GPR55 Antibody, Biotin conjugated

Shipped with Ice Packs
In Stock

Description

Structure and Target Specificity

GPR55 is a 319-amino-acid receptor with low homology to classical cannabinoid receptors (CB1R/CB2R) and is expressed in the CNS, immune cells, and bone marrow . The biotin-conjugated GPR55 antibody typically targets specific epitopes:

  • Antigenic Regions: Commonly raised against sequences in the extracellular loops (e.g., residues 141–240, 160–174, or 210–319) .

  • Host and Clonality: Produced in rabbits as polyclonal antibodies, ensuring broad epitope recognition .

  • Conjugate: Covalently linked to biotin, enabling streptavidin-based amplification in assays .

Cancer Biology

  • Mechanistic Studies: GPR55 promotes cancer cell proliferation and metastasis. Biotin-conjugated antibodies have been used to validate receptor expression in B-lymphoblastoid cell lines (EHEB, DeFew) and glioblastoma models .

  • Therapeutic Targeting: Peptide ligands (e.g., FITC-P1) conjugated to fluorophores or biotin inhibit GPR55 signaling, reducing tumor growth in vitro .

Neurological Research

  • Axon Guidance: GPR55 regulates growth cone dynamics in retinal development, with antibody staining confirming its localization in neuronal tissues .

  • Pain Pathways: Linked to hyperalgesia, GPR55 knockdown via siRNA reduces receptor expression, as quantified using biotinylated probes .

Signal Transduction

  • Calcium Signaling: GPR55 activation by lysophosphatidylinositol (LPI) induces Ca²⁺ release, detectable via biotin-streptavidin ELISA systems .

  • Protein Interactions: Co-immunoprecipitation studies using HA-tagged GPR55 and biotinylated antibodies identified interactions with Gα13 and RhoA .

Validation and Quality Control

  • Specificity: Antibodies are validated using siRNA-mediated GPR55 knockdown, showing reduced binding in HeLa cells .

  • Deglycosylation Assays: Western blotting confirms antibody recognition of both glycosylated (~50 kDa) and deglycosylated (37 kDa) GPR55 forms .

  • Cross-Reactivity: Minimal non-specific binding to unrelated receptors (e.g., CB1R/CB2R) .

Critical Considerations

  • Sensitivity: Biotin conjugation enhances detection limits in low-abundance samples (e.g., bone marrow or tumor biopsies) .

  • Hazard Notes: Contains ProClin 300, a preservative requiring careful handling .

  • Batch Variability: Polyclonal nature may lead to inter-lot differences in epitope recognition .

Recent Advances

  • Peptide-Guided Therapy: Biotinylated peptides like P1 (20 µM affinity) induce GPR55 internalization, offering a strategy to block oncogenic signaling .

  • Multi-Omics Integration: Paired with CRISPR screens, these antibodies help map GPR55’s role in metabolic diseases and neuropathic pain .

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 receiving it. Delivery times may vary depending on the shipping method or location. Please consult your local distributor for specific delivery time information.
Synonyms
G protein coupled receptor 55 antibody; G-protein coupled receptor 55 antibody; GPCR GPR55 antibody; GPR 55 antibody; Gpr55 antibody; GPR55_HUMAN antibody; LPIR1 antibody; Probable G protein coupled receptor 55 antibody
Target Names
Uniprot No.

