GPR116 (also known as ADGRF5, adhesion G protein-coupled receptor F5) is a member of the adhesion G protein-coupled receptor family involved in cell surface receptor signaling pathways. It plays critical roles in surfactant homeostasis in lungs, glomerular filtration in kidneys, and pharyngeal arch artery morphogenesis . It has gained significant research interest due to its emerging roles in cancer progression and its tissue-specific functions. Detecting GPR116 with antibodies is crucial for understanding its expression patterns and physiological roles.
GPR116 antibodies are validated for multiple applications including:
| Application | Recommended Dilution | Notes |
|---|---|---|
| Western Blot (WB) | 1:500-1:1000 | Validated for human samples |
| Immunohistochemistry (IHC) | 1:50-1:500 | Effective on FFPE tissues |
| Immunohistochemistry-Paraffin | 6 μg/ml | For fixed, paraffin-embedded tissues |
| ELISA | Application-dependent | Requires optimization |
| Flow Cytometry | Application-dependent | Validated for transfected cell lines |
Note: Optimal dilutions should be determined by each laboratory for their specific application .
Selection should be based on:
Species reactivity - confirm the antibody recognizes your target species (human GPR116 antibodies show cross-reactivity with primate samples and predicted reactivity with mouse, rat, and other mammals at varying homology percentages)
Application compatibility - ensure validation for your intended application (WB, IHC, flow cytometry)
Epitope location - antibodies targeting different domains (extracellular, transmembrane, or cytoplasmic) may yield different results
Clonality - polyclonal antibodies offer broader epitope recognition while monoclonal antibodies provide higher specificity
GPR116 shows differential expression across tissues:
| Tissue | Expression Level | Detection Method | Notes |
|---|---|---|---|
| Lung | Highest (CT = 26.1 ± 0.5; ΔCT = 0.6 ± 0.3) | qPCR | Primary site of expression |
| Kidney | High (CT = 28.1 ± 0.3; ΔCT = 5.0 ± 0.6) | qPCR | Higher in medulla than cortex |
| Heart | Moderate (CT = 28.0 ± 0.5; ΔCT = 5.8 ± 0.5) | qPCR | Detectable expression |
| Cancer tissues | Variable upregulation | WB, IHC | Increased in gastric and colorectal cancers |
This expression pattern has been confirmed at the protein level by Western blot analysis .
GPR116 shows specific cellular localization in the kidney:
Primarily expressed in acid-secreting A-intercalated cells (A-ICs) in the cortical and medullary collecting ducts
Colocalization with the B subunit of vacuolar-type H+-ATPase in the medulla
In outer medullary collecting ducts, GPR116-expressing cells are interspersed among AQP2-expressing principal cells
In the renal cortex, GPR116 localizes to only a subset of AQP2-negative cells
For optimal detection of this pattern, immunofluorescence imaging on fixed-frozen kidney sections using validated antibodies is recommended. Colocalization studies with markers like V-ATPase (for A-ICs) and AQP2 (for principal cells) provide definitive cellular identification .
For optimal Western blot detection of GPR116:
Sample preparation:
Use whole-organ lysates or cell lysates (HEK293, HeLa, or Y79 cells show positive expression)
Include positive controls (e.g., HEK293 cells expressing murine or human GPR116)
Gel electrophoresis:
Use appropriate percentage gels to resolve the 149 kDa protein
Load adequate protein amount (30-50 μg total protein)
Transfer and detection:
Use PVDF membrane for optimal protein binding
Block with 5% non-fat milk or BSA
Apply primary antibody at 1:500-1:1000 dilution
Incubate overnight at 4°C
Use appropriate HRP-conjugated secondary antibody
Expected results:
For successful GPR116 immunohistochemistry:
Primary antigen retrieval methods:
TE buffer pH 9.0 (recommended as first choice)
Citrate buffer pH 6.0 (alternative method)
Protocol specifics:
For FFPE tissue sections: 5-7 μm thickness
Heat-induced epitope retrieval: 95-98°C for 15-20 minutes
Cool to room temperature: 20 minutes
Block endogenous peroxidase activity: 3% H₂O₂
Block non-specific binding: 5-10% normal serum
Primary antibody incubation: 1:50-1:500 dilution, overnight at 4°C
Detection system: HRP-polymer or ABC method with DAB visualization
Special considerations:
Investigating GPR116 activation requires specialized approaches:
Tethered agonist activation studies:
Generate constructs with mutations near GPS cleavage site (e.g., H991A mutation)
Use antibodies to detect receptor cleavage products (NTF and CTF)
Compare wild-type and mutant receptors using conformational antibodies
Molecular mechanism analysis:
Employ antibodies recognizing specific domains to monitor conformational changes
Detect interactions with downstream signaling proteins (GNAQ/GNA11)
Track receptor internalization upon activation
In vivo validation using knock-out models:
For cancer prognostic studies:
Tissue microarray analysis:
Optimize antibody dilution (1:50-1:500) for IHC on FFPE cancer tissues
Establish scoring system (e.g., intensity score 0-3+)
Correlate staining intensity with clinicopathological parameters
Survival analysis methodology:
Stratify patients based on GPR116 expression levels
Generate Kaplan-Meier survival curves based on antibody staining intensity
Perform multivariate Cox regression analysis to establish independent prognostic value
Example findings from gastric cancer studies:
Common challenges and solutions:
High background in IHC:
Increase blocking time/concentration (use 5-10% normal serum)
Optimize antibody dilution (try 1:100 instead of 1:50)
Include additional washing steps
Test alternative antigen retrieval methods (switch from TE pH 9.0 to citrate pH 6.0)
Weak or no signal in Western blot:
Ensure adequate protein loading (≥30 μg)
Optimize transfer conditions for high molecular weight protein (149 kDa)
Increase antibody concentration or incubation time
Use enhanced chemiluminescence detection system
Non-specific bands:
Thorough validation approaches include:
Genetic controls:
Compare staining in wild-type vs. GPR116 knockout tissues
Use siRNA/shRNA knockdown cells versus control cells
Test over-expression systems (e.g., HEK293 cells transfected with GPR116)
Epitope-specific validation:
Use blocking peptides specific to the antibody epitope
Compare antibodies targeting different domains of GPR116
Analyze GPR116 mutants (e.g., Δexon17 Gpr116) to confirm specificity
Cross-reactivity assessment:
GPR116 activates several signaling pathways that can be monitored using antibody-based techniques:
Primary signaling pathways:
PI3K-Akt signaling pathway
Extracellular matrix-receptor interaction
Focal adhesion signaling
Cell adhesion pathways
Detection methods:
Phospho-specific antibodies to track Akt activation
Co-immunoprecipitation to detect GPR116 interaction with GNAQ/GNA11
Western blotting for downstream signaling intermediates
Immunofluorescence to track protein translocation upon activation
Functional correlation:
Investigating GPR116 in surfactant regulation:
Cellular localization studies:
Use antibodies to confirm GPR116 expression in alveolar type II epithelial (AT2) cells
Perform immunofluorescence co-staining with AT2 cell markers
Functional studies:
Compare GPR116 expression in normal vs. surfactant-accumulation conditions
Analyze effects of Δexon17 GPR116 mutation using domain-specific antibodies
Monitor localization changes in response to surfactant stimulation
Mechanistic investigation:
Emerging therapeutic targeting approaches:
Expression profiling in disease states:
Use antibodies to quantify GPR116 upregulation in cancer tissues
Compare expression levels between primary tumors and metastases
Correlate with treatment response in patient cohorts
Mechanistic studies for drug development:
Screen for compounds that modulate GPR116 activity using antibody-based assays
Detect conformational changes induced by potential therapeutic agents
Monitor downstream signaling modulation using phospho-specific antibodies
Therapeutic antibody development:
Current knowledge and detection methods:
Known GPR116 variants:
Exon17 deletion affects transmembrane domains
H991A mutation prevents GPS autocatalytic cleavage
Various splice variants may exist across tissues
Detection strategies:
Domain-specific antibodies to distinguish variant forms
Western blotting to assess molecular weight differences
Immunoprecipitation followed by mass spectrometry for detailed characterization
Functional implications: