OR10K1 and OR10K2 are members of the olfactory receptor family, specifically belonging to subfamily 10. These G-protein coupled receptors are primarily involved in olfactory signal transduction. While traditionally associated with sensory functions, recent research has identified differential expression patterns of these receptors in various tissues beyond the olfactory epithelium, suggesting potential roles in other physiological processes. Notably, OR10K1 has been identified among the top downregulated differentially expressed genes (DEGs) in certain immune microenvironment studies, while OR10K2 appears among upregulated genes in similar contexts, indicating potentially divergent biological functions .
The observed molecular weight for OR10K1/OR10K2 in Western blot applications is approximately 72 kDa, though the calculated molecular weight based on amino acid sequence is approximately 35 kDa (35,079 Da specifically) . This discrepancy is not uncommon for membrane proteins like olfactory receptors, which often demonstrate higher apparent molecular weights on SDS-PAGE due to post-translational modifications (particularly glycosylation) and the hydrophobic nature of transmembrane domains affecting SDS binding and protein migration patterns. When troubleshooting, researchers should anticipate this higher molecular weight band rather than the theoretical weight calculated from the primary sequence .
Based on available antibody specifications, OR10K1/OR10K2 antibodies primarily demonstrate reactivity with human, mouse, and rat samples . While traditionally associated with olfactory epithelium, researchers should be aware that olfactory receptors have been detected in multiple non-olfactory tissues. Recent research has implicated these receptors in immune microenvironment contexts, suggesting expression in additional tissues beyond primary sensory organs . When designing experiments, researchers should include appropriate positive control tissues, particularly when investigating novel expression patterns in non-canonical locations.
Validation of OR10K1/OR10K2 antibodies should follow a multi-step approach:
Western blot validation: Confirm detection of a band at the expected 72 kDa molecular weight in appropriate positive control samples .
Knockout/knockdown validation: If possible, compare signal between wild-type samples and those where OR10K1/OR10K2 expression has been genetically reduced.
Peptide competition assay: Pre-incubate antibody with the immunogen peptide (derived from the internal region of human OR10K1/2) to confirm signal specificity .
Cross-reactivity assessment: Test the antibody against recombinant OR10K1 and OR10K2 separately to determine specificity for each target.
Application-specific validation: For each intended application (WB, ICC/IF, ELISA), perform concentration gradients to determine optimal working dilutions as suggested in the product specifications .
Distinguishing between OR10K1 and OR10K2 signals presents a technical challenge due to their sequence homology. Most commercially available antibodies recognize both proteins, as evidenced by product naming conventions (OR10K1/OR10K2) . To differentiate between these targets:
Complement antibody-based detection with transcript-specific qPCR to determine which isoform predominates in your experimental system.
Consider using epitope-tagged recombinant constructs for each receptor when overexpression is possible.
Employ competitive binding assays with peptides specific to non-conserved regions of each receptor.
Use bioinformatic analysis to identify unique epitopes and potentially develop custom antibodies targeting these regions if commercially available options cannot provide sufficient discrimination.
Implement orthogonal methods like mass spectrometry for definitive protein identification in complex samples.
For optimal preservation of OR10K1/OR10K2 antibody activity, researchers should:
For short-term storage and frequent use (up to one month), store at 4°C to minimize freeze-thaw cycles .
Prepare small working aliquots upon receipt to avoid repeated freeze-thaw cycles, which significantly diminish antibody performance .
Store in buffer containing 50% glycerol, 0.5% BSA, and 0.02% sodium azide as specified by manufacturers to maintain stability .
When retrieving from frozen storage, thaw antibodies gradually on ice rather than at room temperature to preserve binding capacity.
Monitor antibody performance periodically using positive controls to ensure continued reactivity over the storage period.
For optimal Western blotting results with OR10K1/OR10K2 antibodies:
Sample preparation: Extract proteins using RIPA buffer supplemented with protease inhibitors; heat samples at 95°C for 5 minutes in reducing sample buffer.
Gel selection: Use 10% SDS-PAGE gels for optimal resolution of the 72 kDa target protein .
Transfer conditions: Transfer to PVDF membrane at 100V for 60-90 minutes in cold transfer buffer containing 10-20% methanol.
Blocking: Block membranes with 5% non-fat dry milk in TBST for 1 hour at room temperature.
Primary antibody: Dilute OR10K1/OR10K2 antibody at 1:500 to 1:2000 in blocking buffer and incubate overnight at 4°C .
