CYP2A13 is highly efficient in metabolizing 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK), a tobacco-specific procarcinogen . The FITC-conjugated antibody has been used to localize CYP2A13 in human bronchial epithelia and pancreatic α-islet cells, linking its expression to site-specific toxicity .
In CYP2A13/2F1-humanized mouse models, CYP2A13 mediates naphthalene (NA)-induced cytotoxicity in the olfactory mucosa and lung, as confirmed by IF and IHC . The antibody’s FITC conjugate allows precise tracking of enzyme distribution in these tissues.
Double-labeling studies using FITC-conjugated secondary antibodies (paired with anti-CYP2A13 primary antibodies) revealed CYP2A13 expression in pancreatic α-cells, which produce glucagon . This finding highlights potential roles in islet cell function and toxin metabolism.
Supplier | Catalog Number | Conjugate | Applications | Price |
---|---|---|---|---|
Santa Cruz Biotechnology | sc-xxxx | FITC | IF, IHC, WB | $470.46/100 µL |
antibodies-online | ABINxxxxxxx | FITC | IF, ICC | $269.00/100 µL |
Novus Biologicals | NBP2-50184V | DyLight 405 | ELISA, IHC, WB | $399.00/100 µL |
Note: The Novus Biologicals antibody uses DyLight 405 but targets the same epitope as FITC-conjugated variants .
Specificity: The monoclonal clone F16 P2 D8 binds a C-terminal epitope conserved across CYP2A6, CYP2A7, and CYP2A13 . Cross-reactivity should be accounted for in multi-isoform studies.
Limitations: Not suitable for distinguishing CYP2A13 from CYP2A6/7 due to shared epitopes .
Optimization: Titration is required for IF/IHC to minimize background fluorescence .
CYP2A13 is a human cytochrome P450 monooxygenase encoded by a functional member of the human CYP2A gene subfamily. It has garnered significant attention in toxicology research due to its exceptional catalytic efficiency in the metabolic activation of tobacco-specific carcinogens, particularly 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK). Studies have demonstrated that CYP2A13 has a higher efficiency in NNK activation than any other human P450 enzymes examined to date . Beyond NNK, CYP2A13 also catalyzes the metabolic activation of several other chemical carcinogens, including 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanol (NNAL), 4-aminobiphenyl, aflatoxin B1, naphthalene, and styrene . This metabolic capacity makes CYP2A13 a critical enzyme in understanding tobacco-related carcinogenesis mechanisms, particularly in tissues where it is highly expressed.
CYP2A13 displays a highly selective expression pattern in human tissues. Immunohistochemical studies have shown that CYP2A13 protein is:
Highly expressed in the epithelial cells of human bronchus and trachea
Selectively expressed in the pancreatic islets, but not in the exocrine portion of adult human pancreas
Within pancreatic islets, predominantly expressed in α-islet cells (glucagon-producing cells), which constitute approximately 15-20% of total islet cells
Mainly localized in the peripheral islet region with some single cells in the central part of the islet
Not detected in human liver, heart, testis, and ovary
Expression also observed in human fetal pancreatic islet cells
This tissue-specific distribution correlates with the incidence of smoking-related disease in corresponding tissues, supporting the hypothesis that CYP2A13-mediated in situ activation of tobacco toxicants contributes to pathology.
Validating antibody specificity is critical for accurate CYP2A13 research. A methodological approach should include:
Cross-reactivity testing: Confirm that the antibody does not cross-react with similar P450 proteins, particularly CYP2A6 and CYP2A7, which share significant sequence homology with CYP2A13. Validated antibodies should be tested against human liver microsomes containing CYP1A1, CYP1A2, CYP2C9, CYP2C19, CYP2E1, and CYP3A4 .
Control tissue validation: Include positive and negative control tissues in experiments. Human bronchial epithelial tissue serves as an excellent positive control, while human liver tissue provides a negative control for CYP2A13 expression .
