VIPR2 Antibody, Biotin conjugated

Shipped with Ice Packs
In Stock

Description

Definition and Composition

VIPR2 antibodies conjugated to biotin are immunoglobulin-based reagents designed to bind specifically to VIPR2 antigens. The biotin tag enables high-sensitivity detection via streptavidin-linked enzymes (e.g., HRP) or fluorescent probes in assays such as ELISA, Western blot (WB), and flow cytometry. Key components include:

  • Target Epitope: Typically raised against specific regions of VIPR2, such as the N-terminal (amino acids 95–123 in humans) or internal domains (e.g., AA 81–170 in rats) .

  • Host Species: Commonly produced in rabbits (polyclonal) or mice (monoclonal, e.g., clone 2G9) .

  • Conjugation Method: Biotin is chemically linked to the antibody’s Fc region, preserving antigen-binding specificity .

3.1. Immunoassays

  • ELISA: Used to quantify VIPR2 in serum or cell lysates. Example protocol:

    1. Coat plates with samples or standards.

    2. Incubate with biotin-conjugated VIPR2 antibody (1:100–1:5000 dilution).

    3. Detect with streptavidin-HRP and TMB substrate .

  • Western Blot: Detects VIPR2 at ~49 kDa in human tissues (1:1000 dilution recommended) .

3.2. Cell Signaling Studies

  • VIPR2 antagonists (e.g., KS-133) reduce cancer cell proliferation by blocking cAMP/PKA/ERK and PI3K/AKT/GSK3β pathways. Biotinylated antibodies enable tracking of VIPR2 expression changes in these contexts .

3.3. Neuroimmunology

  • In Parkinson’s disease models, VIPR2 agonists (e.g., LBT-3627) upregulate regulatory T cells (Tregs). Biotin-conjugated antibodies help map VIPR2 distribution in brain tissues .

4.1. Cancer Biology

  • KS-133, a VIPR2 antagonist, suppresses cyclin D1 via ERK and PI3K pathways, reducing breast cancer (MCF-7) cell proliferation by 40% .

  • Biotinylated VIPR2 antibodies confirmed receptor overexpression in tumor microenvironments, correlating with poor prognosis .

4.2. Neuroprotection

  • In α-synuclein-overexpressing rats, VIPR2 activation with LBT-3627 reduced dopaminergic neuron loss by 36% and microglial inflammation by 27% .

4.3. Circadian Rhythms

  • VIPR2 antibodies localized receptors in >90% of suprachiasmatic nucleus (SCN) neurons, critical for maintaining circadian synchrony .

Technical Considerations

  • Biotinylation Advantages:

    • High affinity for streptavidin (Kd = 4 × 10⁻¹⁴ M) minimizes nonspecific binding .

    • Site-specific conjugation preserves antibody efficacy .

  • Limitations:

    • Endogenous biotin in samples (e.g., serum) may require blocking steps .

    • Batch-dependent variability in concentration necessitates validation .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship your order within 1-3 business days of receipt. Delivery times may vary depending on the shipping method and destination. Please contact your local distributor for specific delivery timeframes.
Synonyms
VIPR2; VIP2R; Vasoactive intestinal polypeptide receptor 2; VIP-R-2; Helodermin-preferring VIP receptor; Pituitary adenylate cyclase-activating polypeptide type III receptor; PACAP type III receptor; PACAP-R-3; PACAP-R3; VPAC2
Target Names
Uniprot No.

