AIP1 inhibits VEGF-induced angiogenesis by binding to the VEGFR2-PI3K complex. In HUVECs, AIP1 knockdown enhances EC migration via upregulated VEGFR2 signaling, while its overexpression suppresses it .
Co-immunoprecipitation assays show AIP1 associates with phosphorylated VEGFR2 at late phases (15–30 min post-VEGF stimulation) .
AIP1-KO mice exhibit accelerated tumor metastasis and pathological angiogenesis .
AIP1 acts as a tumor suppressor by inhibiting Ras, PI3K/Akt, and NF-κB pathways. Epigenetic silencing of AIP1 via EZH2 promotes metastasis in prostate and breast cancers .
Subcutaneous B16 melanoma models show AIP1-KO increases lung metastasis incidence by 80% (vs. 0% in WT) .
AIP1 re-expression reduces tumor nodule count and area in metastatic sites .
AIP1 isoforms differentially regulate inflammation:
AIP1A: Suppresses TNFα-induced ROS and VCAM-1/ICAM-1 expression .
AIP1B: Enhances mitochondrial ROS (mitoROS) via ASK1, exacerbating EC activation .
TNFα downregulates AIP1A via Smurf1-mediated degradation while upregulating AIP1B through RIF1/H3K9me3 modulation .
AIP1-2 antibodies are critical for:
AIP1-2 (ASK1-Interacting Protein 1-2) is a signaling scaffold protein that interacts with actin and modulates several pathways associated with inflammation, apoptosis, and cellular development. In plants such as Arabidopsis, AIP1-2 specifically binds to actin isoforms including ACT2 and ACT7, regulating cytoskeletal organization . In mammalian systems, AIP1 functions in TNF-α-induced ASK1 activation by facilitating dissociation of inhibitory proteins like 14-3-3, thereby initiating apoptotic signaling pathways . Additionally, AIP1 modulates several MAPK signaling pathways including JNK, p38 MAPK, and ERK1/2, which are critical for inflammatory responses and insulin sensitivity .
For optimal Western blot detection using AIP1-2 antibodies:
Sample preparation: Extract proteins using RIPA buffer with protease and phosphatase inhibitors to preserve protein integrity .
Protein loading: Load 20-30 μg of total protein per lane for cell lysates; higher amounts (50-75 μg) may be needed for tissue extracts .
Gel conditions: Use 7.5% SDS-PAGE for optimal separation, as AIP1 has a molecular weight around 130 kDa .
Transfer conditions: Transfer to PVDF membrane at 100V for 90 minutes in standard transfer buffer with 20% methanol.
Blocking: Block with 5% non-fat dry milk or BSA in TBST for 1 hour at room temperature.
Primary antibody: Dilute anti-AIP1-2 antibody typically 1:500-1:1000 in blocking buffer; incubate overnight at 4°C.
Secondary antibody: Use appropriate HRP-conjugated secondary antibody (1:5000) for 1 hour at room temperature.
Detection: Visualize using ECL substrates with exposure times of 1-5 minutes depending on signal strength .
Expected band size for AIP1 is approximately 130 kDa, though specific isoforms may vary slightly in molecular weight .
Distinguishing between AIP1 isoforms requires careful experimental design:
Isoform-specific antibodies: Use antibodies raised against unique epitopes. For example, antibodies targeting the C-terminal region (aa 1350 to C-terminus for MAGI2/AIP1) can provide isoform specificity .
RT-PCR approach: Design primers that span unique exon-exon junctions or target isoform-specific sequences. For AIP1-2, semi-quantitative RT-PCR can confirm expression as demonstrated in Arabidopsis studies with primers flanking the insertion sites in the fifth exon (aip1.2-1) or fifth intron (aip1.2-2) .
Complementation assays: Re-express specific isoforms in knockout cells to confirm isoform-specific functions. This approach successfully identified the unique roles of AIP1-2 in Arabidopsis development when combined with act7 mutations .
Rigorous validation of AIP1-2 antibody specificity requires multiple controls:
Genetic knockout/knockdown validation: Compare antibody signal in wild-type versus AIP1-2 knockout/knockdown samples. In research with Arabidopsis, T-DNA insertion mutants (aip1.2-1 and aip1.2-2) provided essential negative controls for antibody validation .
Recombinant protein controls: Use purified GST-AIP1-2 or His-tagged AIP1-2 as positive controls in immunoblotting. These recombinant proteins have been successfully used in pull-down assays to verify interactions with actin .
Peptide competition assay: Pre-incubate the antibody with excess immunizing peptide prior to immunostaining or Western blotting. Signal reduction confirms specificity for the target epitope.
Cross-reactivity assessment: Test the antibody against related proteins (e.g., AIP1-1 in plant systems) to ensure specificity for the desired isoform .
Multiple antibody validation: Compare results using antibodies targeting different epitopes of the same protein. This approach helps confirm signal specificity across different experimental conditions.