Target Background

Function
GPR55 is a receptor involved in various biological processes, including the regulation of pain perception, calcium signaling, and bone physiology. It plays a crucial role in hyperalgesia associated with inflammatory and neuropathic pain. GPR55 serves as a receptor for L-alpha-lysophosphatidylinositol (LPI), a lipid that induces the release of calcium from intracellular stores via the G protein GNA13 and RHOA signaling pathways. Additionally, GPR55 is considered a putative cannabinoid receptor, suggesting its involvement in the endocannabinoid system. Furthermore, GPR55 is implicated in regulating bone physiology by influencing the number and function of osteoclasts, cells responsible for bone resorption.
Gene References Into Functions
  1. GPR55 plays a role in the migration of human breast cancer cells. PMID: 29188802
  2. Inhibiting GPR55 activity can have antitumor effects by attenuating the MEK/ERK and PI3K-AKT signaling pathways, leading to a decrease in the expression and function of multidrug resistance proteins. PMID: 27423937
  3. Research suggests that GPR55 acts as an oncogene, while CB1 acts as a tumor suppressor in colon carcinogenesis. PMID: 28875496
  4. GPR55 is highly expressed in both human and mouse pancreatic beta-cells. It is also present in a smaller proportion of mouse islet alpha-cells and a majority of human islet alpha-cells. However, it is absent or present at very low levels in delta-cells. Notably, GPR55 is preferentially expressed in the endocrine pancreas, with only faint immunoreactivity in the exocrine pancreas. PMID: 27561953
  5. Studies have shown significantly higher expression of GPR55 mRNA in patients with Crohn's disease (CD) compared to ulcerative colitis (UC) and healthy controls. Furthermore, CD patients exhibit higher GPR55 mRNA expression in inflamed compared to non-inflamed colonic tissues. In contrast, GPR55 mRNA expression in UC patients does not show statistically significant differences between inflamed and non-inflamed colonic tissue. PMID: 28272905
  6. Crucial amino acid residues involved in agonist signaling at the GPR55 receptor have been identified. PMID: 28005346
  7. Signaling pathways activated by lysophosphatidylinositol through its receptor GPR55 play a pivotal role in different types of cancer. PMID: 26588872
  8. GPR55 is involved in the migratory behavior of colon carcinoma cells. PMID: 26436760
  9. Inhibition of the pro-angiogenic L-alpha-lysophosphatidylinositol/GPR55 pathway appears to be a promising therapeutic target against angiogenesis in ovarian carcinoma. PMID: 25989290
  10. GPR55 is expressed in the placenta, specifically at the placental endothelium. L-alpha-lysophosphatidylinositol (LPI), the endogenous ligand of GPR55, increases migratory activity of venous but not arterial placental endothelial cells, suggesting a role for the LPI-GPR55 axis in placental venous endothelium function. PMID: 25869640
  11. GPR55 may play a detrimental role in ox-LDL-induced foam cells and could be a potential pharmacological target for managing atherosclerosis and other cardiovascular diseases. PMID: 25970609
  12. GPR55 has been identified as a direct target gene of miR-675-5p. PMID: 25889562
  13. GPR55 plays a proinflammatory role in innate immunity. PMID: 25344934
  14. Expression of CB1 and GPR55 in proximal tubules is altered in response to elevated levels of glucose and albumin. PMID: 25545857
  15. This review provides an overview of the current understanding of GPR55 expression and function in tissues involved in metabolic regulation and the signaling cascades through which GPR55 is reported to act. PMID: 24972076
  16. Heteromerization of GPR55 and cannabinoid CB2 receptors modulates signaling. PMID: 25048571
  17. Research suggests that GPR55, ABCC1 (ATP-binding cassette sub-family C), and MPR1/ABCB1 (multidrug resistance protein 1) participate in autocrine communication and tumorigenesis involving lysophosphatidylinositol. PMID: 25233417
  18. CB2R and GPR55 form heteromers in cancer cells. These structures possess unique signaling properties, and modulation of these heteromers can modify the antitumoral activity of cannabinoids in vivo. PMID: 24942731
  19. GPR55 receptors are expressed in urothelial cell lines and interact with CB1 receptors. PMID: 24652077
  20. GPR55 antagonists occupy a horizontal binding pocket extending into the extracellular loop region, while a central ligand portion fits vertically in the receptor binding pocket. PMID: 24274581
  21. Data suggest that GPR55 is strongly expressed on myenteric neurons of the colon. PMID: 23603203
  22. GPR55 is functionally expressed in vascular endothelium and platelets and is involved in the regulation of calcium signaling. Lysophosphatidylinositol is a ligand/agonist for GPR55. PMID: 23639801
  23. Studies have investigated the expression levels of CB1R, CB2R, and GPR55 in HIV-infected human cells injected into immunodeficient mice. PMID: 20549374
  24. GPR55 drives skin carcinogenesis and is upregulated in human squamous cell carcinomas. PMID: 22751111
  25. This article reviews current data about GPR55 pharmacology and signaling, highlighting its involvement in various pathophysiological conditions. PMID: 23151004
  26. This review summarizes current knowledge about the G-protein coupled receptors GPR35 and GPR55 and their potential characterization as lysophospholipid or cannabinoid receptors, respectively. PMID: 22820167
  27. CB1 modulates the signaling properties of the lysophosphatidylinositol receptor GPR55. PMID: 23161546
  28. Research suggests that the lysophosphatidylinositol (LPI)/orphan G protein-coupled receptor GPR55 axis plays a significant role in various physiological and pathological contexts. PMID: 22285325
  29. GASP-1 is a key regulator of the trafficking and functional expression of GPR55. PMID: 21718301
  30. The alpha-lysophosphatidylinositol(LPI)/GPR55 system is positively associated with obesity in humans. PMID: 22179809
  31. This article explores the partnership between GPR55 and CB2. PMID: 21537344
  32. GPR55 limits tissue-injuring inflammatory responses mediated by CB(2)R, while it synergizes with CB(2)R in recruiting neutrophils to sites of inflammation. PMID: 21467997
  33. GPR55 is expressed in human tumors and drives proliferation. Its expression correlates with tumor aggressiveness. PMID: 21367464
  34. Three lead compounds, combined with the identification of key GPR55 receptor interaction sites, provide a foundation for the design of more efficacious second-generation GPR55 ligands that retain selectivity for GPR55. PMID: 21534610
  35. Studies have shown that the low-functioning Val195 allele of GPR55 is a risk factor for anorexia nervosa. PMID: 20506567
  36. GPR55 expression in human tumors from different origins correlates with tumor aggressiveness. GPR55 promotes cancer cell proliferation through the overactivation of the extracellular signal-regulated kinase cascade. PMID: 20818416
  37. Results suggest that GPR55 and its endogenous ligand LPI play essential roles in the homeostatic responses to stress signals in various mammalian tissues and cells, including certain types of immune cells. PMID: 20051382
  38. These findings strongly suggest that GPR55 is a specific and functional receptor for lysophosphatidylinositol. PMID: 17765871
  39. GPR55 is a novel cannabinoid receptor that binds to and is activated by the cannabinoid ligand CP55940 and couples to Galpha13. It can mediate the activation of rhoA, cdc42, and rac1. PMID: 17876302
  40. Research establishes GPR55 as a cannabinoid receptor with signaling distinct from CB(1) and CB(2). PMID: 18263732
  41. In endothelial cells, two receptors for anandamide have been identified: cannabinoid 1 receptor and G-protein-coupled receptor 55. Integrin clustering enables anandamide-induced Ca2+ signaling in endothelial cells via GPR55. PMID: 18445684
  42. Treatment with lysophosphatidylinositol induces marked GPR55 internalization and stimulates a sustained, oscillatory calcium release pathway that is dependent on Galpha13 and requires RhoA activation. PMID: 18757503
  43. GPR55 is an atypical cannabinoid responder. PMID: 19723626
  44. Research reveals a role for GPR55 in bone physiology by regulating osteoclast number and function. PMID: 19805329

