Antigen Target: The antibody recognizes an epitope within the internal region (AA 24–126) of human VIPR2, a receptor for VIP and PACAP-38/27 .
Conjugation: FITC (fluorescein isothiocyanate) is covalently attached to the antibody, enabling fluorescence detection in imaging and flow cytometry .
Host and Clonality: It is a rabbit polyclonal antibody, offering broad epitope recognition .
Reactivity: Primarily reacts with human VIPR2, with predicted cross-reactivity in rat and mouse .
VIPR2 promotes cancer cell migration via PI3K/AKT signaling and WAVE2-mediated actin remodeling .
FITC-conjugated antibodies (e.g., from R&D Systems) were used to confirm VIPR2 membrane localization in lamellipodia of migrating cells .
In the suprachiasmatic nucleus (SCN), VIPR2 antibodies (e.g., Alomone #AVR-002) revealed VIPR2’s role in circadian rhythm regulation .
VIPR2 is expressed in CD4+ T cells and plasmacytoid dendritic cells, with antibodies aiding in immune cell profiling .
VIPR2 (Vasoactive Intestinal Peptide Receptor 2) is a G protein-coupled receptor that belongs to the vasoactive intestinal peptide receptor family. It functions as a receptor for vasoactive intestinal peptide (VIP), a neuropeptide with diverse physiological roles. VIPR2 mediates multiple critical biological processes, including:
Regulation of circadian rhythm
Modulation of neurotransmission
Immune response regulation
Smooth muscle relaxation
Exocrine and endocrine secretion regulation
Water and ion homeostasis
Dysregulation of VIPR2 signaling has been implicated in various neurological and endocrine disorders, making it a promising target for therapeutic development. Understanding the function of VIPR2 is crucial for elucidating its role in these diseases and potentially identifying novel treatment strategies .
FITC (Fluorescein Isothiocyanate) conjugation provides direct fluorescent labeling of VIPR2 antibodies, enabling visualization without secondary detection reagents. This modification affects antibody functionality in several ways:
Enables direct detection in flow cytometry, immunofluorescence, and live cell imaging
Eliminates potential cross-reactivity issues from secondary antibodies
May slightly reduce binding affinity compared to unconjugated antibodies due to steric hindrance
Can affect the shelf-life and stability (FITC is sensitive to photobleaching)
Optimal pH for FITC fluorescence is 8.0, which may influence experimental conditions
For applications requiring quantitative analysis, researchers should be aware that the degree of labeling (number of FITC molecules per antibody) can affect both binding capacity and signal intensity .
VIPR2 Antibody, FITC conjugated has been validated for several research applications, including:
Application | Recommended Dilution | Special Considerations |
---|---|---|
Flow Cytometry (FACS) | 1:50-1:200 | Optimal for cell surface receptor detection |
Immunocytochemistry (ICC) | 1:100-1:400 | Live cell imaging possible |
Immunofluorescence (IF) | 1:100-1:400 | Low background with proper blocking |
ELISA | 1:2000-1:5000 | Direct detection format |
The antibody shows high reactivity with human samples and has cross-reactivity with rat and mouse VIPR2, making it versatile for comparative studies across species. For optimal results, validation in your specific experimental system is recommended .
VIPR2 Antibody, FITC conjugated can be used effectively with various biological sample types:
Cell lines expressing VIPR2 (particularly neuronal, endocrine, and immune cell lineages)
Primary tissue sections (brain, pancreas, intestine, immune tissues)
Peripheral blood mononuclear cells (PBMCs)
Isolated T-cells (important for immune regulation studies)
Tissue microarrays for high-throughput screening
When working with tissue samples, proper fixation protocols that preserve epitope accessibility while maintaining tissue architecture are critical. For cell suspensions, gentle fixation methods that don't disrupt membrane proteins are recommended .
For optimal detection of VIPR2 using FITC-conjugated antibodies, sample preparation protocols should be tailored to the application:
For flow cytometry:
Use single-cell suspensions (1-5×10^6 cells/mL)
Gentle fixation with 0.5-2% paraformaldehyde
Permeabilization only if detecting intracellular epitopes
Blocking with 5% normal serum from the same species as secondary antibody
For immunofluorescence:
4% paraformaldehyde fixation (10-15 minutes)
Antigen retrieval if using paraffin sections
0.1-0.3% Triton X-100 for permeabilization
Extended blocking (1-2 hours) to reduce background
Counterstaining nuclei with DAPI
For all applications, inclusion of proper negative controls (isotype control, FITC-conjugated non-specific antibody) is essential to establish specificity of staining patterns .