Target Background

Function
This receptor is a binding site for VIP, PACAP-38, and PACAP-27. Its activity is mediated by G proteins, which activate adenylyl cyclase. It can also be coupled to phospholipase C.
Gene References Into Functions
  1. A recent study reported a novel gene duplication syndrome (10q21.2q21.3 microduplication) and provided new evidence for VIPR2 duplication as a potential candidate gene for autism. PMID: 27796743
  2. Research suggests that VIPR2, a negative regulator of smooth muscle cell proliferation, may be a novel tumor suppressor gene in uterine leiomyosarcomas. PMID: 29063609
  3. The 'CC' genotype of the VIPR2 gene was nominally associated with an increased risk of schizophrenia in male Han Chinese patients. PMID: 27156032
  4. Lower CpG methylation of VIPR2 was observed in the saliva of children with ADHD. PMID: 26304033
  5. CD4+ T cells in HIV infection exhibit increased levels of VPAC2 expression. PMID: 24469917
  6. This study suggests that carriers of microduplication genotypes of VIPR2 are predisposed to schizophrenia in Han Chinese. PMID: 24794882
  7. Monocytes from Sjogren's syndrome patients display increased vasoactive intestinal peptide receptor 2 expression and impaired apoptotic cell phagocytosis. PMID: 24827637
  8. Data indicate that VIP and PACAP enhance macrophage resistance to HIV-1 replication by inducing the synthesis of beta-chemokines CCL3 and CCL5, and IL-10 following preferential activation of the receptors VPAC2 and PAC1. PMID: 23818986
  9. This is the first study to suggest a role for VIPR2 in the genetic susceptibility to high myopia. EGR1, JUN, FOS, and VIP are unlikely to be important in predisposing humans to high myopia. PMID: 23637909
  10. Genetic testing for VIPR2-LCR-associated inversions should be performed on available cohorts of psychiatric patients to evaluate their potential pathogenic role. PMID: 23073313
  11. The study concludes that VPAC2/PAC1 receptors require NO in series to effect cutaneous active vasodilation during heat stress in humans. PMID: 22961270
  12. PACAP causes PAC1/VPAC2 receptor mediated hypertension and sympathoexcitation in normal and hypertensive rats. PMID: 22886412
  13. Gene expression levels and cAMP signaling of VIPR2 were increased in patients carrying 7q36.3 microduplications, thus implicating VIPR2 in the etiology of schizophrenia. [review] PMID: 21721910
  14. VPAC(2) receptor presents an extranuclear localization, and its protein expression is lower than that of the VPAC(1) receptor in human breast tissue samples. PMID: 20691743
  15. VPAC2 and/or PAC1 receptor activation is involved in cutaneous active vasodilation in humans. PMID: 20395540
  16. Detection of beta-galactosidase marker for human VPAC2/VIPR2 in cells within the suprachiasmatic nucleus (SCN) of transgenic mice indicates that VPAC2 may contribute to autoregulation and/or coupling within the SCN core and to control of the SCN shell. PMID: 15090046
  17. Analysis of a mutant form of VPAC2 demonstrates its role in signaling and ligand binding. PMID: 15302876
  18. A novel recombinant agonist for VPAC2 is not active against PAC1. PMID: 16500728
  19. The abnormal expression of VPCAP2-R mRNA in gallbladder tissue may play a role in the formation of gallbladder stones and gallbladder polyps. PMID: 16552823
  20. VPAC2-R mRNA was visualized only in the cerebellum of 7-22-year-old subjects. PMID: 16572459
  21. Splice variants may modify the immunoregulatory contributions of the VIP-VPAC2 axis. PMID: 16888203
  22. Identification and characterization of novel five-transmembrane (5TM) isoforms of VPAC2. PMID: 16934434
  23. Altered expression of VPAC2 in activated CD4+ T cells derived from multiple sclerosis (MS) patients rendered CD4+ T cells less responsive to VIP and skewed the system to a predominantly T(h)1 direction. PMID: 17077178
  24. Daily stimulation of VPAC2, but not VPAC1 or PAC1, resulted in up to 90% inhibition of X4 or R5 productive infections in either cell lines or PBMCs. PMID: 17257640
  25. A novel mechanism of calmodulin in regulating PACAP signaling by possible interaction with the inactive state of PAC1 and VPAC2 receptors. PMID: 19269029

Show More

Hide All

Database Links

HGNC: 12695

OMIM: 601970

KEGG: hsa:7434

STRING: 9606.ENSP00000262178

UniGene: Hs.585052

Protein Families
G-protein coupled receptor 2 family
Subcellular Location
Cell membrane; Multi-pass membrane protein.
Tissue Specificity
Expressed in CD4+ T-cells, but not in CD8+ T-cells. Expressed in the T-cell lines Jurkat, Peer, MOLT-4, HSB, YT and SUP-T1, but not in the T-cell lines HARRIS and HuT 78.