Show More

Hide All

Database Links

HGNC: 4511

OMIM: 604107

KEGG: hsa:9290

STRING: 9606.ENSP00000375893

UniGene: Hs.114545

Protein Families
G-protein coupled receptor 1 family
Subcellular Location
Cell membrane; Multi-pass membrane protein.
Tissue Specificity
Expressed in the caudate nucleus and putamen, but not detected in the hippocampus, thalamus, pons cerebellum, frontal cortex of the brain or in the liver. Expressed in osteoclasts and osteoblasts.

Q&A

What is GPR55 and why is it important in research?

GPR55 is a G-protein-coupled receptor encoded by the GPR55 gene (Gene ID: 9290, Swiss Prot: Q9Y2T6). It functions as a receptor for L-alpha-lysophosphatidylinositol (LPI), which induces Ca²⁺ release from intracellular stores via the heterotrimeric G protein GNA13 and RHOA. GPR55 may be involved in hyperalgesia associated with inflammatory and neuropathic pain. It also plays a significant role in bone physiology by regulating osteoclast number and function, and in neurodevelopment by modulating axon growth and target innervation . As a putative cannabinoid receptor, GPR55 is relevant to studies investigating endocannabinoid signaling systems and potential therapeutic applications.

What is a biotin-conjugated GPR55 antibody?

A biotin-conjugated GPR55 antibody is an immunoglobulin that specifically recognizes and binds to the GPR55 protein and has been chemically linked to biotin molecules. This biotin conjugation enhances the antibody's utility in various detection systems due to the strong affinity between biotin and streptavidin/avidin. Commonly available GPR55 biotin-conjugated antibodies are polyclonal, derived from rabbits immunized with synthetic peptides corresponding to specific amino acid sequences of human GPR55 (such as regions 141-240/319 or 203-222) . This conjugation allows for amplified signal detection and versatility in experimental applications.

What are the primary applications for GPR55 biotin-conjugated antibodies?

GPR55 biotin-conjugated antibodies are primarily used in:

  • Western Blot (WB): To detect and quantify GPR55 protein expression in tissue or cell lysates, typically at dilutions between 1:300-1:5000 .

  • ELISA (Enzyme-Linked Immunosorbent Assay): For quantitative determination of GPR55 in solution, typically at dilutions between 1:500-1:1000 .

  • Immunohistochemistry (IHC): To visualize GPR55 expression in tissue sections when used with appropriate detection systems.

  • Immunofluorescence: For cellular localization studies, particularly when investigating GPR55's subcellular distribution (primarily in cell membranes) .

The biotin conjugation provides amplification advantages through secondary detection with streptavidin-conjugated reporter molecules.

What tissue/cell types are known to express GPR55?

GPR55 has been detected in various tissues and cell types:

  • Central nervous system: Particularly in the retina during development where it regulates growth cone morphology and axon growth .

  • Bone marrow: Where it may influence osteoclast function and bone remodeling .

  • Immune cells: Contributing to inflammatory responses.

  • Endocrine tissues: With potential roles in metabolic regulation.

  • Cancer cells: With emerging evidence for involvement in cancer cell proliferation and migration.

When designing experiments with GPR55 antibodies, researchers should consider that expression levels may vary significantly between tissues, developmental stages, and pathological conditions.

How do GPR55 agonists and antagonists affect antibody binding and experimental outcomes?

The presence of GPR55 ligands in experimental systems can significantly influence antibody binding characteristics and experimental outcomes. When GPR55 binds agonists like lysophosphatidylinositol (LPI) or O-1602, the receptor undergoes conformational changes that can potentially mask or expose epitopes recognized by antibodies . Antagonists like cannabidiol (CBD) or ML192 derivatives may stabilize the receptor in an inactive conformation.

In immunodetection experiments:

  • Epitope accessibility may be altered depending on receptor activation state

  • Pre-treatment of samples with ligands might require adjusted antibody concentrations

  • Fixation methods can differentially preserve ligand-induced conformations

For functional studies, researchers should consider whether antibody binding itself might have agonistic or antagonistic effects on receptor function. Control experiments comparing antibody binding in the presence and absence of known ligands can help interpret complex results, especially in live cell applications.

What are the implications of GPR55 knockout models for antibody validation?

GPR55 knockout (gpr55-/-) models provide crucial controls for antibody validation and specificity determination. Research shows that neurons from gpr55-/- mouse embryos exhibit smaller growth cones, fewer growth cone filopodia, and decreased axonal outgrowth compared to wild-type neurons . These models offer several advantages for antibody validation:

  • True negative controls: Tissue from knockout animals should show no specific staining, allowing identification of non-specific binding.

