Western Blot (WB): 1:5,000–1:50,000
Immunoprecipitation (IP): 0.5–4.0 µg per 1.0–3.0 mg lysate
Immunofluorescence (IF): 1:1,000–1:4,000
WB: 1:5,000–1:50,000
IHC: 1:500–1:2,000 (antigen retrieval recommended)
IF: 1:1,000–1:4,000
Optimal dilutions vary by sample type; validation data and protocols are available for download .
AP2B1 antibodies have been used to investigate:
AP2B1 facilitates clathrin-mediated endocytosis of African swine fever virus .
Pseudorabies virus exploits AP-2 to inhibit lysosomal TRPML1 inactivation during entry .
AP2B1 dysfunction is implicated in Alzheimer’s disease due to disrupted autophagy and lysosomal pathways .
AP-2 complexes interact with synaptic vesicle proteins, linking them to neurodegenerative disorders .
AP2A1 (a partner subunit) is a biomarker in ovarian cancer and pediatric medulloblastoma .
AP2B1 regulates β1 integrin transport, influencing cancer cell metastasis .
AP2B1 interacts with:
BUB1B (mitotic checkpoint protein)
TGF-β receptor 2 (signaling pathway regulator)
These interactions highlight its role in cell cycle regulation and receptor trafficking .
15690-1-AP has been cited in 12 WB, 3 IF, and 1 KO study, including research on CD1d-mediated viral entry .
68349-1-Ig shows cross-reactivity with pig and rabbit tissues, expanding its utility in comparative studies .
AP2B1 (adaptor related protein complex 2 subunit beta 1) is a component of the adaptor protein complex 2 (AP-2), which functions as a cytosolic heterotetramer that mediates sorting of membrane proteins in secretory and endocytic pathways. The protein plays a pivotal role in the formation of clathrin-coated vesicles (CCVs) by recruiting the scaffold protein clathrin. With a molecular weight of approximately 104.6-105 kDa, AP2B1 is crucial for understanding membrane trafficking, endocytosis, and related cellular processes . Research focused on AP2B1 provides insights into fundamental cell biology mechanisms that impact numerous physiological and pathological conditions.
AP2B1 antibodies are primarily used in Western Blotting (WB), Immunofluorescence (IF)/Immunocytochemistry (ICC), Immunoprecipitation (IP), and ELISA applications. Western Blotting is particularly common, with typical dilution ranges of 1:5000-1:50000 depending on the specific antibody and experimental system . Immunofluorescence applications typically use dilutions of 1:1000-1:4000, while immunoprecipitation protocols generally require 0.5-4.0 μg of antibody for 1.0-3.0 mg of total protein lysate . When designing experiments, it's essential to optimize antibody concentrations for your specific application and cell/tissue type to maximize signal-to-noise ratio.
Most commercially available AP2B1 antibodies show reactivity with human, mouse, and rat samples as confirmed through validation studies . Some antibodies have cited reactivity with additional species including pig and monkey models . When selecting an antibody for cross-species applications, it's advisable to check the manufacturer's validation data and consider sequence homology between your species of interest and the immunogen used to generate the antibody. For unusual model organisms, preliminary validation experiments are recommended before proceeding with full-scale studies.
For optimal Western Blot results with AP2B1 antibodies, follow these methodological guidelines:
Sample preparation: Lyse cells in RIPA buffer supplemented with protease inhibitors
Protein quantification: Use BCA or Bradford assay to normalize loading (20-50 μg total protein recommended)
Gel electrophoresis: Use 8-10% SDS-PAGE gels to properly resolve the 100-105 kDa AP2B1 protein
Transfer: Perform wet transfer to PVDF membrane (overnight at 30V, 4°C recommended for large proteins)
Blocking: Block with 5% non-fat milk in TBST for 1 hour at room temperature
Primary antibody: Dilute AP2B1 antibody 1:5000-1:50000 in blocking buffer and incubate overnight at 4°C
Secondary antibody: Use appropriate HRP-conjugated secondary antibody (typically 1:5000-1:10000)
Detection: Develop using ECL substrate and capture images using a digital imaging system
This protocol has been validated with multiple cell lines including HeLa, SH-SY5Y, K-562, C6, and NIH/3T3 cells .
AP2B1 antibodies are typically supplied in a storage buffer containing PBS with 0.02% sodium azide and 50% glycerol at pH 7.3 . For optimal stability and activity maintenance, store antibodies at -20°C, where they remain stable for approximately one year after shipment. Unlike some antibodies, small volume (20μl) AP2B1 antibody preparations often contain 0.1% BSA as a stabilizer . Aliquoting is generally unnecessary for -20°C storage of these antibody preparations, which helps minimize freeze-thaw cycles while maintaining convenience. Always briefly centrifuge antibody vials before opening to collect liquid at the bottom of the tube and avoid using antibodies past their expiration date.
When designing experiments with AP2B1 antibodies, include these essential controls:
Positive controls: Use validated cell lines known to express AP2B1 such as HeLa, SH-SY5Y, K-562, C6, or NIH/3T3 cells
Negative controls: Include samples where AP2B1 is knocked down using validated shRNAs
Loading controls: Use housekeeping proteins (β-actin, GAPDH) to normalize expression levels
Primary antibody omission: To assess non-specific binding of secondary antibody
Isotype control: Use matched concentration of non-specific rabbit IgG to evaluate non-specific binding
These controls help ensure experimental validity and facilitate accurate interpretation of results, particularly when studying novel tissue types or experimental conditions.
For effective AP2B1 knockdown studies, consider this methodological approach:
Design multiple shRNA sequences targeting different regions of AP2B1 mRNA (at least 3 different sequences recommended)
Clone shRNAs into appropriate vectors (pSuper or similar) with selection markers and/or fluorescent reporters
Transfect neurons or other target cells at appropriate developmental stages (e.g., DIV8 for hippocampal neurons)
Include scrambled shRNA sequences as negative controls
Validate knockdown efficiency by Western blot using AP2B1 antibodies (expect 70-90% reduction in protein levels)
Assess phenotypic changes using morphological analysis, trafficking assays, or other functional readouts
Perform rescue experiments with shRNA-resistant AP2B1 constructs to confirm specificity
This approach has been successfully used to study AP2B1's role in dendrite morphology and mTOR signaling , providing a robust framework for investigating AP2B1 function in various cellular contexts.
Distinguishing AP2B1 from other adaptin subunits presents several technical challenges that must be addressed through careful experimental design:
Sequence homology: Beta-adaptins share structural similarities across different AP complexes (AP-1, AP-2, etc.)
Co-immunoprecipitation concerns: AP-2 complex subunits typically co-precipitate, complicating individual analysis
Antibody specificity: Ensure antibodies target unique epitopes in AP2B1 not present in other beta-adaptin subunits
Expression pattern overlap: Multiple adaptin subunits may be expressed in the same tissues/cells
To overcome these challenges, researchers should:
Use antibodies validated for specificity against the unique regions of AP2B1
Employ mass spectrometry to distinguish between closely related adaptins in immunoprecipitation experiments
Consider using tagged AP2B1 constructs in overexpression studies
Implement subunit-specific knockdown to parse individual contributions to phenotypes
For high-quality co-localization studies using AP2B1 antibodies in immunofluorescence applications:
Cell preparation:
Culture cells on glass coverslips coated with appropriate substrates
Fix with 4% paraformaldehyde (10 minutes at room temperature)
Permeabilize with 0.1% Triton X-100 (5-10 minutes)
Blocking and antibody incubation:
Block with 3-5% BSA or normal serum (1 hour at room temperature)
Dilute AP2B1 primary antibody 1:1000-1:4000 in blocking solution
Incubate with second primary antibody (for co-localization target) simultaneously or sequentially
Use fluorophore-conjugated secondary antibodies with non-overlapping emission spectra
Imaging considerations:
Use confocal microscopy with appropriate filter sets
Capture z-stacks to ensure complete sampling of subcellular structures
Apply deconvolution algorithms to improve signal-to-noise ratio
Perform quantitative co-localization analysis using Pearson's or Mander's coefficients
Controls:
Single-antibody controls to assess bleed-through
Secondary-only controls to evaluate background
Known co-localizing and non-co-localizing proteins as references
This protocol has been validated with HeLa cells and can be adapted for other cell types with appropriate optimization .
When troubleshooting weak or absent AP2B1 signals in Western blots, consider these common issues and solutions:
Remember that AP2B1 should appear as a 100-105 kDa band in Western blots based on validated observations .
To distinguish between specific and non-specific binding in AP2B1 immunostaining experiments:
Pattern recognition: Specific AP2B1 staining typically shows punctate patterns near plasma membrane and cytoplasmic regions, consistent with its role in clathrin-coated vesicle formation. Non-specific staining often presents as diffuse signals or unusual subcellular localizations.
Controls validation:
Colocalization verification:
Co-stain with established markers of clathrin-coated structures
Verify expected co-localization with other AP-2 complex components
Assess changes in localization after perturbation of endocytic pathways
Staining optimization:
Titrate antibody concentrations to determine optimal signal-to-noise ratio
Modify fixation methods (PFA vs. methanol) to identify ideal epitope preservation
Adjust blocking conditions to reduce background
When comparing AP2B1 expression across different experimental models, researchers should consider these critical factors:
Expression baseline variations:
Different cell lines show varied endogenous AP2B1 expression levels
Tissue-specific expression patterns may influence interpretation
Developmental stage-dependent expression changes should be normalized appropriately
Technical normalization:
Use multiple housekeeping genes/proteins specific to each model system
Consider whole-protein staining methods (Ponceau S) for loading normalization
Apply appropriate statistical methods for cross-model comparisons
Antibody performance variables:
Validate antibody specificity in each model system separately
Adjust antibody dilutions based on expression level differences
Consider species-specific secondary antibodies for cross-species studies
Biological context interpretation:
AP2B1 functions within the AP-2 complex, requiring consideration of other subunit expressions
Changes in AP2B1 levels may represent compensatory mechanisms rather than direct experimental effects
Correlation with functional endocytosis assays provides context for expression data
AP2B1 antibodies can serve as powerful tools for investigating clathrin-mediated endocytosis (CME) dynamics through these methodological approaches:
Live-cell imaging:
Express fluorescently-tagged AP2B1 constructs in cells
Use antibodies to validate construct localization and function
Track vesicle formation, movement, and internalization in real-time
Proximity labeling techniques:
Couple AP2B1 antibodies with enzyme tags (HRP, APEX2, BioID)
Identify transient interaction partners during different stages of CME
Map the dynamic protein landscape of forming clathrin-coated pits
Super-resolution microscopy:
Use directly-conjugated or secondary fluorescent antibodies compatible with STORM/PALM
Achieve nanometer-scale resolution of AP2B1 localization within endocytic structures
Combine with other endocytic markers to create detailed structural maps
Functional blockade:
Microinject function-blocking AP2B1 antibodies to acutely disrupt function
Monitor effects on cargo internalization using fluorescent tracers
Compare with genetic approaches (knockdown/knockout) to distinguish acute vs. compensated effects
These approaches enable researchers to move beyond static snapshots to understand the dynamic processes of vesicle formation and trafficking.
When planning immunohistochemistry (IHC) studies with AP2B1 antibodies in tissue sections, researchers should consider:
Tissue preparation protocols:
Perfusion fixation generally yields better results than immersion fixation
Optimize fixative composition and duration for AP2B1 epitope preservation
Consider antigen retrieval methods (citrate buffer, pH 6.0 often effective)
Antibody selection criteria:
Choose antibodies specifically validated for IHC applications
Verify species reactivity matches your tissue source
Select antibodies targeting regions well-conserved across species for comparative studies
Signal amplification strategies:
Consider tyramide signal amplification for low-abundance detection
Polymer-based detection systems can improve sensitivity while reducing background
Biotin-streptavidin systems may require biotin blocking in certain tissues
Interpretation guidelines:
AP2B1 typically shows punctate cytoplasmic staining in positive cells
Expression patterns may vary by tissue type and cellular differentiation state
Compare with known expression databases to validate expected distribution
Controls:
Include positive control tissues with known AP2B1 expression
Use tissues from AP2B1 knockdown/knockout models as negative controls when available
Employ isotype controls at matched concentrations
Several AP2B1 antibodies have been specifically validated for IHC-p (paraffin) applications, making them suitable for archival tissue research .
AP2B1 antibodies provide valuable tools for investigating neurodegenerative disease mechanisms through these approaches:
Pathological sample analysis:
Compare AP2B1 expression and localization in post-mortem brain tissues from patients versus controls
Analyze co-localization with disease-specific aggregates (Aβ, tau, α-synuclein)
Assess endosomal-lysosomal pathway alterations using AP2B1 as a marker
Receptor trafficking studies:
Investigate AP2B1-dependent trafficking of disease-relevant receptors (glutamate receptors, APP)
Monitor internalization kinetics using surface biotinylation and AP2B1 co-immunoprecipitation
Examine effects of disease mutations on AP2B1-receptor interactions
Neuronal morphology research:
Therapeutic target validation:
Employ AP2B1 antibodies to confirm target engagement of compounds affecting CME
Monitor AP2B1 complex integrity after drug treatment
Validate downstream pathway modulation in response to interventions
These applications leverage AP2B1's role in critical neuronal processes including receptor trafficking and synaptic function, which are frequently disrupted in neurodegenerative conditions.