VPAC1 is a G protein-coupled receptor (GPCR) that binds vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase-activating polypeptide (PACAP) . Antibodies targeting VPAC1 enable researchers to:
Quantify receptor expression in immune cells via flow cytometry
Study receptor localization through immunofluorescence microscopy
Investigate VIP/VPAC1 signaling pathways in inflammation and immunity
Specificity: No cross-reactivity with human VPAC1, VPAC2, or PAC1 receptors .
Applications:
Technical Notes:
Design: DyLight 650-conjugated nanobodies (CA7277, CA7281, etc.) .
Performance: Comparable to monoclonal antibodies in flow cytometry for primary human leukocytes .
Target Cells:
Immune Regulation:
Therapeutic Potential:
KEGG: ath:AT3G01310
UniGene: At.47813
VIP1 antibodies fall into two distinct categories based on their target proteins:
Plant VIP1 antibodies target the plant-specific VirE2-interacting protein 1, a bZIP transcription factor involved in stress responses and Agrobacterium-mediated transformation .
Mammalian VPAC1 (VIP1 receptor) antibodies recognize the Vasoactive Intestinal Peptide Receptor 1, a G-protein coupled receptor that mediates VIP and PACAP signaling in various tissues .
Selection criteria should include:
Species specificity: Confirm the antibody recognizes your target species. For example, the α-mVPAC1 polyclonal antibody described in recognizes mouse but not human VPAC1.
Application compatibility: Verify suitability for your intended application (Western blot, flow cytometry, immunohistochemistry, etc.)
Epitope location: For VPAC1, antibodies targeting extracellular domains are preferable for detecting native receptor on cell surfaces .
Validation data: Look for antibodies validated in knockout/mutant models that demonstrate specificity.
Available formats include:
Unconjugated primary antibodies
Biotinylated antibodies for sensitivity enhancement (though not always beneficial)
Rigorous validation is critical due to potential cross-reactivity with related proteins:
For VPAC1 antibodies:
Cross-reactivity testing: Test antibody against cells expressing related receptors. The α-mVPAC1 pAb described in was validated by demonstrating it does not recognize other family receptors (mouse VPAC2 and PAC1, human VPAC1, VPAC2 and PAC1) by flow cytometry.
Knockout validation: Compare staining between wild-type and knockout samples. This provides the most definitive specificity control .
Cell type expression patterns: Validate using cell types with known differential expression patterns. For example, flow cytometry should detect VPAC1 on resting T cells but not activated T cells or B cells .
Peptide competition: Pre-incubate antibody with immunizing peptide to confirm signal elimination.
For plant VIP1 antibodies:
Mutant comparison: Compare signal between wild-type plants and vip1 mutants (such as vip1-1 or vip1-2) .
Recombinant protein controls: Test antibody against purified recombinant VIP1.
Subcellular localization: VIP1 should show nuclear localization during stress conditions .
For mammalian VPAC1:
The expected molecular weight ranges from 55-70 kDa on Western blots .
Post-translational modifications (particularly glycosylation) can cause variation in the observed molecular weight.
Denaturation conditions can affect epitope accessibility, particularly for antibodies targeting conformational epitopes.
For plant VIP1:
VIP1 is a 341 amino acid protein with a calculated molecular weight of approximately 39 kDa .
The vip1-1 mutant produces a truncated protein of the first 244 amino acids, while vip1-2 encodes only the first 140 amino acids plus five additional amino acids from a frameshift mutation .
When troubleshooting unexpected molecular weights:
Try different sample preparation methods (reducing vs. non-reducing conditions)
Compare with positive control samples
Consider using multiple antibodies targeting different epitopes
Verify protein identity using mass spectrometry
Flow cytometry is particularly valuable for studying VPAC1 expression in immune cell populations. Based on the successful methodology described in :
Sample preparation:
Freshly isolated primary cells yield optimal results
Maintain cells at 4°C during staining to prevent receptor internalization
Include a viability dye to exclude dead cells
Antibody selection and optimization:
Multiparameter analysis:
Design panels to simultaneously measure VPAC1 and activation markers (e.g., CD44)
Include lineage markers to identify specific cell populations
Critical findings:
Resting T cells (CD44 low) express high levels of VPAC1
Activated T cells (CD44 high) downregulate VPAC1 to undetectable levels
B cells express minimal VPAC1
Cell Population | VPAC1 Expression | CD44 Status |
---|---|---|
Resting CD4+ T cells | High | CD44 Low-Int |
Activated CD4+ T cells | Undetectable | CD44 High |
Resting CD8+ T cells | High | CD44 Low-Int |
Activated CD8+ T cells | Undetectable | CD44 High |
B cells (CD19+) | Undetectable | N/A |
This pattern confirms that VPAC1 protein downregulation parallels previously observed mRNA downregulation during T cell activation, suggesting that VPAC1 regulation occurs primarily at the transcriptional or post-transcriptional level .
To investigate plant VIP1's function as a transcription factor:
DNA binding assays:
Transcriptional activation assays:
Reporter gene systems: Using VRE-containing promoters fused to reporter genes. VIP1 strongly enhances expression from synthetic promoters harboring multiple VRE copies .
Point mutation analysis: Studies have shown correlation between VIP1-VRE binding and transcriptional activation by introducing mutations in the VRE sequence .
Protein interaction studies:
Promoter activation by VIP1 is influenced by bacterial and plant proteins that interact with VIP1 during Agrobacterium infection (VirE2, VirF, VIP2) .
VirF, an F-box protein, strongly decreased VIP1 transcriptional activation ability without affecting its binding to VRE in vitro, likely by triggering proteasomal degradation of VIP1 .
Functional analysis:
For mammalian VPAC1:
Tissue preparation:
Fixation should preserve membrane proteins without masking epitopes
For paraffin sections, appropriate antigen retrieval is critical
Fresh frozen sections may better preserve epitopes but require different fixation protocols
Antibody optimization:
Visualization approaches:
Controls:
Include tissue from VPAC1 knockout animals when available
Peptide competition controls can confirm specificity
Include tissues with known high and low expression
For plant VIP1:
Tissue fixation:
Paraformaldehyde fixation preserves protein localization
Permeabilization is crucial for detecting nuclear proteins like VIP1
Detection strategies:
Confocal microscopy can track nuclear translocation following stress
Dual staining with nuclear markers confirms localization
Experimental design:
The observation that VPAC1 is downregulated during T cell activation has important functional implications. Design your experiments considering:
Kinetic analysis:
Use flow cytometry with anti-VPAC1 antibodies to track the timing of VPAC1 downregulation relative to activation markers
Compare protein downregulation with previously established mRNA downregulation kinetics
Determine whether receptor internalization precedes degradation
Mechanism investigation:
Previous studies showed VPAC1 mRNA downregulation was blocked by inhibitors against Fyn, Lck, and JNK kinases
Design experiments using these inhibitors with flow cytometric detection of VPAC1 protein
Develop co-immunoprecipitation protocols to identify proteins interacting with VPAC1 during activation
Functional consequences:
VPAC1 signaling suppresses T cell activation by inhibiting IL-2 and IL-4 expression
Design experiments to test if preventing VPAC1 downregulation affects cytokine production and T cell proliferation
Investigate effects on regulatory T cell (Treg) differentiation, as VIP/VPAC1 signaling enhances Treg development
In vivo relevance:
Plant VIP1 functions as a stress-responsive transcription factor with roles in defense. Design your experiments to investigate:
Stress-induced translocation:
Pathogen response studies:
Target gene regulation:
Protein interaction networks:
Inconsistent flow cytometry results with VPAC1 antibodies can have several causes:
Variable expression levels:
Technical considerations:
Receptor internalization: Maintain cells at 4°C during processing
Antibody concentration: Optimal concentration may differ between applications
Cell viability: Dead cells can give false-positive signals
Antibody-specific factors:
Lot-to-lot variation in polyclonal antibodies
Storage conditions affecting antibody quality
Freezing/thawing cycles reducing antibody activity
Optimization strategies:
Alternative approaches:
Contradictory results with different antibodies are a common challenge. Address this by:
Epitope mapping:
Validation hierarchy:
Prioritize results from antibodies validated with genetic knockout models
Compare antibody results with orthogonal techniques (mRNA analysis, mass spectrometry)
Consider using multiple antibodies targeting different epitopes in parallel
Experimental conditions:
Cross-reactivity assessment:
Species considerations: