OR2H1 antibodies are polyclonal or monoclonal reagents designed to bind specific epitopes of the OR2H1 protein. Key features include:
Property | Details |
---|---|
Target Protein | Olfactory Receptor OR2H1 (UniProt: Q9GZK4; Gene ID: 26716) |
Host Species | Rabbit (common), others vary by product |
Applications | IHC, ELISA, Western Blot (WB), Immunocytochemistry (ICC) |
Reactivity | Human-specific |
Immunogen | Synthetic peptides (e.g., residues 1–100) or fusion proteins containing OR2H1 sequences |
Molecular Weight | ~35 kDa (predicted) |
Proteintech 26954-1-AP: Validated for IHC (1:50–1:500 dilution) and ELISA, detects OR2H1 in lung cancer tissues .
Thermo Fisher OSR00280W: Targets extracellular domains, validated for WB and ICC .
OR2H1 exhibits tumor-specific overexpression, making it a promising therapeutic target. Key data include:
Mechanistic Role: OR2H1 enhances glucose uptake via 2-NBDG-sensitive pathways, promoting tumor growth . CRISPR/Cas9 knockout of OR2H1 reduces cancer cell proliferation and glucose metabolism .
OR2H1 antibodies underpin the development of CAR T-cell therapies for epithelial cancers:
Antibody specificity is confirmed through multiple methods:
OR2H1-targeted CAR T cells are in preclinical development for cancers with unmet needs:
OR2H1 is an olfactory receptor from the G-protein coupled receptor (GPCR) family with transmembrane domains that interact with odorant molecules in olfactory neurons. Its significance in cancer research stems from its unique expression pattern – widely expressed in various solid epithelial tumors while showing limited expression in normal tissues (primarily restricted to testis). This selective expression profile makes OR2H1 a potentially valuable target for cancer immunotherapy, particularly for CAR T cell approaches targeting solid tumors .
OR2H1 expression has been documented across multiple solid tumor types with varying frequencies:
Intrahepatic cholangiocarcinoma: ~69% of cases
Prostate cancer: >38% of cases
Serous endometrial carcinoma: ~27% of cases
Ovarian carcinomas: 20% of cases (8/40 in immunohistochemistry studies)
Lung carcinomas: 13% of cases (8/60)
Cholangiocarcinomas: 59% of cases (43/73)
Colon cancer: ~4% of cases
Immunohistochemistry, RT-QPCR, and RNAscope analyses have confirmed OR2H1 expression in pancreatic, ovarian, lung, and breast cancers of various histologies .
Multiple complementary methods have been validated for detecting OR2H1 expression:
Method | Sample Type | Detection Target | Advantages |
---|---|---|---|
RT-QPCR | Fresh/frozen tissue | mRNA | Quantitative, high sensitivity |
Western Blot | Protein lysates | Protein | Size verification, semi-quantitative |
RNAscope | FFPE tissue sections | RNA | In situ visualization, high specificity |
Immunohistochemistry | FFPE tissue sections | Protein | Protein localization, clinical compatibility |
Immunocytochemistry | Fixed cells | Protein | Cellular localization |
For RT-QPCR, researchers have successfully used primers (Forward: 5'-TCACTCAGTACAGCTCCCATGC-3'; Reverse: 5'-TTCAGTTCTTGCAATTAAGTCAGACTCT-3') with GAPDH as a reference gene for normalization .
Validating OR2H1 antibody specificity requires a multi-faceted approach:
Gene knockout controls: Utilize CRISPR/Cas9-mediated OR2H1 knockout cells as a negative control to confirm antibody specificity. This approach has been successfully employed to validate OR2H1 detection in published studies .
Multiple detection methods: Compare protein detection via immunohistochemistry with mRNA expression data (RT-QPCR or RNAscope). Concordance between protein and mRNA detection strengthens confidence in antibody specificity.
Tissue panel screening: Screen a diverse panel of normal tissues known to be OR2H1-negative (based on databases like GTEX Portal and The Human Protein Atlas) alongside positive controls (testis tissue or known OR2H1-expressing tumor cells).
Blocking experiments: Conduct peptide competition assays using the synthetic peptide immunogen to confirm binding specificity of the antibody.
Antibody validation in multiple applications: Verify consistent results across Western blot, IHC, and flow cytometry applications, confirming expected molecular weight and cellular localization patterns .
Several critical factors impact OR2H1 antibody performance in FFPE tissue staining:
Antigen retrieval method: Studies have shown that heat-induced epitope retrieval is essential for OR2H1 detection in FFPE tissues. The precise pH and buffer composition should be optimized based on the specific antibody.
Antibody concentration: Titration experiments are necessary to determine optimal antibody concentration, typically ranging from 1:500 to 1:2000 for primary antibodies .
Incubation conditions: Temperature and duration significantly impact staining quality. Overnight incubation at 4°C often yields better signal-to-noise ratio than shorter incubations at room temperature.
Detection system: The choice between chromogenic (DAB) versus fluorescent detection should be based on the expression level of OR2H1 and the need for multiplexing with other markers.
Tissue fixation variables: Fixation time, type of fixative, and tissue processing methods all influence epitope preservation. When possible, standardize these parameters across all samples being compared .
Controls: Include testis tissue as a positive control and ileum as a negative control, as established in published research protocols .
Quantification of OR2H1 protein can be approached through several methodologies:
Fluorescence-based flow cytometry: Conjugate OR2H1 antibody to fluorophores (such as PE) and quantify relative expression levels using calibration beads. This allows estimation of receptor density per cell.
Western blot densitometry: Semi-quantitative analysis using housekeeping proteins (like GAPDH) as loading controls can provide relative expression levels across samples.
IHC scoring systems: Implement H-score or Allred scoring systems that account for both staining intensity and percentage of positive cells. Digital pathology platforms can provide objective quantification of staining intensity.
Quantitative mass spectrometry: For absolute quantification, targeted proteomics approaches using labeled peptide standards can determine exact copy numbers of OR2H1 protein.
For optimal results, researchers should combine multiple quantification methods to validate findings across different platforms .
Designing experiments to elucidate OR2H1's functional role requires a comprehensive approach:
Gene modulation strategies:
CRISPR/Cas9 knockout: Generate complete OR2H1 knockout cell lines
shRNA/siRNA: Create transient and stable knockdown models
Overexpression models: Introduce OR2H1 in cell lines with low/no expression
Functional assays:
Cell proliferation: Measure growth rates using real-time cell analysis systems
Metabolism: Assess glucose uptake using fluorescent glucose analogs (e.g., 2-NBDG)
Migration/invasion: Determine metastatic potential using Boyden chamber assays
Colony formation: Evaluate clonogenic potential in 2D and 3D cultures
Signaling pathway analysis:
Investigate G-protein coupled signaling pathways typically associated with olfactory receptors
Assess metabolic signaling changes, as OR2H1 has been implicated in glucose metabolism
Examine potential cross-talk with oncogenic pathways
In vivo models:
Xenograft studies comparing OR2H1 wildtype versus knockout cells
Patient-derived xenografts to maintain tumor heterogeneity
Metastasis models to evaluate impact on tumor spread
Multiomics approach:
When evaluating OR2H1 antibody specificity for immunohistochemistry, these controls are essential:
Positive tissue controls:
Testis tissue (known to express OR2H1)
Validated OR2H1-positive tumor samples (cholangiocarcinoma, ovarian cancer)
Negative tissue controls:
Ileum tissue (confirmed OR2H1-negative)
Panel of normal tissues (58 normal tissue types have been confirmed OR2H1-negative)
Cellular controls:
Cell lines with known OR2H1 expression status
Isogenic cell lines with CRISPR/Cas9-mediated OR2H1 knockout
Technical controls:
Secondary antibody-only control (omit primary antibody)
Isotype control antibody (same species and isotype as OR2H1 antibody)
Peptide competition (pre-incubation with immunizing peptide)
Method validation controls:
Parallel RNAscope analysis for OR2H1 mRNA detection
Western blot verification of antibody specificity
These controls collectively ensure that any observed staining is specific to OR2H1 rather than technical artifacts or non-specific binding .
Determining the optimal cutoff for OR2H1 positivity requires a systematic approach:
Baseline expression determination:
Analyze a large panel of normal tissues (>50) to establish background levels
Quantify expression in positive control tissues (testis) to establish reference levels
Statistical approaches to cutoff determination:
ROC curve analysis comparing tumor vs. normal tissue staining
Survival analysis using various cutpoints to identify clinically meaningful thresholds
Tertile or quartile classification based on expression distribution within a cohort
Validation across detection methods:
Compare IHC positivity with RT-QPCR quantification
Validate with RNAscope to confirm RNA-protein correlation
Correlate with functional outcomes in experimental models
Clinical outcome correlation:
Retrospective analysis linking expression levels to patient survival
Association with response to standard therapies
Stratification by tumor type and stage
Standardization considerations:
Implement digital pathology quantification for objective scoring
Use H-score or Allred scoring systems that account for both intensity and percentage
Consider tissue-specific thresholds based on background expression
Based on published research, cutoffs for positivity have varied by tumor type, with cholangiocarcinomas showing a 59% positivity rate compared to 13% in lung carcinomas using the same evaluation criteria .
For optimal western blot detection of OR2H1, follow this detailed protocol:
Sample preparation:
Lyse cells or tissues in RIPA buffer supplemented with protease inhibitors
For membrane proteins like OR2H1, include brief sonication steps (3 x 10s pulses)
Determine protein concentration using BCA or Bradford assay
Gel electrophoresis:
Load 20-40μg total protein per lane
Use 10-12% polyacrylamide gels to properly resolve OR2H1 (~35 kDa molecular weight)
Include positive controls (testis tissue lysate) and negative controls
Transfer conditions:
Semi-dry or wet transfer to PVDF membrane (preferred over nitrocellulose for OR2H1)
Transfer at 100V for 1 hour or 30V overnight at 4°C
Blocking and antibody incubation:
Block with 5% non-fat milk in TBST for 1 hour at room temperature
Incubate with primary OR2H1 antibody at 1:500-1:2000 dilution overnight at 4°C
Wash 3x5 minutes with TBST
Incubate with HRP-conjugated secondary antibody (1:5000) for 1 hour at room temperature
Detection and analysis:
Develop using enhanced chemiluminescence (ECL) reagents
Expect OR2H1 band at approximately 35 kDa
Strip and reprobe for loading control (GAPDH or β-actin)
For quantification, normalize OR2H1 signal to loading control
Validation steps:
Optimizing immunohistochemistry for OR2H1 across tumor types requires systematic protocol refinement:
Fixation optimization:
Standard 10% neutral buffered formalin fixation for 24-48 hours
Consistent processing conditions to ensure comparable results
Antigen retrieval optimization:
Test multiple retrieval methods: citrate buffer (pH 6.0), EDTA buffer (pH 9.0), and enzymatic retrieval
Optimize retrieval time (10-30 minutes) and temperature
Blocking optimization:
Test 3-5% BSA, 5-10% normal serum, or commercial blocking reagents
Include avidin/biotin blocking if using biotin-based detection systems
Antibody titration:
Perform serial dilutions (1:100 to 1:2000) of OR2H1 antibody
Determine optimal concentration that maximizes specific signal while minimizing background
Consider extended incubation times (overnight at 4°C) for weak signals
Detection system selection:
For low expression: Use amplification systems (e.g., tyramide signal amplification)
For routine detection: Use polymer-based detection systems
For multiplex staining: Consider fluorescent secondary antibodies
Tumor-specific adaptations:
Lung tissue: Additional peroxidase blocking (3% H₂O₂, 15 minutes)
Highly pigmented tumors: Consider azure B treatment to reduce melanin interference
Necrotic areas: Implement careful region-of-interest selection
Counterstain optimization:
Adjust hematoxylin counterstaining time (30 seconds to 2 minutes) based on tumor type
For challenging tissues, consider nuclear fast red as an alternative counterstain
Based on published protocols, successful detection of OR2H1 has been achieved using DAB chromogen development following antigen retrieval in a variety of epithelial tumors .
For optimal quantification of OR2H1 mRNA expression via RT-QPCR:
RNA isolation optimization:
For FFPE samples: Use specialized kits designed for degraded RNA
For fresh/frozen tissues: Standard TRIzol or column-based methods
Include DNase treatment to eliminate genomic DNA contamination
cDNA synthesis considerations:
Use SuperScriptTM IV First-Strand Synthesis System or comparable high-efficiency reverse transcriptases
Include no-RT controls to detect genomic DNA contamination
Consider gene-specific primers for reverse transcription if expression is low
Primer design and validation:
Validated OR2H1-specific primers:
Forward: 5'-TCACTCAGTACAGCTCCCATGC-3'
Reverse: 5'-TTCAGTTCTTGCAATTAAGTCAGACTCT-3'
Design primers spanning exon-exon junctions to avoid genomic amplification
Verify primer specificity through melt curve analysis and sequencing of amplicons
Reference gene selection:
GAPDH has been validated as a suitable reference gene for OR2H1 studies
Forward: 5'-CCTGCACCACCAACTGCTTA-3'
Reverse: 5'-AGTGATGGCATGGACTGTGGT-3'
Consider multiple reference genes (ACTB, B2M, HPRT1) for more robust normalization
Validate reference gene stability across your specific sample types
Quantification strategy:
Absolute quantification: Generate standard curves using plasmids containing OR2H1
Relative quantification: Use 2^(-ΔΔCt) method with appropriate calibrator samples
Include inter-run calibrators when analyzing large sample sets across multiple plates
Data validation:
Discrepancies between OR2H1 mRNA and protein detection can arise from multiple factors:
Post-transcriptional regulation:
MicroRNA-mediated suppression of OR2H1 translation
RNA binding proteins affecting mRNA stability or translation efficiency
Alternative splicing generating protein isoforms not detected by certain antibodies
Methodological limitations:
Different sensitivity thresholds between RT-QPCR (typically more sensitive) and IHC/WB
Antibody epitope accessibility issues in fixed tissues
RNA quality differences affecting RT-QPCR reliability
Biological heterogeneity:
Intratumoral heterogeneity leading to sampling discrepancies
Temporal variations in OR2H1 expression during tumor progression
Microenvironmental factors influencing protein but not mRNA levels
Technical considerations:
FFPE fixation causing RNA degradation but preserving protein epitopes
Antibody cross-reactivity with related olfactory receptors
Reference gene instability affecting RT-QPCR normalization
Resolution strategies:
Perform single-cell analyses to account for heterogeneity
Use multiple detection methods on the same sample
Implement RNAscope as a bridge technology that provides spatial context for mRNA
Consider protein half-life vs. mRNA stability differences
Research shows OR2H1 detection rates are comparable between mRNA-based and protein-based methods in most tumor types, but discrepancies can occur in highly heterogeneous samples .
To address weak or inconsistent OR2H1 signals in western blots:
Sample preparation optimization:
Use membrane protein extraction kits specific for GPCRs
Avoid repeated freeze-thaw cycles of protein lysates
Include protease inhibitor cocktails optimized for membrane proteins
Consider non-denaturing conditions if epitope conformation is critical
Loading adjustments:
Increase total protein loading (up to 50-80μg for low abundance targets)
Concentrate samples using TCA precipitation or similar methods
Load positive controls (testis lysate) at various dilutions
Detection enhancement strategies:
Switch to high-sensitivity ECL substrates
Extend primary antibody incubation (up to 48 hours at 4°C)
Use signal amplification systems (biotin-streptavidin)
Consider fluorescent secondary antibodies with longer exposure times
Antibody optimization:
Try alternative OR2H1 antibodies targeting different epitopes
Reduce antibody dilution (1:200-1:500)
Add 0.05% Tween-20 to antibody dilution buffer to reduce non-specific binding
Test different blocking buffers (BSA vs. milk vs. commercial blockers)
Transfer optimization:
For membrane proteins like OR2H1, use PVDF membranes
Add 0.1% SDS to transfer buffer to improve elution of hydrophobic proteins
Reduce methanol concentration in transfer buffer to 10%
Consider longer transfer times at lower voltage
Sample handling guidelines:
To minimize non-specific binding in OR2H1 immunostaining:
Blocking optimization:
Test multiple blocking agents: 5-10% normal serum matching secondary antibody species
Include 0.1-0.3% Triton X-100 for permeabilization
Consider dual blocking with both serum and BSA
Add 0.1% gelatin to blocking buffer for highly autofluorescent tissues
Antibody concentration adjustment:
Titrate primary antibody across broader range (1:100-1:5000)
Increase washing duration and number of washes (5x5 minutes)
Dilute antibodies in blocking buffer rather than PBS/TBS alone
Pre-absorb antibody with tissue powder from negative tissues
Protocol modifications:
Implement endogenous peroxidase quenching (3% H₂O₂, 10 minutes)
Add avidin/biotin blocking for biotin-based detection systems
Include mouse-on-mouse blocking for mouse antibodies on mouse tissues
Reduce secondary antibody concentration
Advanced techniques:
Use monovalent Fab fragments to block endogenous immunoglobulins
Implement protein A/G pre-treatment to block endogenous immunoglobulins
Consider direct conjugated primary antibodies to eliminate secondary antibody issues
Use isotype-matched control antibodies to identify non-specific binding patterns
Validation strategies:
OR2H1 antibodies are instrumental in CAR T cell therapy development through multiple applications:
Target validation and screening:
Evaluate OR2H1 expression across tumor types and normal tissues
Quantify receptor density using antibody-based flow cytometry
Screen patient biopsies to identify suitable candidates for OR2H1-targeted therapy
CAR design and optimization:
Derive single-chain variable fragments (scFvs) from OR2H1 antibodies
Screen antibody libraries to identify optimal OR2H1-binding domains
Test various antibody-derived binding domains for CAR construction
Preclinical efficacy assessment:
Monitor target engagement via competitive binding assays
Assess on-target/off-tumor binding in tissue cross-reactivity studies
Evaluate CAR T cell infiltration in tumor models using OR2H1 co-staining
Patient selection biomarkers:
Develop IHC-based companion diagnostics for patient stratification
Establish quantitative thresholds for OR2H1 positivity
Create standardized scoring systems for clinical implementation
Therapy monitoring:
Track changes in OR2H1 expression during treatment
Identify escape mechanisms through epitope mapping
Monitor emergence of OR2H1-negative tumor subpopulations
Research has demonstrated that scFvs derived from OR2H1 antibodies can successfully redirect T cells against OR2H1-expressing tumors, showing cytotoxic activity both in vitro and in vivo against ovarian, lung, and other epithelial tumors .
OR2H1's involvement in glucose metabolism has significant implications for cancer research:
Metabolic targeting strategies:
OR2H1 knockout via CRISPR/Cas9 has demonstrated reduced glucose uptake in cancer cells
The 2-NBDG assay reveals functional consequences of OR2H1 expression on glucose metabolism
This metabolic role suggests OR2H1 may contribute to the Warburg effect in tumors
Dual-targeting approaches:
Combined targeting of OR2H1 and glucose metabolism pathways may yield synergistic effects
Glycolysis inhibitors could potentially sensitize tumors to OR2H1-targeted therapies
Metabolic imaging could serve as a biomarker for OR2H1 functional activity
Mechanistic research directions:
Investigation of signaling pathways connecting OR2H1 to glucose transporters
Analysis of how OR2H1 affects metabolic enzyme expression or activity
Exploration of potential role in hypoxia response and metabolic adaptation
Clinical correlations:
Assessment of relationship between OR2H1 expression and FDG-PET avidity
Evaluation of OR2H1 status in treatment-resistant tumors with altered metabolism
Analysis of metabolic signatures as predictive biomarkers for OR2H1-targeted therapies
Therapeutic implications:
Development of bispecific antibodies targeting both OR2H1 and metabolic enzymes
Design of OR2H1 antagonists that may disrupt metabolic signaling
Exploration of combination strategies with metabolic inhibitors
CRISPR/Cas9-mediated ablation of OR2H1, followed by glucose uptake analysis using fluorescent glucose analogs, has confirmed OR2H1's tumor-promoting role in glucose metabolism, suggesting novel therapeutic opportunities through metabolic disruption .
Multiplexed detection of OR2H1 with other biomarkers can significantly advance cancer research:
Tumor heterogeneity characterization:
Co-stain OR2H1 with other tumor markers to identify distinct subpopulations
Quantify spatial relationships between OR2H1+ cells and stromal/immune components
Correlate OR2H1 expression with regions of hypoxia, proliferation, or apoptosis
Pathway analysis applications:
Multiplex OR2H1 with glucose transporters (GLUT1/3) to validate metabolic correlations
Co-stain with phospho-proteins in relevant signaling pathways
Evaluate relationship with hypoxia markers (HIF-1α) and metabolic enzymes
Immune contexture evaluation:
Assess OR2H1 expression relative to tumor-infiltrating lymphocytes
Correlate with immune checkpoint molecules (PD-L1, CTLA-4)
Evaluate relationship with myeloid cell infiltration patterns
Technical approaches:
Multiplex immunofluorescence with tyramide signal amplification
Cyclic immunofluorescence for high-parameter tissue analysis
Mass cytometry imaging (IMC) for highly multiplexed protein detection
Digital spatial profiling for quantitative spatial analysis
Clinical applications:
Develop prognostic algorithms incorporating OR2H1 with established biomarkers
Create patient stratification approaches for combination therapies
Design precision medicine strategies based on multiplexed biomarker profiles
Researchers have successfully implemented multiplexed detection approaches combining OR2H1 with other cancer biomarkers to enhance understanding of its biological context and therapeutic implications in various epithelial tumors .