Q&A

What is the relationship between VIP and VIPR2 in neurological research?

VIPR2 (also known as VPAC2R) functions as a key receptor for Vasoactive Intestinal Peptide (VIP), playing crucial roles in neural signaling pathways. In neurological research, this receptor-ligand interaction is particularly important in the suprachiasmatic nucleus (SCN) where VPAC2R is strategically positioned for specialized synaptic communication from VIP neurons throughout the SCN . Understanding this relationship is essential when designing experiments targeting either the peptide or its receptor.

The relationship can be functionally validated through several approaches:

  • Pre-adsorption studies showing that VIP completely abolishes immunostaining with anti-VIP antibodies

  • Cross-reactivity testing demonstrating that related peptides (secretin, gastric inhibitory polypeptide, somatostatin, etc.) do not interfere with VIP immunolabeling

  • Genetic validation using knockout models (e.g., Vipr2−/− mice) which show background-level staining when probed with anti-VPAC2R antibodies

These validation methods confirm the specificity of both the peptide-receptor interaction and the antibodies used to study them.

How should I validate VIPR2 antibody specificity for my experimental design?

Proper validation of VIPR2 antibody specificity requires a multi-faceted approach:

  • Genetic validation: Compare staining between wild-type and Vipr2−/− (knockout) tissues. Authentic antibodies will show intense staining in wild-type samples and only background levels in knockout tissues .

  • Pre-adsorption controls: Pre-incubate your antibody with rat VIP at concentrations around 10^-5 M before application to your samples. Specific antibodies will show dramatically reduced or abolished staining after pre-adsorption . Commercial pre-adsorption controls are available (e.g., Sigma Aldrich, Catalog # V6130) .

  • Concentration optimization: Test different dilutions to determine optimal signal-to-noise ratio. For biotin-streptavidin/HRP systems with VIPR2/VPAC2R antibodies, effective dilutions are typically 1:15,000 for DAB visualization and 1:1,000 for immunofluorescence .

  • Cross-reactivity assessment: Verify that staining patterns are consistent with known VIPR2 expression patterns across different tissues and species, particularly in regions like the SCN where VIPR2 is well-characterized .

What are the key differences between VIPR2 antibodies and VIP antibodies in research applications?

While both antibodies target components of the same signaling system, they have distinct research applications:

CharacteristicVIPR2 AntibodiesVIP Antibodies
TargetMembrane-bound G-protein coupled receptor (VIPR2/VPAC2R)Secreted 28-amino acid neuropeptide
Cellular LocalizationPrimarily cell membrane, sometimes endosomesCell bodies, axons, and terminals of VIP-producing neurons
Knockout ControlsVipr2−/− miceVip−/− mice
Common Host SpeciesRabbit, MouseRabbit, Sheep
Typical ApplicationsReceptor mapping, signaling studiesPeptide localization, neural circuit mapping
Key Validation MethodPre-adsorption with VIP peptide (should not affect VIPR2 antibody staining)Pre-adsorption with VIP peptide (should abolish staining)

Both antibody types can be used complementarily to study different aspects of the VIP-VIPR2 signaling axis, with VIP antibodies revealing the source of the signal and VIPR2 antibodies identifying target cells .

What are the optimal protocols for immunodetection using biotin-conjugated VIPR2 antibodies?

When working with biotin-conjugated VIPR2 antibodies, protocol optimization is critical:

For ELISA applications:

  • Dilute concentrated biotin-conjugated VIPR2 antibody 1:100 with appropriate diluent before use .

  • Apply to wells pre-coated with capture antibody and containing bound VIPR2.

  • Incubate according to manufacturer recommendations (typically 1-2 hours at room temperature).

  • Wash thoroughly to remove unbound antibody.

  • Add streptavidin-HRP (commonly diluted 1:100 from concentrate) .

  • Follow with substrate solution and measure absorbance.

For Immunohistochemistry/Immunofluorescence:

  • Begin with paraformaldehyde-fixed tissues (commonly used for neural tissues).