RNAi rescue experiment: Restore protein expression using RNAi-resistant constructs to validate antibody-detected phenotypes. The RNase protection assay with AIP1 shRNA has been used to confirm specific silencing .
To effectively study AIP1-2 interactions with signaling proteins:
Co-immunoprecipitation (Co-IP):
Use anti-AIP1 antibody for immunoprecipitation followed by immunoblotting with antibodies against potential interacting partners (e.g., ASK1, 14-3-3, actins)
Include appropriate detergent conditions (typically 0.5-1% NP-40 or Triton X-100) to maintain protein-protein interactions
Validate with reverse Co-IP (precipitate with partner antibody, blot for AIP1)
Pull-down assays:
Proximity ligation assay (PLA):
Use for in situ detection of protein-protein interactions in fixed cells
Requires specific primary antibodies against both AIP1-2 and its interaction partner
Provides spatial information about interaction sites within cells
Mutational analysis:
Functional validation:
AIP1 expression changes have been documented in several pathological conditions:
Type 2 Diabetes:
AIP1 is significantly downregulated in omental adipose tissue of obese patients with T2D
AIP1 levels negatively correlate with insulin resistance markers (HOMA-IR, r = -0.4829) and waist-to-hip ratio (r = -0.2614)
Anti-AIP1 antibodies can quantify expression changes via Western blot and IHC in adipose tissue samples
After bariatric surgery (RYGB), AIP1 expression in adipose tissue normalizes, correlating with metabolic improvement
Alzheimer's Disease:
AIP1 is elevated in brain tissue from AD Tg2576 mice at both mRNA and protein levels
Aβ1-42 treatment increases AIP1 expression in cerebral microvascular endothelial cells
Antibodies against AIP1 can track these expression changes in brain tissue sections and primary cell cultures via Western blot and immunohistochemistry
Inflammatory Conditions:
When studying these changes, it's critical to use consistent sample collection protocols, appropriate normalization controls, and validated antibody dilutions for quantitative comparisons across disease stages.
For optimal immunohistochemical analysis of AIP1-2 in tissue sections:
Tissue fixation and processing:
Antigen retrieval:
Blocking and background reduction:
Antibody concentrations and incubation:
Visualization and counterstaining:
To effectively integrate AIP1-2 antibodies into multiplex protein analysis:
Multiplex immunofluorescence:
Use spectrally distinct fluorophores for simultaneous detection of AIP1-2 and interacting proteins
For example, co-staining of AIP1 with phosphorylated ASK1 (pSer-967) and 14-3-3 can visualize the dynamic regulation of this signaling complex
Select antibodies raised in different host species to avoid cross-reactivity with secondary antibodies
Include single-stain controls to establish specificity and assess bleed-through
Protein array applications:
Use purified AIP1-2 antibodies in reverse-phase protein arrays to screen multiple samples simultaneously
Include appropriate controls for normalization and antibody validation
This approach is valuable for screening AIP1-2 expression across multiple patient samples or experimental conditions
Mass cytometry (CyTOF):
Label AIP1-2 antibodies with metal isotopes for inclusion in high-dimensional CyTOF panels
Enables simultaneous measurement of AIP1-2 with dozens of other cellular markers
Particularly useful for analyzing AIP1-2 expression in heterogeneous cell populations
Proximity extension assay:
Utilize antibody pairs recognizing different epitopes on AIP1-2
Conjugate with DNA oligonucleotides that can form amplifiable reporters when in close proximity
Enables highly sensitive detection of AIP1-2 in complex biological samples
Implementation considerations:
Validate antibody performance in multiplexed format against single-plex controls
Optimize antibody concentrations to achieve comparable sensitivity across targets
Include both technical and biological replicates to ensure reproducibility
Common Western blot challenges with AIP1-2 antibodies include:
| Issue | Possible Causes | Solutions |
|---|---|---|
| No signal | Degraded protein, insufficient antibody concentration, poor transfer | Use fresh lysates with protease inhibitors; increase antibody concentration to 1:250-1:500; optimize transfer conditions for high molecular weight proteins (130 kDa) |
| Multiple bands | Cross-reactivity, protein degradation, post-translational modifications | Validate with knockout controls; use fresher samples with protease inhibitors; consider phosphatase inhibitors to preserve modification state |
| High background | Insufficient blocking, excessive antibody concentration, poor washing | Increase blocking time to 2 hours; titrate antibody concentration; add 0.1% Tween-20 to wash buffers and perform 5x10-minute washes |
| Inconsistent signal across experiments | Variable expression levels, inconsistent loading, antibody batch variation | Use housekeeping proteins (β-actin, β-tubulin) for normalization; perform Bradford assays for accurate loading; prepare sufficient antibody aliquots from single lot |
For optimal AIP1-2 detection, SDS-PAGE conditions should be adjusted for its molecular weight (approximately 130 kDa), and 7.5% gels are recommended for better resolution in this size range .