VIPR2 is critically involved in circadian rhythm regulation, particularly in the suprachiasmatic nucleus (SCN). FITC-conjugated VIPR2 antibodies enable precise investigation of this system through:
Temporal expression profiling: Track VIPR2 expression throughout circadian cycles using timed tissue collection and flow cytometry quantification
Co-localization studies: Combine with markers for other circadian regulators (PER, CRY, CLOCK) using multi-channel immunofluorescence
Ex vivo SCN slice cultures: Visualize real-time VIPR2 dynamics in response to phase-shifting stimuli
Receptor trafficking: Monitor internalization and recycling of VIPR2 following VIP stimulation
A methodological approach would involve:
Harvest SCN tissue at 4-hour intervals across a 24-hour period
Process for either flow cytometry or tissue sectioning
Label with VIPR2 Antibody, FITC conjugated (1:100 dilution)
Quantify fluorescence intensity and localization patterns
Correlate with circadian time points and behavioral outputs
This approach has revealed that VIPR2 undergoes rhythmic expression changes with peak levels during the subjective night, coinciding with periods of maximal VIP sensitivity .
When using VIPR2 Antibody, FITC conjugated for flow cytometry, the following controls are essential for reliable data interpretation:
Control Type | Purpose | Implementation |
---|---|---|
Unstained Cells | Establish autofluorescence baseline | Process identical cells without any antibody |
Isotype Control | Identify non-specific binding | Use FITC-conjugated IgG matching the antibody's host and isotype |
Fluorescence Minus One (FMO) | Set accurate gating | Include all fluorophores except FITC |
Blocking Peptide | Confirm specificity | Pre-incubate antibody with immunizing peptide |
Positive Control | Validate detection | Use cells known to express VIPR2 (e.g., T cells) |
Negative Control | Confirm specificity | Use VIPR2-knockout or siRNA-treated cells |
Additionally, titration experiments should be performed to determine optimal antibody concentration, as both insufficient and excessive antibody can compromise data quality. For multipanel flow cytometry, spectral overlap between FITC and other fluorophores (particularly PE) should be carefully compensated .
Validating VIPR2 Antibody specificity in neurological tissues requires a multi-faceted approach:
Comparative immunohistochemistry:
Parallel staining with multiple VIPR2 antibodies targeting different epitopes
Comparison of staining patterns with published atlas data
Cross-validation with in situ hybridization for VIPR2 mRNA
Knockout/knockdown validation:
Use of VIPR2 knockout tissues as negative controls
RNAi-mediated knockdown in primary neuronal cultures
CRISPR-Cas9 edited cell lines or tissues
Peptide competition assays:
Pre-incubation of antibody with excess immunizing peptide
Titration of blocking peptide to determine concentration-dependent inhibition
Use of related but distinct peptides to confirm epitope specificity
Western blot correlation:
Confirmation that tissue lysates show bands of expected molecular weight
Correlation between band intensity and immunofluorescence signal strength
This comprehensive validation approach ensures that observed staining truly represents VIPR2 distribution, which is particularly critical in neurological tissues where non-specific binding can be problematic .
Recent research has revealed important connections between VIPR2 signaling and T-cell-mediated anti-leukemic activity:
VIPR2 signaling typically exerts immunosuppressive effects on T cells. When VIPR2 is engaged by VIP, it triggers cAMP production that dampens T-cell activation. Conversely, VIPR2 antagonism enhances T-cell responses against leukemia cells through multiple mechanisms:
Increased T-cell proliferation following activation
Enhanced production of pro-inflammatory cytokines (IFN-γ, TNF-α)
Augmented cytotoxic activity against leukemia cells
Reduced regulatory T-cell suppressive function
Improved memory T-cell formation
Experimental evidence shows that VIP antagonists (VIPhyb) enhance T-cell activation and induce T-cell-dependent anti-leukemic activity in murine models of acute myeloid leukemia. The efficacy of these antagonists correlates positively with their predicted binding affinity to VIP receptors .
FITC-conjugated VIPR2 antibodies can be used to:
Monitor receptor expression during different phases of T-cell activation
Sort VIPR2-high and VIPR2-low T-cell populations for functional studies
Track receptor internalization following antagonist treatment
Correlate VIPR2 expression with anti-tumor activity in patient samples
These approaches have identified ANT308 and ANT195 as promising VIP-R antagonists with enhanced potency for inducing anti-leukemia immune responses .
VIPR2 antagonists and VIPR2 antibodies represent complementary but distinct research tools with different applications and mechanisms:
Aspect | VIPR2 Antagonists | VIPR2 Antibodies (FITC Conjugated) |
---|---|---|
Mechanism | Compete with VIP for receptor binding | Bind to receptor epitopes (may not affect function) |
Function | Block VIP signaling | Detect/visualize VIPR2 |
Applications | Functional studies, therapeutic development | Detection, localization, quantification |
Delivery Systems | Can be administered in vivo | Primarily for ex vivo/in vitro use |
Readouts | Physiological responses, signaling changes | Receptor expression, distribution, trafficking |
Specificity | May cross-react with VPAC1 | Epitope-specific binding |
The development of C-terminal sequence variations of VIPhyb has yielded improved VIP-R antagonists with enhanced receptor binding and plasma stability. In contrast, FITC-conjugated VIPR2 antibodies are primarily analytical tools that enable visualization and quantification of the receptor in research samples.
For comprehensive studies, researchers often use both approaches: antagonists to modulate function and antibodies to monitor expression and localization changes resulting from that modulation. This combination has proven particularly valuable in cancer immunology research investigating how VIPR2 signaling affects T-cell responses to malignancies .
To maintain optimal activity of VIPR2 Antibody, FITC conjugated, adhere to these storage and handling guidelines:
Store at -20°C in small aliquots to minimize freeze-thaw cycles
Protect from light using amber tubes or by wrapping in aluminum foil
Store in stabilizing buffer (typically pH 7.4 PBS with 0.05% NaN3 and 40% glycerol)
When working with the antibody, keep on ice and protected from direct light
Centrifuge briefly before opening vial to collect solution at the bottom
Do not dilute until immediately before use
Use within 12 months for optimal results
FITC conjugates are particularly sensitive to photobleaching, so minimize exposure to light during all handling steps. For long-term storage beyond 12 months, -80°C is recommended. Always verify activity with positive controls before use in critical experiments .
Optimizing multiplexed immunofluorescence with FITC-conjugated VIPR2 antibody requires careful consideration of fluorophore combinations and staining protocols:
Fluorophore selection strategies:
Pair FITC (excitation: 495nm, emission: 519nm) with fluorophores having minimal spectral overlap
Recommended partners: Cy5 (far-red), APC (red), or coumarin derivatives (blue)
Avoid PE or rhodamine derivatives which have significant overlap with FITC
Sequential staining protocol:
Apply VIPR2 Antibody, FITC conjugated first (1:100 dilution)
Wash thoroughly (3-5 times with PBS + 0.05% Tween-20)
Block with 2% normal serum from host of second primary antibody
Apply second primary antibody
Use fluorophore-conjugated secondary antibody for detecting second primary
Controls for co-localization studies:
Single-color controls for each fluorophore
Isotype controls for each primary antibody
Absorption controls (pre-incubation with blocking peptides)
Co-localization threshold controls using known interacting and non-interacting proteins
For optimal results, confocal microscopy with sequential scanning is preferred over wide-field fluorescence to minimize bleed-through. Post-acquisition processing should include background subtraction and careful thresholding based on control samples .
When encountering issues with VIPR2 Antibody, FITC conjugated staining, consider these systematic troubleshooting approaches:
For weak signal:
Increase antibody concentration (try 2-fold increments)
Extend incubation time (overnight at 4°C)
Optimize fixation (try shorter fixation times to preserve epitopes)
Enhance antigen retrieval (test multiple methods: citrate, EDTA, enzymatic)
Use signal amplification systems (tyramide signal amplification)
Check sample viability and VIPR2 expression level in your specific sample type
For high background/non-specific staining:
Increase blocking duration and concentration (5-10% normal serum, 2 hours)
Add 0.1-0.3% Triton X-100 to blocking solution
Include 0.1-0.3% BSA in all antibody dilution buffers
Increase wash steps (5-6 washes, 5 minutes each)
Pre-adsorb antibody with tissue powder from non-expressing tissues
Include human Fc block when working with human samples
For autofluorescence issues:
Include Sudan Black B treatment (0.1% in 70% ethanol, 20 minutes)
Use specialized autofluorescence quenching reagents
Employ spectral unmixing during confocal microscopy acquisition
Consider time-resolved fluorescence to separate FITC signal from autofluorescence
Documentation of all optimization steps in a laboratory notebook will facilitate reproducible protocols for future experiments .
VIPR2 Antibody, FITC conjugated provides valuable tools for investigating neurological disorders through several methodological approaches:
Receptor distribution mapping:
Comparative analysis of VIPR2 expression in healthy vs. diseased brain tissue
High-resolution mapping of receptor localization in specific neuronal populations
Quantitative assessment of receptor density changes during disease progression
Schizophrenia research applications:
The VIPR2 gene (designated as SCZD16 in some databases) has been implicated in schizophrenia
Fluorescent antibodies enable visualization of altered receptor distribution in patient-derived samples
Flow cytometric quantification of VIPR2 levels in peripheral immune cells as potential biomarkers
Circadian rhythm disorder investigations:
Visualization of altered VIPR2 expression in suprachiasmatic nucleus tissue
Correlation of receptor distribution with sleep-wake cycle abnormalities
Monitoring changes in receptor trafficking following chronobiotic treatments
Neurodevelopmental studies:
Tracking VIPR2 expression changes during critical periods of brain development
Investigation of receptor clustering in developing neuronal networks
Analysis of VIPR2-associated signaling during neurite outgrowth and synaptogenesis
These applications rely on the specificity of FITC-conjugated VIPR2 antibodies for visualizing subtle changes in receptor distribution that may contribute to neurological dysfunction .
Studying VIPR2 internalization and trafficking using FITC-conjugated antibodies requires specialized techniques to capture receptor dynamics:
Live-cell imaging approaches:
Pulse-chase labeling with VIPR2 Antibody, FITC conjugated
Time-lapse confocal microscopy to track receptor movement
Photobleaching techniques (FRAP/FLIP) to measure mobility
Endocytic pathway characterization:
Co-localization with endosomal markers (Rab5, Rab7, Rab11)
Pharmacological inhibitors of different endocytic routes
Dynamin inhibitors to block clathrin-dependent internalization
Quantitative internalization assays:
Acid wash removal of surface-bound antibody
Flow cytometry to measure internalization rate
Biotinylation-based biochemical assays
Recycling vs. degradation fate determination:
Dual-pulse labeling with different color conjugates
Co-localization with lysosomal markers (LAMP1)
Inhibitors of lysosomal degradation (chloroquine, bafilomycin A)
A typical protocol would involve:
Surface labeling with VIPR2 Antibody, FITC conjugated at 4°C (prevents internalization)
Washing to remove unbound antibody
Warming to 37°C to permit internalization
Imaging at defined time intervals (0, 5, 15, 30, 60 min)
Quantification of surface vs. intracellular fluorescence
These approaches can reveal how VIPR2 trafficking is regulated in response to VIP stimulation or antagonist treatment, providing insights into receptor desensitization mechanisms .
VIPR2 Antibody, FITC conjugated plays a critical role in immunotherapy development research through several methodological approaches:
Target validation and expression profiling:
Flow cytometric screening of patient samples to identify VIPR2-high populations
Correlation of VIPR2 expression with immunosuppressive tumor microenvironments
Isolation of VIPR2+ cells for functional characterization
Therapeutic response monitoring:
Before/after treatment analysis of VIPR2 expression on immune cells
Real-time monitoring of receptor occupancy during VIP antagonist therapy
Correlation of receptor modulation with clinical outcomes
Mechanism of action studies:
Visualization of VIPR2 distribution changes following antagonist treatment
Assessment of receptor clustering and signaling complex formation
Co-localization with downstream signaling molecules
Companion diagnostic development:
Standardization of VIPR2 detection protocols for patient stratification
Threshold determination for predicting response to VIP-targeted therapies
Validation across different tissue types and disease states
Research has demonstrated that VIP antagonists enhance T-cell activation and induce T-cell-dependent anti-leukemic activity. FITC-conjugated VIPR2 antibodies enable monitoring of receptor expression and distribution changes during antagonist treatment, providing mechanistic insights that guide further therapeutic development .