Washing: Wash membranes 3-5 times with TBST, 5-10 minutes per wash.
Secondary antibody: Incubate with HRP-conjugated anti-rabbit secondary antibody (1:5000) for 1 hour at room temperature.
Detection: Develop using enhanced chemiluminescence and expect a band at approximately 72 kDa .
Controls: Include positive control tissues and negative controls (secondary antibody only) in each experiment.
For immunofluorescence staining using OR10K1/OR10K2 antibodies:
Fixation: Fix cells with 4% paraformaldehyde for 15 minutes at room temperature or tissues with 10% neutral buffered formalin.
Permeabilization: Permeabilize with 0.1-0.2% Triton X-100 in PBS for 10 minutes for intracellular epitope access.
Blocking: Block with 5% normal serum (from the species of secondary antibody) in PBS with 0.1% Tween-20 for 1 hour.
Primary antibody: Dilute OR10K1/OR10K2 antibody at 1:200 to 1:1000 in blocking buffer and incubate overnight at 4°C .
Washing: Wash 3 times with PBS-T, 5 minutes each.
Secondary antibody: Incubate with fluorochrome-conjugated anti-rabbit secondary antibody at 1:500 in blocking buffer for 1 hour at room temperature.
Nuclear counterstaining: Counterstain with DAPI (1:1000) for 5 minutes.
Mounting: Mount with antifade mounting medium and seal with nail polish.
Controls: Include peptide competition controls and secondary-only controls to confirm specificity.
Imaging parameters: Begin imaging with lower exposure settings and adjust as needed, as olfactory receptors often show relatively low expression levels in non-olfactory tissues.
For ELISA applications using OR10K1/OR10K2 antibodies:
Antibody dilution: Use a high dilution factor of 1:20,000 for ELISA applications, significantly higher than for other applications like WB (1:500-1:2000) or IF/ICC (1:200-1:1000) .
Sample types: The antibody has been tested with cell culture supernatant, plasma, serum, and tissue homogenate samples .
Assay optimization: Perform preliminary titration experiments to determine optimal coating concentration, blocking conditions, and incubation times.
Controls: Include a standard curve using recombinant OR10K1/OR10K2 protein if available, or use samples with known expression levels as positive controls.
Cross-reactivity prevention: Pre-absorb the antibody with related proteins if cross-reactivity is a concern, particularly with other olfactory receptor family members.
Signal development: Monitor colorimetric development carefully, as signal-to-noise ratio can be challenging with low-abundance membrane proteins.
Data analysis: Use appropriate statistical methods to determine the limit of detection and quantification range for your specific experimental conditions.
Validation: Confirm ELISA results with orthogonal methods such as Western blotting when establishing the assay for novel sample types.
When facing weak or absent signals with OR10K1/OR10K2 antibodies:
Sample preparation: Ensure adequate protein extraction, particularly for membrane proteins, by using detergent-containing buffers (RIPA or NP-40) with complete protease inhibitor cocktails.
Antibody concentration: Increase primary antibody concentration; try the lower end of the recommended dilution range (1:500 for WB, 1:200 for IF/ICC) .
Incubation conditions: Extend primary antibody incubation to overnight at 4°C to increase binding efficiency.
Detection system: Switch to a more sensitive detection system (e.g., from colorimetric to chemiluminescent for WB, or implement signal amplification for IF).
Epitope retrieval: For fixed tissues/cells, optimize antigen retrieval methods (heat-induced or enzymatic) to improve epitope accessibility.
Expression levels: Verify target expression in your samples via RT-PCR before immunodetection, as OR10K1/OR10K2 may have tissue-specific expression patterns.
Antibody quality: Check antibody viability by testing on positive control samples known to express the target protein.
Buffer conditions: Ensure the antibody storage buffer maintains proper pH and contains stabilizers as recommended (50% glycerol, 0.5% BSA, 0.02% sodium azide) .
To reduce background and non-specific signals when working with OR10K1/OR10K2 antibodies:
Blocking optimization: Extend blocking time (2-3 hours) or switch blocking agents (BSA, normal serum, commercial blockers) to find optimal conditions.
Antibody dilution: Increase dilution of primary antibody; test the upper end of recommended ranges (1:2000 for WB, 1:1000 for IF/ICC) .
Washing stringency: Increase the number and duration of washes (5-6 washes of 10 minutes each with 0.1% Tween-20 in PBS/TBS).
Secondary antibody: Dilute secondary antibody further or pre-absorb with tissue/cell lysate from the species being analyzed.
Cross-adsorption: Pre-incubate primary antibody with non-specific proteins (e.g., non-relevant tissue lysate) to reduce non-specific binding.
Detergent adjustment: Increase detergent concentration in washing and antibody dilution buffers (0.2-0.3% Tween-20 or 0.1% Triton X-100).
Fixation optimization: For IF applications, test different fixation methods as over-fixation can increase background.
Filter selection: For fluorescence applications, ensure proper filter sets to minimize autofluorescence from tissues.
Endogenous enzyme blocking: For IHC applications, block endogenous peroxidase or phosphatase activity before antibody incubation.
When encountering unexpected bands with OR10K1/OR10K2 antibodies:
Expected vs. observed weight: While the calculated molecular weight is approximately 35 kDa, the observed weight is typically 72 kDa due to post-translational modifications and membrane protein properties .
Higher molecular weight bands: Bands above 72 kDa may represent:
Glycosylated forms (olfactory receptors often undergo N-linked glycosylation)
Dimers or oligomers if sample preparation was insufficient to fully denature complexes
Ubiquitinated forms marking the protein for degradation
Lower molecular weight bands: Bands below 72 kDa may indicate:
Proteolytic degradation products (enhance protease inhibition during sample preparation)
Splice variants (verify with transcript analysis)
Cross-reactivity with related olfactory receptor family members
Validation approaches:
Peptide competition assay to confirm which bands disappear when the antibody is pre-absorbed with the immunizing peptide
Sample treatment with glycosidases to identify glycosylated forms
Mass spectrometry analysis of excised bands for definitive identification
Literature comparison: Compare your results with published Western blot images of OR10K1/OR10K2 to identify common patterns.
OR10K1/OR10K2 antibodies can be valuable tools in investigating thymic epithelial tumor (THYM) immune microenvironments:
Expression pattern analysis: Use immunohistochemistry or immunofluorescence with OR10K1/OR10K2 antibodies to map receptor distribution in different THYM immunotypes. This is particularly relevant as OR10K1 has been identified among top downregulated genes in certain THYM immunotypes, while OR10K2 appears among upregulated genes .
Correlation studies: Combine OR10K1/OR10K2 immunostaining with immune cell markers to correlate receptor expression with immune infiltration patterns.
Prognostic assessment: Evaluate OR10K1/OR10K2 expression levels as potential prognostic biomarkers in THYM patient cohorts, given their inclusion in an 11-gene prognostic model .
Functional investigations:
Co-immunoprecipitation using validated OR10K1/OR10K2 antibodies to identify binding partners in tumor cells
Chromatin immunoprecipitation (ChIP) assays with transcription factors potentially regulating OR10K1/OR10K2 expression
Therapeutic response monitoring: Assess changes in OR10K1/OR10K2 expression before and after immunotherapeutic interventions.
Multi-omics integration: Combine antibody-based protein detection with transcriptomic data to build comprehensive models of receptor involvement in tumor immune microenvironments.
To investigate functional differences between OR10K1 and OR10K2:
Differential expression analysis:
Use antibodies in combination with transcript-specific probes to map differential expression patterns across tissues
Quantify relative abundance ratios between OR10K1 and OR10K2 in different physiological and pathological states
Receptor-specific knockdown/knockout:
Design siRNA or CRISPR approaches targeting unique regions of each receptor
Use OR10K1/OR10K2 antibodies to confirm protein reduction and assess phenotypic consequences
Ligand binding studies:
Express each receptor separately in heterologous systems
Use calcium imaging or cAMP assays to identify potential differential responses to ligands
Protein interaction networks:
Perform immunoprecipitation using epitope-tagged versions of each receptor
Identify specific interaction partners through mass spectrometry analysis
Signaling pathway analysis:
Assess downstream signaling activation (e.g., phosphorylation events) following receptor stimulation
Use phospho-specific antibodies in combination with OR10K1/OR10K2 detection
Structural biology approaches:
Generate structural models based on sequence differences
Design experiments to test predicted functional differences in ligand binding pockets or G-protein coupling domains
For successful multiplexed immunofluorescence including OR10K1/OR10K2 antibodies:
Antibody compatibility assessment:
Spectral considerations:
Select fluorophores with minimal spectral overlap for secondary antibodies
Include appropriate single-stain controls for spectral unmixing if using confocal or spectral imaging
Antigen abundance balancing:
Adjust exposure times for each channel based on relative abundance of targets
OR10K1/OR10K2 may require longer exposure or signal amplification if expression is low compared to other targets
Epitope retrieval optimization:
Test different antigen retrieval methods to ensure compatibility with all antibodies in the panel
Consider tyramide signal amplification for OR10K1/OR10K2 detection while maintaining compatibility with other retrieval methods
Staining sequence determination:
Test different staining sequences to determine optimal order
Generally stain for lower abundance targets (potentially OR10K1/OR10K2) first
Validation controls:
Include tissue microarrays or cell line controls with known expression patterns for all targets
Perform parallel single-marker staining to confirm multiplexed results
Analysis considerations:
Develop quantitative image analysis protocols that account for potential differences in subcellular localization
Use colocalization analysis to investigate potential functional relationships with other markers
The opposing regulation patterns of OR10K1 (downregulated) and OR10K2 (upregulated) in thymic epithelial tumors generate several research hypotheses:
Functional antagonism hypothesis: These receptors may have opposing functions in tumor microenvironment regulation, with OR10K1 potentially serving as a tumor suppressor while OR10K2 functions as an oncogenic factor.