To optimize immunoprecipitation (IP) protocols for AIP1-2:
Lysis buffer optimization:
Use buffers that preserve protein-protein interactions while efficiently extracting AIP1-2
For co-IP of AIP1-2 with ASK1, a buffer containing 50 mM HEPES (pH 7.4), 150 mM NaCl, 1% Triton X-100, 10% glycerol, and protease/phosphatase inhibitors works effectively
Avoid harsh detergents like SDS that disrupt protein-protein interactions
Antibody selection and coupling:
For AIP1 IP, anti-AIP1 antibodies should be carefully validated for specificity
Pre-couple antibodies to Protein A/G beads (2-5 μg antibody per 20 μl beads) for more efficient capture
For difficult interactions, consider crosslinking antibodies to beads using dimethyl pimelimidate (DMP)
Pre-clearing samples:
Pre-clear lysates with Protein A/G beads for 1 hour at 4°C before adding antibody
This reduces nonspecific binding and decreases background
IP conditions optimization:
Perform IP overnight at 4°C with gentle rotation to maximize specific interactions
After IP, wash beads 3-5 times with lysis buffer to remove nonspecific binding
For detecting transient interactions, consider shorter incubation times or chemical crosslinking
Elution conditions:
For Western blot analysis, elute in Laemmli buffer at 70°C (not boiling) for 10 minutes
For mass spectrometry analysis, consider gentler elution using competing peptides or low pH
This approach has successfully detected interactions between AIP1 and ASK1, as well as the dissociation of 14-3-3 from ASK1 in response to Aβ1-42 treatment .
The choice between monoclonal and polyclonal antibodies for AIP1-2 research depends on specific experimental goals:
Use monoclonal antibodies when:
High reproducibility between experiments is critical
Specific epitopes or post-translational modifications need to be detected
Background must be minimized for clean Western blots or immunostaining
Long-term studies requiring consistent antibody performance are planned
Applications involve flow cytometry or immunoprecipitation of specific AIP1-2 forms
Use polyclonal antibodies when:
Maximum sensitivity is needed (multiple epitopes enhance signal)
Detecting AIP1-2 across multiple species (broader epitope recognition)
Studying native protein conformation or protein complexes
Performing initial characterization before investing in monoclonals
Working with fixed tissues where epitope availability may be limited
Research-based recommendations:
For studying specific AIP1-2 isoforms, monoclonal antibodies against unique regions provide better discrimination
For detecting endogenous AIP1-2 in tissues, polyclonal antibodies like those used in adipose tissue studies show good sensitivity
For co-immunoprecipitation experiments studying AIP1-ASK1 interactions, polyclonal antibodies have been successfully used
When studying signaling dynamics, monoclonal phospho-specific antibodies against downstream targets provide clearer results
The rabbit polyclonal antibody against AIP1 has demonstrated successful application in Western blot, immunohistochemistry, and co-immunoprecipitation experiments .
AIP1-2 antibodies show potential in engineered therapeutic applications:
Synthetic cytokine receptor systems:
Anti-idiotypic nanobodies (AIP VHH) against palivizumab have been developed as components of synthetic cytokine receptors
These systems utilize engineered IgG2 subclass antibodies to activate designer receptors
The AIP2gp130Δstalk construct demonstrates how AIP components can be incorporated into functional signaling systems
This approach allows for controlled activation of signaling pathways in therapeutic cell engineering
Monitoring therapeutic response:
Target validation for drug development:
Cell-based therapy monitoring:
Emerging technologies are expanding AIP1-2 antibody applications:
Single-cell protein analysis:
Integration of AIP1-2 antibodies into single-cell Western blot platforms
Allows correlation of AIP1-2 expression with cellular heterogeneity
Particularly valuable for analyzing AIP1-2 expression in mixed cell populations from tissue samples
Spatial transcriptomics with protein correlation:
Combining anti-AIP1-2 immunofluorescence with spatial transcriptomics
Reveals relationships between AIP1-2 protein expression and spatial gene expression patterns
Critical for understanding AIP1-2 function in complex tissues like adipose depots or brain sections
CRISPR screening validation:
Living cell imaging of AIP1-2 dynamics:
Anti-AIP1-2 nanobodies conjugated to fluorescent proteins for live-cell imaging
Enables real-time visualization of AIP1-2 translocation and complex formation
Critical for understanding the temporal dynamics of AIP1-ASK1-14-3-3 interactions
Mass spectrometry integration:
Immunoprecipitation with AIP1-2 antibodies followed by mass spectrometry
Identifies novel interaction partners and post-translational modifications
Provides comprehensive understanding of AIP1-2 in signaling networks
Key challenges in AIP1-2 antibody reproducibility include:
Epitope specificity issues:
Isoform cross-reactivity:
Species-specific validation gaps:
Many antibodies are validated in limited species (often human only)
Solution: Perform cross-species validation using sequence alignment and recombinant proteins from target species
Application-specific optimization:
Standardization of protocols:
Variable results between labs due to protocol differences
Solution: Adopt detailed standardized protocols for sample preparation, antibody dilutions, and detection methods
Reproducibility documentation: