown that mutations in ATP6V1 family proteins affect lysosomal morphology, localization, and function, resulting in defective autophagic flux and accumulation of lysosomal substrates. This work also showed that upregulated V-ATPase function affects cilium biogenesis, documenting pleiotropy .
Cutting-edge methodologies for ATP6V1D investigation:
Single-cell analysis:
Single-cell RNA-seq to map ATP6V1D expression heterogeneity
Single-cell proteomics for protein-level analysis
Spatial transcriptomics to assess tissue-specific expression patterns
Advanced imaging technologies:
Super-resolution microscopy for nanoscale localization
Live-cell pH sensors to monitor V-ATPase activity in real-time
Correlative light and electron microscopy for structure-function studies
Genome editing innovations:
Base editing for introducing specific ATP6V1D mutations
Prime editing for precise genetic modifications
Inducible/conditional knockout systems for temporal control
Computational approaches:
AI-driven protein structure prediction for ATP6V1D complexes
Systems biology modeling of V-ATPase in cellular networks
Multi-omics data integration for comprehensive pathway analysis
Organoid and tissue-specific models:
Patient-derived organoids to study ATP6V1D in disease contexts
Tissue-specific conditional knockout models
Humanized mouse models for translational research
These advanced approaches will enable researchers to better understand ATP6V1D's complex roles in cellular physiology and pathology, potentially leading to novel therapeutic interventions for diseases involving V-ATPase dysfunction.