Compensatory mechanism hypothesis: Upregulation of OR10K2 may represent a compensatory response to OR10K1 downregulation, suggesting overlapping but non-identical functions.
Immune microenvironment modulation hypothesis: The differential expression may influence distinct aspects of immune cell recruitment, activation, or suppression within the tumor microenvironment.
Prognostic biomarker development: The ratio of OR10K2 to OR10K1 expression might serve as a more robust prognostic indicator than either marker alone.
Research approaches to investigate these hypotheses include:
Correlation analyses between receptor expression and clinical outcomes
In vitro functional studies with receptor overexpression and knockdown
Immune cell co-culture experiments to assess effects on immune function
Integration with other components of the 11-gene prognostic model to understand pathway interactions
To investigate the molecular weight discrepancy between calculated (35 kDa) and observed (72 kDa) values for OR10K1/OR10K2 :
Post-translational modification analysis:
Treatment with glycosidases (PNGase F, Endo H) to remove N-linked glycans
Phosphatase treatment to remove phosphate groups
Treatment with deubiquitinating enzymes to remove ubiquitin modifications
Western blot analysis after each treatment to assess shifts in molecular weight
Expression system comparison:
Express recombinant OR10K1/OR10K2 with epitope tags in different systems (bacteria, insect cells, mammalian cells)
Compare molecular weights across expression systems with different post-translational modification capabilities
Use antibodies against both the epitope tag and OR10K1/OR10K2 to confirm identity
Mass spectrometry analysis:
Immunoprecipitate OR10K1/OR10K2 from native sources
Perform LC-MS/MS analysis to identify the protein and characterize modifications
Quantify the stoichiometry of different modifications
Membrane protein solubilization comparison:
Test different detergents and solubilization methods
Compare observed molecular weights under different denaturation conditions
Assess potential detergent-resistant oligomerization
Domain deletion analysis:
Express truncated versions of the receptors lacking specific domains
Determine which regions contribute most significantly to the aberrant migration pattern
Integration of OR10K1/OR10K2 antibody-based detection with other -omics approaches offers several innovative research opportunities:
Spatial transcriptomics integration:
Combine OR10K1/OR10K2 immunofluorescence with spatial transcriptomics to correlate protein expression with local transcriptional profiles
Map receptor distribution in relation to tissue microenvironments and signaling gradients
Single-cell proteogenomics:
Integrate antibody-based flow cytometry for OR10K1/OR10K2 with single-cell RNA sequencing
Identify cell populations with discordant mRNA and protein expression patterns
Interactome mapping:
Use proximity labeling approaches (BioID, APEX) with OR10K1/OR10K2 as baits
Identify tissue-specific protein interaction networks through mass spectrometry
Validate key interactions with co-immunoprecipitation using OR10K1/OR10K2 antibodies
Clinical proteogenomics:
Correlate OR10K1/OR10K2 protein levels (by IHC or targeted proteomics) with genomic alterations in large patient cohorts
Develop integrated prognostic models incorporating both genetic and protein expression data
Drug discovery applications:
Use OR10K1/OR10K2 antibodies in high-content screening to identify compounds that modulate receptor expression or localization
Combine with transcriptional profiling to understand mechanism of action
Multi-modal imaging:
Develop correlative imaging workflows combining OR10K1/OR10K2 immunodetection with techniques like MALDI imaging mass spectrometry
Map receptor expression in relation to metabolite distributions in tissues