TAS2R7 antibodies are critical for studying receptor localization, expression patterns, and functional interactions.
Western Blot: Detects TAS2R7 in tissue lysates, with optimal dilution ranges between 1:500 and 1:5000 .
Immunofluorescence: Visualizes receptor distribution in taste buds and intestinal epithelia .
ELISA: Quantifies TAS2R7 levels using conjugated variants (e.g., HRP, FITC, or biotin) .
| Conjugate | Product Code (Example) | Application |
|---|---|---|
| HRP | CSB-PA868363OB01HU | Quantitative ELISA |
| FITC | CSB-PA868363OC01HU | Cell imaging |
| Biotin | CSB-PA868363OD01HU | Streptavidin assays |
TAS2R7 detects bitter compounds via gustducin-coupled signaling, activating phospholipase Cβ2 (PLCβ2) and transient receptor potential channel TRPM5 . It responds to divalent/trivalent salts (e.g., Zn²⁺, Cu²⁺, Al³⁺) and bitter substances like cromolyn .
Regulates enteroendocrine hormone release and bile acid metabolism .
Modulates immune responses and metabolic pathways linked to obesity .
No 3D structure available for TAS2R7, complicating epitope mapping .
Variable efficacy reported for bitter ligands (e.g., quinine, diphenidol) .
Preclinical studies highlight TAS2R7's potential as a drug target for metabolic disorders. For example:
TAS2R7 (Taste Receptor Type 2 Member 7) is a G protein-coupled receptor that functions as a gustducin-coupled receptor implicated in the perception of bitter compounds. It is primarily expressed in subsets of taste receptor cells of the tongue and palate epithelium, exclusively in gustducin-positive cells . TAS2R7 plays a crucial role in bitter taste perception in the oral cavity and also functions in sensing the chemical composition of gastrointestinal content. Recent research has identified TAS2R7 as a unique metal cation receptor that responds to a broad range of divalent and trivalent salts, including zinc, calcium, magnesium, copper, manganese, and aluminum, which contributes to the bitter taste perception of these metal ions .
TAS2R7 is a membrane-bound G protein-coupled receptor with seven transmembrane domains. The protein has 318 amino acids with molecular weight of approximately 36-37 kDa . Functionally, TAS2R7 signals through PLCB2 and the calcium-regulated cation channel TRPM5 . Molecular modeling and mutagenesis analysis have identified specific residues, particularly H94, that are critical for metal ion binding and receptor activation . The receptor demonstrates selective responses to divalent and trivalent salts but not to monovalent salts like KCl, suggesting a specific tuning mechanism for higher-valence cations .
For Western blot applications using TAS2R7 antibodies, the following methodological considerations are important:
Sample preparation: Cell lysates from relevant cell lines (e.g., HL-60, K562) have been successfully used to detect TAS2R7 .
Dilution range: Recommended dilutions typically range from 1:500 to 1:2000 for Western blot applications . For specific antibodies like that from Novus Biologicals, a concentration of 1.0 μg/ml has been recommended .
Detection system: Standard secondary antibody detection systems compatible with rabbit IgG should be used.
Protein size: The expected molecular weight for detection is approximately 36-37 kDa .
Storage conditions: Most antibodies should be stored at -20°C for long-term storage to maintain reactivity, with aliquoting recommended to avoid freeze-thaw cycles .
Validating TAS2R7 antibody specificity requires multiple approaches:
Positive controls: Use cell lines known to express TAS2R7, such as taste receptor cells or transfected cell lines (e.g., HEK293 cells transiently transfected with TAS2R7) .
Negative controls: Include mock-transfected cells or cells known not to express TAS2R7.
Immunostaining validation: Compare immunostaining patterns with expected cellular localization (membrane-bound for TAS2R7) .
Peptide competition assays: Pre-incubate the antibody with the immunizing peptide to confirm binding specificity.
Molecular techniques: Consider validating with complementary techniques such as RT-PCR to confirm expression at the mRNA level.
Knockdown experiments: siRNA knockdown of TAS2R7 should reduce antibody signal if the antibody is specific.
Several functional assays can be employed alongside antibody-based detection:
Calcium mobilization assay: This assay monitors TAS2R7 activation by measuring intracellular calcium release. HEK293 cells transfected with TAS2R7 along with a coupling chimeric G protein (Gα16-gust44) can be used, and receptor activation monitored using calcium-sensitive dyes .