  • Validation of developmental phenotypes: GPR55-/- mice show decreased branching in the dorsal terminal nucleus and lower levels of eye-specific segregation of retinal projections in the superior colliculus and dorsal lateral geniculate nucleus .

  • Ligand response verification: In absence of GPR55, pharmacological effects of GPR55 ligands (LPI, O-1602, CBD) are not observed, confirming antibody specificity .

When validating biotin-conjugated GPR55 antibodies, comparing staining patterns between wild-type and knockout tissues across multiple detection methods (WB, IHC, IF) provides comprehensive evidence of specificity and appropriate experimental conditions.

How do post-translational modifications of GPR55 affect antibody recognition?

GPR55, like other GPCRs, undergoes various post-translational modifications (PTMs) that can affect antibody recognition. Although specific data on GPR55 PTMs is limited in the provided search results, general principles apply:

  • Phosphorylation: Occurs during receptor desensitization and may alter epitope accessibility, particularly for antibodies targeting intracellular domains.

  • Glycosylation: GPR55 contains potential N-glycosylation sites in extracellular domains that might affect antibody binding to these regions.

  • Palmitoylation: May influence receptor trafficking and membrane localization, potentially affecting antibody accessibility in intact cells.

  • Ubiquitination: Can signal receptor degradation and may affect detection in protein turnover studies.

When designing experiments, researchers should consider:

  • Using multiple antibodies targeting different epitopes to ensure comprehensive detection

  • Comparing native versus denatured conditions to assess conformational epitope recognition

  • Employing enzymatic treatments (phosphatases, glycosidases) to evaluate PTM effects on detection

  • Validating results across different cell types where PTM patterns may vary

What are the optimal protocols for using biotin-conjugated GPR55 antibodies in Western blotting?

For optimal Western blot results with biotin-conjugated GPR55 antibodies, researchers should consider the following protocol recommendations:

Sample Preparation:

  • Extract proteins using buffers containing protease inhibitors to prevent GPR55 degradation

  • For membrane proteins like GPR55, include gentle detergents (0.5-1% Triton X-100 or CHAPS)

  • Heat samples at 37°C instead of boiling to prevent aggregation of membrane proteins

Electrophoresis and Transfer:

  • Use 10-12% polyacrylamide gels for optimal resolution of GPR55 (~37 kDa)

  • Transfer to PVDF membranes (preferred over nitrocellulose for hydrophobic proteins)

  • Verify transfer efficiency with reversible staining

Immunodetection:

  • Block with 3-5% BSA in TBS with 0.1% Tween-20 (BSA is preferred over milk for biotin-conjugated antibodies)

  • Dilute primary antibody 1:300-1:5000 in blocking buffer

  • Incubate overnight at 4°C for highest sensitivity

  • Use streptavidin-HRP (1:2000-1:10000) for detection

  • Develop using enhanced chemiluminescence

Controls:

  • Include GPR55-overexpressing positive control

  • Use GPR55 knockout or knockdown samples as negative controls

  • Consider peptide competition assays to confirm specificity

This approach maximizes detection sensitivity while minimizing background and non-specific binding commonly encountered with membrane proteins like GPR55.

How can biotin-conjugated GPR55 antibodies be utilized in colocalization studies?

Biotin-conjugated GPR55 antibodies are valuable tools for colocalization studies examining GPR55's spatial relationship with other proteins. The following methodology optimizes their use:

Sample Preparation:

  • Fix cells/tissues with 4% paraformaldehyde (10-15 minutes)

  • Permeabilize with 0.1-0.3% Triton X-100 for intracellular epitopes

  • For membrane preservation, consider milder detergents like 0.1% saponin

Staining Protocol:

  • Block with 5% normal serum and 1% BSA

  • Apply biotin-conjugated GPR55 antibody (typically 2-5 μg/ml)

  • For colocalization, simultaneously or sequentially apply unconjugated primary antibodies against proteins of interest

  • Detect GPR55 using streptavidin conjugated to a fluorophore spectrally distinct from secondary antibodies used for other targets