  • Use biotin-conjugated VIPR2 antibodies at dilutions between 1:1,000-1:15,000 depending on visualization method .

  • For the biotin-streptavidin/HRP detection system, particularly successful results have been reported in rat amygdala, cortex, and suprachiasmatic nucleus at dilutions of 1:8,000-1:10,000 .

  • Include appropriate negative controls (knockout tissue or pre-adsorbed antibody) .

In both applications, preliminary titration experiments are essential to determine optimal antibody concentration for your specific tissue or sample type.

How does the biotin conjugation affect VIPR2 antibody performance compared to unconjugated versions?

Biotin conjugation provides several advantages but also introduces specific considerations:

Advantages:

  • Enhanced signal amplification through the biotin-streptavidin system

  • Flexibility in detection methods (can be paired with various streptavidin-conjugated reporters)

  • Improved sensitivity, particularly for low-abundance VIPR2 expression

  • Compatible with multiple immunodetection techniques including ELISA, IHC, and flow cytometry

Performance Considerations:

  • Biotin conjugation may affect antibody binding kinetics compared to unconjugated versions

  • Background can increase due to endogenous biotin in some tissues (liver, kidney)

  • May require blocking of endogenous biotin or peroxidase activity

  • Optimal dilution ranges typically differ from unconjugated antibodies (e.g., 1:8,000-1:10,000 for biotin-conjugated vs potentially different ranges for unconjugated)

For direct comparisons, it's advisable to test both conjugated and unconjugated versions on identical samples to determine which performs optimally for your specific application and tissue type.

What are the critical storage and handling considerations for maintaining biotin-conjugated VIPR2 antibody activity?

Proper storage and handling are essential for maintaining the activity of biotin-conjugated VIPR2 antibodies:

Storage Recommendations:

  • Unopened kits containing biotin-conjugated antibodies can typically be stored at 2-8°C for up to 1 year

  • Once opened, concentrated biotin-conjugated antibodies should be stored at 2-8°C and used within 1 month

  • Avoid repeated freeze-thaw cycles which can compromise antibody activity

  • Store antibody solutions away from strong light, which can affect biotin activity

Handling Considerations:

  • Some VIPR2 antibodies are provided as lyophilized whole serum containing 0.09% sodium azide . Reconstitute these according to manufacturer instructions

  • When diluting concentrated (e.g., 100×) biotin-conjugated antibodies, use only the recommended diluent

  • Prepare working dilutions shortly before use rather than storing diluted antibody for extended periods

  • For antibodies purified by Protein G (>95% purity), minimize exposure to proteases and bacterial contamination

Adherence to these storage and handling guidelines will help ensure consistent antibody performance across experiments and maximize shelf life.

How can I optimize detection in tissues with low VIPR2 expression?

Detecting low-abundance VIPR2 can be challenging but several methodological approaches can enhance sensitivity:

  • Signal Amplification Systems:

    • Utilize tyramide signal amplification (TSA) in conjunction with biotin-conjugated antibodies

    • Employ multistep detection with avidin-biotin complex (ABC) method followed by enhanced chemiluminescence

    • Consider sequential application of primary and secondary detection reagents with extended incubation times

  • Sample Preparation Optimization:

    • Evaluate different fixation protocols (paraformaldehyde concentration, duration)

    • Test various antigen retrieval methods (heat-induced vs. enzymatic)

    • Reduce background through careful blocking of endogenous biotin and peroxidase

  • Antibody Selection:

    • Choose antibodies targeting specific epitopes known to be accessible

    • For human samples, select antibodies recognizing amino acids 24-126 of VIPR2

    • Compare polyclonal vs. monoclonal options (polyclonals may provide better detection of low abundance targets)

  • Detection Enhancement Protocol:
    a) Use extended primary antibody incubation (overnight at 4°C)
    b) Increase streptavidin-HRP concentration slightly beyond standard recommendations
    c) Extend substrate development time while monitoring background
    d) Consider fluorescent detection with photomultiplier amplification for quantitative studies

These approaches should be systematically tested with appropriate positive and negative controls to determine the optimal protocol for your specific experimental system.

How should I address cross-reactivity concerns between VIPR1 and VIPR2 antibodies?

Distinguishing between the closely related VIPR1 and VIPR2 requires careful antibody selection and validation:

  • Epitope Selection:

    • Choose antibodies targeting regions with minimal sequence homology between VIPR1 and VIPR2

    • Antibodies recognizing the N-terminal region (AA 24-126) of VIPR2 are often more specific

    • Verify that the manufacturer has performed cross-reactivity testing against VIPR1

  • Validation Approaches:

    • Compare staining patterns with published distribution data (VIPR1 and VIPR2 have distinct tissue expression profiles)

    • Use tissues from Vipr1−/− or Vipr2−/− knockout models as negative controls

    • Perform pre-adsorption tests with both VIPR1 and VIPR2 peptides separately

  • Parallel Testing Protocol:
    a) Run serial sections probed with anti-VIPR1 and anti-VIPR2 antibodies
    b) Include appropriate positive control tissues known to express predominantly one receptor subtype
    c) Compare staining distribution and intensity patterns
    d) Confirm with alternative methods (e.g., in situ hybridization or RT-PCR) when possible

  • Western Blot Verification:

    • VIPR1 and VIPR2 have different molecular weights

    • Confirm antibody specificity by Western blot before immunohistochemical applications

    • Look for single bands at the appropriate molecular weight

Following these approaches will help ensure that your experimental results specifically reflect VIPR2 rather than VIPR1 or non-specific binding.

What are the most effective blocking strategies to reduce non-specific binding with biotin-conjugated VIPR2 antibodies?

Non-specific binding is a common challenge with biotin-conjugated antibodies but can be addressed through optimized blocking strategies:

  • Endogenous Biotin Blocking:

    • Pre-treat sections with avidin followed by biotin to saturate endogenous biotin

    • Commercial avidin/biotin blocking kits are available and effective

    • Critical for biotin-rich tissues (liver, kidney, brain regions)

  • Protein Blocking Optimization:

    • Test different blocking solutions:

      • 5-10% normal serum from the same species as the secondary antibody

      • 1-5% BSA in combination with 0.1-0.3% Triton X-100

      • Commercial blocking reagents formulated for biotin-streptavidin systems

    • Extend blocking time to 1-2 hours at room temperature for challenging tissues

  • Pre-absorption Protocol:

    • Pre-absorb the biotin-conjugated VIPR2 antibody with the target peptide at 10^-5 M concentration

    • This should abolish specific staining but not non-specific binding, helping to identify background

    • Include this as a negative control in each experiment

  • Additional Technical Considerations:

    • Use TBS rather than PBS for wash steps when using alkaline phosphatase detection systems

    • Include 0.05-0.1% Tween-20 in wash buffers to reduce hydrophobic interactions

    • Consider low-antibody diluent formulations that enhance signal-to-noise ratio

    • Ensure all incubation steps occur at consistent, controlled temperatures

These blocking strategies should be systematically evaluated and optimized for each new tissue type or experimental condition.

How should I design experiments to distinguish between VIPR2 expression levels and detection sensitivity?

Distinguishing true biological variation in VIPR2 expression from technical detection limitations requires careful experimental design:

  • Standard Curve Calibration:

    • For ELISA applications, establish a standard curve using recombinant VIPR2 protein at known concentrations

    • Include low concentration standards to define the lower limit of detection

    • Standard curve analysis should include tests for linearity and determine the dynamic range of detection

  • Internal Controls Implementation:

    • Include reference tissues with established VIPR2 expression levels in each experiment

    • Process all experimental and control samples simultaneously with identical protocols

    • Use housekeeping proteins for normalization in Western blot applications

  • Titration Series Design:

    • Test serial dilutions of your biotin-conjugated VIPR2 antibody

    • Plot the signal-to-noise ratio at each dilution

    • Determine the optimal concentration that balances sensitivity and specificity

    • For biotin-streptavidin/HRP systems, dilutions between 1:8,000-1:10,000 often provide optimal balance

  • Comparative Method Validation:

    • Validate immunohistochemistry findings with complementary methods:

      • qRT-PCR for VIPR2 mRNA expression

      • Receptor binding assays using labeled VIP

      • Western blot quantification of total protein

    • Discrepancies between methods can reveal post-transcriptional regulation or technical limitations

This systematic approach will help distinguish biological variations in VIPR2 expression from artifacts of detection methodology.

What controls are essential when using biotin-conjugated VIPR2 antibodies across different species?

When applying biotin-conjugated VIPR2 antibodies across different species, robust controls are essential:

  • Species Validation Controls:

    • Verify cross-reactivity with your species of interest

    • VIPR2 antibodies have documented reactivity with numerous species including human, rat, mouse, monkey, and other mammals

    • Include positive control tissues from well-characterized species alongside your experimental samples

  • Negative Controls Suite:

    • Include knockout tissues when available (Vipr2−/− mice are ideal)

    • Use pre-adsorption controls with VIPR2 peptide (10^-5 M concentration)

    • Process tissue sections with primary antibody omitted

    • Include isotype controls matching the primary antibody's host species and isotype

  • Cross-Species Validation Protocol:

    • Compare staining patterns with published distribution data for each species

    • Verify antibody performance in your species through Western blot before immunohistochemistry

    • Test antibody at multiple concentrations as optimal dilutions may vary between species

    • Consider sequence homology between species in the epitope region (AA 24-126 for many VIPR2 antibodies)

  • Technical Validation Across Species:

    • For each new species, confirm signal specificity through:

      • Peptide competition assays

      • Comparison of multiple antibodies targeting different VIPR2 epitopes

      • Parallel in situ hybridization for VIPR2 mRNA when possible

    • Document species-specific differences in optimal protocol conditions

These controls will ensure reliable interpretation of VIPR2 expression patterns across different species and minimize false-positive or false-negative results due to species differences.

How can I quantitatively analyze VIPR2 immunolabeling data for comparative studies?

For rigorous quantitative analysis of VIPR2 immunolabeling:

  • Image Acquisition Standardization:

    • Use identical acquisition parameters across all samples (exposure time, gain, offset)

    • Capture images at multiple magnifications for comprehensive analysis

    • Include calibration standards in each imaging session

    • Avoid areas with processing artifacts or uneven staining

  • Quantification Methodology Options:

    • For DAB-Based Immunohistochemistry:

      • Measure optical density using calibrated systems

      • Apply thresholding to distinguish positive cells from background

      • Quantify cell counts, staining intensity, and percent area stained

    • For Fluorescence-Based Detection:

      • Measure mean fluorescence intensity in defined regions of interest

      • Use colocalization analysis for double-labeling experiments

      • Apply background subtraction based on negative control samples

  • Data Normalization Approaches:

    • Normalize to internal references (adjacent regions, housekeeping proteins)

    • Include technical replicates to assess variability

    • Apply appropriate statistical methods for comparing expression levels:

      • ANOVA for multi-group comparisons

      • t-tests for two-group comparisons

      • Non-parametric alternatives when normality cannot be assumed

  • Validation Through Multiple Quantitative Methods:

    • Complement image analysis with biochemical quantification where possible

    • For ELISA-based quantification of VIPR2, generate standard curves with recombinant protein

    • Cross-validate image analysis with flow cytometry for single-cell suspensions

    • Compare protein quantification with mRNA expression data

This systematic approach to quantification will ensure reproducible and reliable comparative analysis of VIPR2 expression across experimental conditions or biological systems.

How can biotin-conjugated VIPR2 antibodies be integrated into multiplexed immunolabeling strategies?

Multiplexed immunolabeling with biotin-conjugated VIPR2 antibodies enables complex spatial relationship studies:

  • Sequential Multiplexing Approaches:

    • Design antibody panels from different host species to avoid cross-reactivity

    • When using multiple biotin-conjugated antibodies, employ sequential detection with stripping steps between rounds

    • Consider tyramide signal amplification for multiplexed fluorescence applications

    • Use spectral unmixing to resolve overlapping fluorescent signals

  • Multiplex Protocol Optimization:

    • Begin with validated antibody pairs known to be compatible

    • Test for potential crossreactivity between detection systems

    • Optimize order of antibody application (typically starting with lowest abundance target)

    • Include appropriate single-stain controls for each antibody in the panel

  • Application in SCN Circadian Rhythm Research:

    • Combine biotin-conjugated VIPR2 antibodies with markers for:

      • Other neuropeptide receptors

      • Clock genes (Per, Cry, Bmal1)

      • Neuronal activation markers

    • This approach can reveal the specialized synaptic communication from VIP neurons in the ventral SCN to cells throughout the SCN at different times of day

  • Technical Considerations for Successful Multiplexing:

    • Carefully order detection steps to prevent cross-reaction

    • For brightfield applications, use different chromogens for each target

    • For fluorescence, select fluorophores with minimal spectral overlap

    • Consider using biotin-conjugated VIPR2 antibody for the most challenging (lowest abundance) target

These multiplexed approaches enable comprehensive spatial and functional mapping of VIPR2 in relation to other signaling components in complex tissues.

What are the emerging applications of VIPR2 antibodies in understanding neurological disorders?

Biotin-conjugated VIPR2 antibodies are becoming valuable tools in neurological disorder research:

  • Circadian Rhythm Disorders:

    • VIPR2/VPAC2R is essential for normal circadian rhythm function

    • Antibodies enable mapping of receptor distribution in the SCN under normal and pathological conditions

    • Studies show VPAC2R is "well positioned for specialized synaptic communication from VIP neurons in the ventral SCN to cells throughout the SCN at all times of day"

    • Changes in VIPR2 expression may correlate with sleep disorders and seasonal affective disorder

  • Neurodevelopmental Disorders:

    • Genetic studies have linked VIPR2 gene copy number variations to schizophrenia risk

    • Immunohistochemical studies can reveal altered VIPR2 expression patterns in developmental disorders

    • Biotin-conjugated antibodies allow precise cellular localization in human postmortem tissue

  • Neurodegenerative Diseases:

    • VIP signaling through VIPR2 has neuroprotective properties

    • Studies can map changes in receptor distribution during disease progression

    • Quantitative analysis of VIPR2 expression may identify vulnerable neuronal populations

  • Future Research Directions:

    • Integration with single-cell transcriptomics

    • Correlation of VIPR2 expression with circuit-level dysfunction

    • Development of receptor-targeted therapeutics based on expression mapping

    • Study of VIPR2 trafficking and signaling dynamics using live-cell imaging

These emerging applications highlight the importance of specific and sensitive VIPR2 antibodies in understanding the role of VIP signaling in neurological health and disease.

How can researcher-optimized protocols for biotin-conjugated VIPR2 antibodies be validated and shared with the scientific community?

Standardizing and sharing optimized protocols benefits the broader scientific community:

  • Comprehensive Validation Framework:

    • Document antibody performance across multiple applications (WB, IHC, IF, ELISA)

    • Test specificity using genetic models (Vipr2−/− tissue)

    • Compare results with multiple antibodies targeting different VIPR2 epitopes

    • Validate across species when possible

    • Include Research Resource Identifiers (RRIDs) for antibodies in publications (e.g., AB_572270)

  • Protocol Documentation Best Practices:

    • Provide detailed methods including:

      • Fixation conditions

      • Antigen retrieval parameters

      • Blocking composition and duration

      • Primary antibody dilution, incubation time and temperature

      • Detection system specifications

      • Imaging parameters

    • Include representative images of positive and negative controls

    • Document antibody lot numbers and sourcing information

  • Community Sharing Mechanisms:

    • Publish detailed methods in peer-reviewed journals

    • Deposit protocols in repositories like protocols.io

    • Share troubleshooting tips in methods-focused publications

    • Include protocol variations for different applications

    • Establish standardized reporting guidelines specific to VIPR2 detection

  • Collaborative Validation Approach:

    • Organize multi-laboratory validation studies

    • Develop consensus protocols for specific applications

    • Create reference materials for interlaboratory standardization

    • Establish online communities for protocol optimization discussions

These approaches to validation and sharing will accelerate research progress by reducing duplication of methodological optimization efforts and enhancing reproducibility across laboratories.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.