Dose-response experiments: These can determine the sensitivity of TAS2R7 toward various ligands, including metal ions. Studies have shown TAS2R7 responds to metal ions in a dose-dependent manner with varying EC50 values (Al2(SO4)3: 39±15 μM; CuSO4: 1.04±0.36 mM; ZnSO4: 3.36±0.14 mM; MgCl2: 6.07±1.07 mM; CaCl2: 5.27±0.50 mM; MnCl2: 6.59±1.73 mM) .
Immunofluorescence assays: These can be used to visualize receptor localization in cells using specific antibodies .
Molecular modeling and mutagenesis: Site-directed mutagenesis can be used to identify critical residues involved in ligand binding and receptor activation, complementing antibody studies .
Research has shown that TAS2R7 interacts differently with various metal ions, with distinct potency and efficacy profiles:
Metal ion sensitivity profile: TAS2R7 demonstrates highest sensitivity to aluminum sulfate (EC50: 39±15 μM), followed by copper sulfate, zinc sulfate, magnesium chloride, calcium chloride, and manganese chloride .
Experimental approaches:
Dose-response analysis: Systematic testing with varying concentrations of metal ions can establish sensitivity profiles.
Calcium-free assays: Performing assays in calcium-free conditions can eliminate potential interference from calcium in the assay buffer.
Anion effect studies: Comparing responses to different salt forms (e.g., ZnSO4 vs. ZnCl2) can determine if anions affect potency or efficacy .
Molecular docking: Computational approaches can predict binding sites and interaction modes. For example, electrostatic potential calculations using APBS and molecular surface generation with water probes can model metal ion interactions .
Site-directed mutagenesis: Mutating specific residues (e.g., H94) can validate predicted binding sites .
TAS2R7 shows distinctive characteristics compared to other bitter taste receptors:
Narrowly tuned receptor: Unlike many TAS2Rs that respond to multiple bitter compounds, TAS2R7 appears to be relatively selective, responding primarily to metal ions and cromolyn (at high concentrations, EC50: 5.9 mM) .
Metal ion sensitivity: TAS2R7 is unique in responding to multiple metal ions, a property shared among GPCRs only with the calcium-sensing receptor (CaSR) .
Signal transduction: Like other bitter taste receptors, TAS2R7 signals through PLCB2 and TRPM5 , but its specific downstream pathways when activated by metal ions versus organic compounds might differ.
Experimental approach considerations:
Comparative analysis: Side-by-side testing of TAS2R7 with other TAS2Rs using the same assay conditions.
Pathway inhibitor studies: Using inhibitors of different signaling components to elucidate specific pathways activated by TAS2R7.
Receptor chimeras: Creating chimeric receptors between TAS2R7 and other TAS2Rs to identify domains responsible for specific ligand interactions.
Developing specific TAS2R7 antibodies presents several challenges:
Sequence homology: TAS2Rs share sequence similarities, which can lead to cross-reactivity. Careful epitope selection from unique regions is crucial.
Membrane protein challenges: As a seven-transmembrane protein, TAS2R7 has limited exposed regions for antibody targeting, often requiring use of synthetic peptides rather than full-length protein for immunization.
Validation complexity: Confirming specificity requires multiple approaches including testing in different tissues and cell types.
Potential solutions:
Epitope mapping: Systematic analysis to identify unique regions within TAS2R7.
Recombinant expression: Expression of specific domains for antibody generation.
Affinity purification: Two-step purification against target peptide and cross-absorption against related TAS2R peptides.
Advanced validation: Using CRISPR/Cas9 knockout models to confirm specificity.
Monoclonal development: Development of monoclonal antibodies against specific epitopes for increased specificity.
Emerging research suggests potential roles for bitter taste receptors including TAS2R7 in cancer:
Expression patterns: Studies indicate that TAS2Rs are expressed in various cancerous cell lines and tissues, with some being predominantly downregulated in cancerous compared to non-cancerous samples .
Prognostic significance: Higher expression levels of TAS2Rs in primary cancerous cells and tissues have been associated with improved prognosis in humans .