  • Include DAPI for nuclear counterstaining

Imaging Considerations:

  • Use confocal microscopy to minimize out-of-focus fluorescence

  • Capture sequential scans to prevent crosstalk between channels

  • Apply appropriate controls for spectral bleed-through

  • Consider super-resolution techniques for detailed subcellular localization

Quantitative Analysis:

  • Calculate Pearson's or Mander's coefficients for quantitative colocalization assessment

  • Perform line scan analysis across cellular regions to confirm spatial relationships

  • Consider pixel-by-pixel intensity correlation analysis

This approach enables precise determination of GPR55's spatial relationship with potential interacting partners, signaling components, or cellular compartments.

What is the optimal approach for using GPR55 biotin-conjugated antibodies in flow cytometry?

For optimal flow cytometry results with biotin-conjugated GPR55 antibodies, implement the following protocol:

Cell Preparation:

  • Harvest cells using enzyme-free dissociation methods to preserve membrane proteins

  • Maintain viability above 95% for reliable results

  • Adjust to 1×10^6 cells per sample

Surface Staining Protocol:

  • Block with 2% FBS in PBS for 15 minutes

  • Apply biotin-conjugated GPR55 antibody (1-5 μg/ml) for 30 minutes at 4°C

  • Wash twice with cold PBS containing 2% FBS

  • Incubate with streptavidin-fluorophore conjugate (e.g., streptavidin-PE, APC, or Alexa Fluor dyes)

Intracellular Staining Protocol (if needed):

  • Fix cells with 2-4% paraformaldehyde for 10 minutes

  • Permeabilize with 0.1% saponin or commercially available permeabilization buffers

  • Proceed with blocking and staining as above

Controls:

  • Include unstained cells for autofluorescence assessment

  • Use isotype-biotin controls followed by streptavidin-fluorophore

  • Include secondary-only controls (streptavidin-fluorophore only)

  • Use cells with known GPR55 expression levels as positive controls

  • When possible, include GPR55 knockout or knockdown cells as negative controls

Analysis Considerations:

  • Gate on viable, single cells

  • Compare signal to appropriate negative controls

  • Consider compensation if using multiple fluorophores

This approach enables quantitative assessment of GPR55 expression across cell populations, facilitating studies of expression dynamics under various conditions or treatments.

How can researchers address non-specific binding issues with biotin-conjugated GPR55 antibodies?

Non-specific binding is a common challenge with biotin-conjugated antibodies. For GPR55 detection, consider these strategies:

Sources of Non-specific Binding:

  • Endogenous biotin in samples (particularly in brain, liver, and kidney tissues)

  • Cross-reactivity with structurally similar GPCRs

  • Fc receptor interactions in immune cells

  • Hydrophobic interactions with membrane proteins

Optimization Strategies:

  • Biotin Blocking:

    • Pretreat samples with avidin/biotin blocking kits

    • Include free biotin (10-100 μg/ml) in blocking buffers

  • Blocking Optimization:

    • Use 3-5% BSA instead of milk (which contains endogenous biotin)

    • Add 0.1-0.3% Triton X-100 to reduce hydrophobic interactions

    • Include 5-10% serum from the same species as secondary reagents

  • Antibody Dilution:

    • Perform careful titration experiments to determine optimal concentration

    • For GPR55 biotin-conjugated antibodies, start with manufacturer recommendations (1:300-1:5000 for WB, 1:500-1000 for ELISA)

  • Stringency Adjustment:

    • Increase salt concentration (150-500 mM NaCl) in wash buffers

    • Add 0.05-0.1% SDS to wash buffers for Western blotting

    • Increase number and duration of washes

  • Validation Controls:

    • Use peptide competition assays with the immunizing peptide

    • Include GPR55 knockout or knockdown samples

    • Compare multiple antibodies targeting different GPR55 epitopes

By systematically implementing these strategies, researchers can significantly reduce non-specific binding while maintaining sensitivity for specific GPR55 detection.

What are the considerations for detecting GPR55 in different species using biotin-conjugated antibodies?

When extending GPR55 detection across species, researchers must carefully consider antibody specificity and epitope conservation:

Species Reactivity Analysis:

  • Available biotin-conjugated GPR55 antibodies show confirmed reactivity with human GPR55

  • Predicted reactivity extends to mouse, rat, dog, horse, chicken, and rabbit for some antibodies

  • Epitope sequence conservation should be verified by sequence alignment

Cross-Species Validation Approach:

  • Sequence Comparison:

    • Align the immunogen sequence (e.g., human GPR55 amino acids 141-240 or 203-222) with target species

    • Calculate percent identity and similarity in the epitope region

    • Identify potential species-specific post-translational modifications

  • Positive Controls:

    • Use tissues with known GPR55 expression from the target species

    • Consider transfection controls with species-specific GPR55 constructs

    • Compare detection patterns between human and target species samples

  • Knockout Validation:

    • When available, use tissues from GPR55 knockout animals of the target species

    • Compare staining patterns between wild-type and knockout samples

  • Optimization for Each Species:

    • Adjust antibody concentration (generally higher for less conserved epitopes)

    • Modify fixation protocols based on tissue characteristics

    • Adapt blocking conditions to minimize species-specific background

  • Detection System Considerations:

    • Ensure secondary detection reagents (streptavidin conjugates) lack cross-reactivity with target species proteins

    • Consider endogenous biotin levels which vary between species and tissues

By following this systematic approach, researchers can confidently extend GPR55 studies across species while maintaining experimental rigor and reproducibility.

How do different fixation and permeabilization methods affect GPR55 antibody detection?

Fixation and permeabilization protocols significantly impact GPR55 detection due to its membrane localization and conformation-sensitive epitopes:

Fixation Effects:

  • Paraformaldehyde (PFA) Fixation:

    • 2-4% PFA generally preserves GPR55 epitopes while maintaining cellular architecture

    • Shorter fixation times (10-15 minutes) minimize epitope masking

    • Preferred for immunofluorescence and flow cytometry applications

  • Methanol Fixation:

    • Can improve detection of some intracellular epitopes

    • May disrupt membrane structure and alter conformational epitopes

    • Test side-by-side with PFA for specific antibody clones

  • Glutaraldehyde:

    • Provides superior ultrastructural preservation

    • Often causes excessive cross-linking that masks epitopes

    • If needed, use at low concentrations (0.1-0.5%) in combination with PFA

Permeabilization Considerations:

  • For Transmembrane and Extracellular Epitopes:

    • Gentle permeabilization or no permeabilization is preferred

    • Digitonin (0.001-0.01%) selectively permeabilizes plasma membrane

    • Saponin (0.1-0.3%) creates small pores while preserving membrane structure

  • For Intracellular Epitopes:

    • Triton X-100 (0.1-0.3%) provides reliable access to intracellular domains

    • Consider fixation-permeabilization sequence (simultaneous vs. sequential)

Optimization Strategy:

  • Test matrix of conditions combining:

    • Fixation agent and concentration

    • Fixation duration and temperature

    • Permeabilization method and timing

    • Antigen retrieval options (heat, pH, enzymatic)

  • Evaluate results based on:

    • Signal intensity at expected molecular weight/location

    • Background levels and non-specific binding

    • Morphological preservation of subcellular structures

These considerations are particularly important when studying GPR55 conformation changes induced by ligands like LPI, O-1602, or CBD, as described in neurodevelopmental studies .

How can biotin-conjugated GPR55 antibodies be used to study GPR55 agonist and antagonist mechanisms?