Correlating VIPR2 expression with functional outcomes requires integrated methodological approaches:
Sequential tissue analysis workflow:
Collect matched tissue samples for both staining and functional assays
Section tissue into adjacent slices for different analyses
Perform VIPR2 immunofluorescence with FITC-conjugated antibody
Conduct functional assays on matched sections (enzyme activity, electrophysiology, etc.)
Map expression patterns to functional readouts using image registration
Single-cell analysis techniques:
Flow cytometric sorting of cells based on VIPR2-FITC signal intensity
Functional assessment of VIPR2-high vs. VIPR2-low populations
Correlation of receptor level with cellular responsiveness to VIP
Integration with single-cell transcriptomics for comprehensive profiling
Ex vivo tissue culture approaches:
Precision-cut tissue slices maintained in culture
VIPR2 visualization using FITC-conjugated antibody
Functional testing with receptor agonists/antagonists
Time-lapse correlation of receptor dynamics with functional outputs
In situ hybridization correlation:
RNAscope for VIPR2 mRNA detection on adjacent sections
Correlation of protein expression with transcript levels
Assessment of post-transcriptional regulation
These approaches have revealed that in leukemia research, T cells with higher VIPR2 expression show differential responses to VIP antagonist treatment, with expression levels correlating with anti-tumor activity potential .
VIPR2 Antibody, FITC conjugated is enabling several cutting-edge applications in cancer immunology research:
Immune checkpoint modulation studies:
VIPR2 signaling functions as an immune checkpoint in the tumor microenvironment
FITC-conjugated antibodies enable visualization of receptor distribution on tumor-infiltrating lymphocytes
Flow cytometric analysis of VIPR2 expression correlates with T-cell exhaustion markers
CAR-T cell engineering applications:
Selection of VIPR2-low T cells for CAR modification (enhanced persistence)
Monitoring VIPR2 expression changes during CAR-T manufacturing
Correlation of pre-infusion VIPR2 levels with in vivo efficacy
Combination therapy development:
Visualization of VIPR2 modulation when combining VIP antagonists with other immunotherapies
Assessment of receptor redistribution following checkpoint blockade
Identification of optimal sequencing for multi-modal immunotherapy
Biomarker development:
VIPR2 expression profiling across cancer types to identify responsive subsets
Correlation of receptor levels with tumor microenvironment immune composition
Development of predictive algorithms based on VIPR2 distribution patterns
Recent research has demonstrated that VIP antagonists with improved docking scores for human VIP receptors VPAC1 and VPAC2 enhance T cell-dependent anti-leukemia responses. The predicted binding affinity of these antagonists correlates positively with their ability to augment T-cell proliferation and anti-leukemia activity, highlighting the importance of precisely characterizing VIPR2 expression and modulation in cancer immunotherapy development .
Emerging research directions for VIPR2 Antibody applications span multiple fields and methodological advances:
Integration with advanced imaging technologies:
Super-resolution microscopy for nanoscale receptor clustering analysis
Expansion microscopy for improved spatial resolution of VIPR2 distribution
Light-sheet microscopy for whole-organ receptor mapping
Intravital imaging for real-time receptor dynamics in living tissues
Multi-omics integration approaches:
Correlation of VIPR2 protein expression with spatial transcriptomics
Integrated proteomics to identify VIPR2 interaction networks
Metabolomics correlation with receptor signaling activity
Novel therapeutic targeting strategies:
Development of VIPR2-targeted antibody-drug conjugates
Bispecific antibodies linking VIPR2 with immune effector cells
Nanoparticle-delivered VIP antagonists with improved pharmacokinetics
Expanded disease applications:
Neurodegenerative disorders beyond circadian rhythm disruption
Inflammatory bowel disease and gut-brain axis investigation
Metabolic disorders with neuroendocrine components
Advanced conjugation technologies:
Photoswitchable fluorophores for super-resolution applications
Quantum dot conjugation for improved stability and brightness
Multicolor VIPR2 antibody panels for multiplexed receptor family analysis