Anti-cancer effects: Receptor-specific, agonist-mediated activation of TAS2Rs has been shown to induce various anti-cancer effects, including decreased cell proliferation, migration, and invasion, as well as increased apoptosis .
Research applications of TAS2R7 antibodies:
Expression profiling: Antibodies can be used to assess TAS2R7 expression levels in different cancer types.
Prognostic markers: TAS2R7 detection might serve as a prognostic marker given the association between TAS2R expression and cancer outcomes.
Therapeutic target validation: Antibodies can help validate TAS2R7 as a potential therapeutic target.
Mechanism studies: Immunoprecipitation using TAS2R7 antibodies can identify interacting partners in cancer cells.
Research on TAS2R genetic variations and disease susceptibility represents an emerging field:
Genetic association studies: Variations in TAS2R genes, including TAS2R7, have been studied in relation to cancer risk, though findings remain inconclusive .
Functional consequences: Genetic variations in TAS2Rs might affect receptor function, potentially impacting tissue-specific functions relevant to carcinogenesis .
Role of antibodies in variant research:
Expression analysis: Antibodies can be used to assess whether genetic variants affect protein expression levels.
Localization studies: Immunohistochemistry can determine if variants affect receptor trafficking and localization.
Functional studies: Combined with genetic data, antibody-based detection can link genotype to phenotype through functional analyses.
Methodology considerations: When studying variants, researchers should use antibodies targeting conserved regions not affected by the variations under investigation.
Investigating TAS2R7 in extra-oral tissues requires specific methodological approaches:
Tissue-specific expression verification:
Use validated TAS2R7 antibodies for immunohistochemistry to confirm expression in the tissue of interest.
Complement with RT-PCR or RNA-seq to verify transcript expression.
Include positive controls (taste tissue) and negative controls.
Function in non-taste contexts:
Develop tissue-specific functional assays that may differ from taste cell assays.
Consider cell-type specific differences in signaling components.
Disease context considerations:
Compare expression between normal and pathological tissues using consistent methodology.
Account for potential changes in receptor localization or post-translational modifications in disease states.
Consider environmental factors (e.g., pH changes in tumor microenvironments) that might affect antibody binding or receptor function.
Technical challenges and solutions:
Low expression levels: Use high-sensitivity detection methods or signal amplification.
Background staining: Optimize blocking conditions and use highly specific antibodies.
Tissue heterogeneity: Consider single-cell approaches or laser capture microdissection.
Functional verification: Combine antibody detection with functional assays to confirm not just expression but activity in the tissue of interest.
TAS2R7 antibodies can facilitate high-throughput screening through several approaches:
Cell-based screening platforms:
Develop stable cell lines expressing TAS2R7 that can be validated with antibodies.
Use antibodies to confirm receptor expression levels across screening plates.
Employ antibody-based detection methods as secondary validation for hits identified in primary functional screens.
Binding assays:
Develop competitive binding assays using labeled antibodies.
Use antibodies in proximity-based assays (e.g., BRET, FRET) to detect conformational changes upon ligand binding.
Technical considerations:
Select antibodies recognizing extracellular domains for live-cell applications.
Validate that antibody binding doesn't interfere with ligand interaction sites.
Consider developing conformation-specific antibodies that recognize active or inactive states of the receptor.
Recent advances in TAS2R7 structural biology include:
Homology modeling: Studies have employed homology modeling based on related GPCRs, such as the 5-HT2C serotonin receptor, to predict TAS2R7 structure .
Binding site identification: Molecular modeling and mutagenesis have identified potential binding sites for metal ions and other ligands .
Antibody contributions to structural studies:
Conformational antibodies: Development of antibodies recognizing specific conformational states can provide insights into receptor dynamics.
Crystallization aids: Antibodies can be used as crystallization chaperones to stabilize the receptor for structural studies.
Epitope mapping: Systematic epitope mapping with antibodies can validate predicted structural elements.
Structure-function relationships: Combining antibody binding studies with functional assays can link structural elements to receptor function.
Methodological approach:
Generate panel of antibodies against different domains.
Use antibody accessibility studies to validate topology models.
Employ antibodies in cross-linking studies to identify spatial relationships between domains.
The field of TAS2R7 structural biology remains developing, with antibodies playing a crucial role in validating computational models and advancing understanding of this important taste receptor.