Biotin-conjugated GPR55 antibodies provide valuable tools for investigating ligand interactions and receptor responses:

Receptor Internalization Assays:

  • Track GPR55 redistribution following agonist treatment (LPI, O-1602)

  • Compare surface vs. internal receptor pools using non-permeabilized and permeabilized conditions

  • Quantify internalization kinetics through time-course experiments

Conformational Change Detection:

  • Some antibodies may preferentially recognize active or inactive receptor conformations

  • Compare antibody binding before and after treatment with agonists (LPI) versus antagonists (CBD, ML192 derivatives)

  • Use proximity ligation assays to detect changes in protein-protein interactions following ligand binding

Ligand Screening Applications:

  • Develop competition binding assays using biotin-conjugated antibodies

  • Measure displacement of antibody binding by potential ligands

  • Compare results with functional assays (β-arrestin recruitment, calcium mobilization)

Data Analysis Considerations:

  • Account for different efficacies of ligands (LPI vs. O-1602)

  • Consider biased signaling where ligands preferentially activate different pathways

  • Compare data with results from thienopyrimidine derivatives and other GPR55 antagonists

This approach enables detailed mechanistic studies of GPR55 pharmacology, potentially identifying novel ligands with therapeutic applications in inflammatory pain, cancer, or neurodevelopmental disorders.

What insights can GPR55 antibody studies provide about its role in neurodevelopment?

GPR55 antibody-based studies offer critical insights into neurodevelopmental processes as evidenced by research on retinal ganglion cell projections:

Developmental Expression Patterns:

  • Biotin-conjugated GPR55 antibodies can map temporal and spatial expression throughout neurodevelopment

  • Compare expression between wild-type and GPR55 knockout models to correlate with phenotypic differences

  • Analyze co-expression with developmental markers and axon guidance molecules

Growth Cone Dynamics:

  • Studies show GPR55 regulates growth cone morphology and filopodia formation

  • Antibody labeling can visualize receptor distribution within growth cone structures

  • Live imaging with minimally disruptive labeling techniques can track receptor dynamics during axon extension

Target Innervation Analysis:

  • GPR55 knockout mice show decreased branching in the dorsal terminal nucleus (DTN)

  • Antibody labeling can quantify receptor levels at key decision points during innervation

  • Combined with anterograde tracing, can correlate receptor expression with branching patterns

Mechanistic Insights:

  • Antibody blocking experiments can complement pharmacological approaches

  • Phospho-specific antibodies (if available) can track activation of downstream signaling pathways

  • Co-immunoprecipitation with biotin-conjugated antibodies can identify developmental binding partners

These approaches have revealed that GPR55 plays important roles in eye-specific segregation of retinal projections in the superior colliculus and dorsal lateral geniculate nucleus, suggesting broader implications for activity-dependent refinement of neural circuits .

How can GPR55 biotin-conjugated antibodies be used in multiplex detection systems?

Biotin conjugation makes GPR55 antibodies particularly suitable for multiplex detection strategies:

Sequential Multiplex Immunohistochemistry:

  • Use biotin-conjugated GPR55 antibodies as one component in multiplexed tissue staining

  • Apply tyramide signal amplification for signal enhancement and subsequent antibody stripping

  • Cycle through multiple rounds of staining to detect 5-10 proteins on the same section

  • Useful for characterizing GPR55-expressing cell populations in complex tissues

Mass Cytometry (CyTOF) Applications:

  • Conjugate GPR55 antibodies to metal isotopes via biotin-streptavidin bridges

  • Integrate into panels of 30+ markers for deep cellular phenotyping

  • Quantify GPR55 expression across diverse cell populations simultaneously

Proximity-Based Detection:

  • Pair biotin-conjugated GPR55 antibodies with antibodies against potential interacting proteins

  • Apply proximity ligation assay (PLA) to visualize interactions as fluorescent puncta

  • Quantify interaction dynamics under different ligand conditions (LPI, CBD)

Bead-Based Multiplex Assays:

  • Couple streptavidin-coated beads with biotin-conjugated GPR55 antibodies

  • Develop pull-down assays to capture GPR55 and associated proteins

  • Analyze using flow cytometry or imaging cytometry platforms

Spatial Transcriptomics Integration:

  • Combine antibody detection with in situ hybridization techniques

  • Correlate protein expression with mRNA localization

  • Map receptor distribution across tissue sections with transcriptional profiles

This multiplexing capability enables comprehensive characterization of GPR55 in complex biological systems, providing insights into its diverse functions across different cellular contexts and disease states.

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.