Preimmune serum controls: Always include negative controls where the primary antibody is replaced with preimmune serum from the same species to identify non-specific binding .
Antibody concentration optimization: Perform dilution series experiments to determine optimal antibody concentration that maximizes specific signal while minimizing background. Published studies have used 1:600 dilution for rabbit anti-CYP2A13 antiserum in immunohistochemistry applications .
For effective colocalization studies of CYP2A13 with pancreatic islet cell markers, researchers should follow this methodological approach:
Tissue preparation:
Use 5-μm thick paraffin sections from properly fixed pancreatic tissue
Perform standard deparaffinization and hydration procedures
Apply antigen retrieval with antigen unmasking solution to expose epitopes
Blocking and primary antibody incubation:
Block non-specific binding with normal goat serum
Prepare a mixture of rabbit anti-CYP2A13 antiserum (1:600 dilution) with either:
Mouse anti-proinsulin C-peptide IgG (1:200 dilution) for β-cell identification, or
Mouse anti-glucagon IgG (1:2000 dilution) for α-cell identification
Incubate sections with the antibody mixture in a humidified chamber overnight at 4°C
Secondary antibody application:
This protocol facilitates the visualization of CYP2A13 expression (green fluorescence) in relation to either insulin-producing β-cells or glucagon-producing α-cells (red fluorescence), allowing precise cellular localization within pancreatic islets.
Differentiating between CYP2A13.1 (wild-type) and CYP2A13.2 (variant) requires a combination of molecular and functional approaches:
Genetic characterization:
mRNA quantification:
Functional enzyme activity assays:
Compare metabolic activity toward known CYP2A13 substrates:
NNK (4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone)
N-nitrosomethylphenylamine
N,N-dimethylaniline
2′-methoxyacetophenone
Hexamethylphosphoramide
The CYP2A13.2 variant typically exhibits 20-40% lower activity compared to CYP2A13.1 with these substrates
Promoter activity analysis:
These methodological approaches provide comprehensive characterization of the functional differences between wild-type and variant forms, crucial for studies investigating the role of CYP2A13 polymorphisms in disease susceptibility.
For rigorous immunohistochemical analysis of CYP2A13, researchers should incorporate the following controls:
Antibody specificity controls:
Preimmune serum control: Replace primary anti-CYP2A13 antibody with preimmune serum from the same animal species to assess non-specific binding
Omission control: Exclude primary antibody to identify secondary antibody non-specific binding
Absorption control: Pre-incubate antibody with the immunizing peptide to confirm epitope specificity
Tissue controls:
Positive tissue control: Include human bronchial epithelial tissue sections, known to express high levels of CYP2A13
Negative tissue control: Include human liver sections, which have minimal or no CYP2A13 expression
Technical replicates: Perform staining on several adjacent sections to confirm reproducibility of findings
Method validation:
Implementing these controls ensures reliable and reproducible immunohistochemical data, essential for accurate characterization of CYP2A13 expression patterns.
Discrepancies between mRNA and protein expression levels are common in CYP2A13 research and require careful interpretation:
Potential mechanisms for discrepancies:
Post-transcriptional regulation: miRNAs or RNA-binding proteins may affect translation efficiency
Protein stability differences: Variant forms like CYP2A13.2 may have altered protein stability
Allelic expression imbalance: The CYP2A132 allele shows approximately 40% lower expression than CYP2A131
Tissue-specific translational regulation: Some tissues may have mechanisms that prevent efficient translation of certain mRNAs
Methodological approaches to resolve discrepancies:
Implement parallel mRNA and protein quantification in the same samples
Use allele-specific expression analysis in heterozygous samples
Assess protein half-life through pulse-chase experiments
Examine both global and tissue-specific post-transcriptional regulatory mechanisms
Interpretation framework:
In CYP2A13*2 carriers, lower protein levels may reflect both decreased transcription (due to the 26-nucleotide promoter deletion) and potentially altered protein stability
Functional significance should be assessed through enzyme activity assays, as expression levels alone may not fully explain phenotypic differences
Consider the tissue context, as the relationship between mRNA and protein may vary between tissues
This comprehensive approach allows researchers to better understand the biological significance of observed discrepancies and their implications for CYP2A13 function in different tissues.
Several technical challenges can affect experiments using FITC-conjugated antibodies for CYP2A13 detection:
Tissue autofluorescence:
Challenge: Pancreatic tissue contains endogenous fluorescent compounds that may interfere with FITC signal
Solution: Implement autofluorescence quenching protocols using reagents like Sudan Black B or perform spectral unmixing during image acquisition
Signal fading during microscopy:
Challenge: FITC is prone to photobleaching during extended imaging sessions
Solution: Use anti-fade mounting media, minimize exposure time, and consider alternative more photostable fluorophores like Alexa Fluor 488 for critical applications
Cross-talk between fluorescent channels:
Challenge: In dual labeling experiments (e.g., FITC and rhodamine), signal bleed-through can occur
Solution: Acquire single-labeled control samples for each fluorophore, implement proper filter sets, and use sequential scanning in confocal microscopy
Fixation-induced epitope masking:
Antibody internalization in live-cell applications:
Challenge: FITC-conjugated antibodies may undergo internalization or capping in live-cell experiments
Solution: Use Fab fragments instead of whole IgG molecules or perform fixation before antibody application
Implementing these technical solutions ensures more reliable and reproducible results when using FITC-conjugated antibodies in CYP2A13 research.
CYP2A13 antibodies are powerful tools for investigating smoking-related disease mechanisms:
Tissue-specific expression mapping:
Use immunohistochemistry with CYP2A13 antibodies to map expression in tissues susceptible to smoking-related diseases
Correlate expression patterns with histopathological changes in smokers versus non-smokers
The selective expression of CYP2A13 in human respiratory epithelium aligns with the observation that most smoking-related lung cancers are bronchogenic
Cellular co-localization with carcinogen adducts:
Employ dual immunofluorescence combining CYP2A13 antibodies with antibodies against DNA adducts (e.g., NNK-derived adducts)
This approach can demonstrate spatial relationships between CYP2A13 expression and carcinogen damage
Variant expression analysis in patient cohorts:
Mechanistic studies in pancreatic pathology:
The selective expression of CYP2A13 in pancreatic α-islet cells suggests these cells may be specifically vulnerable to tobacco toxicants
CYP2A13 antibodies can help track islet cell-specific damage in relation to smoking exposure
A recent finding showed an inactive CYP2A13 variant is associated with decreased pancreatic cancer susceptibility, supporting this enzyme's role in pancreatic carcinogenesis
These applications highlight how CYP2A13 antibodies contribute to understanding the tissue-specific mechanisms of smoking-related diseases at the cellular and molecular level.
Demonstrating the functional significance of CYP2A13 requires multi-faceted experimental approaches:
Ex vivo tissue metabolism studies:
Incubate fresh tissue slices (bronchial epithelium or pancreatic tissue) with NNK or other CYP2A13 substrates
Measure metabolite formation using LC-MS/MS
Compare metabolism rates with CYP2A13 expression levels determined by immunohistochemistry
Published research has shown that levels of CYP2A13 protein expression correlate with rates of lung microsomal NNK metabolic activation
Genetic manipulation in relevant cell models:
Use CRISPR/Cas9 to generate CYP2A13 knockouts or to introduce specific variants (e.g., CYP2A13*2)
Engineer isogenic cell lines differing only in CYP2A13 status
Assess differences in carcinogen metabolism, DNA damage, and cellular transformation
Tissue-specific transgenic mouse models:
Generate mice expressing human CYP2A13 in specific tissues (e.g., respiratory epithelium or pancreatic α-cells)
Expose to NNK or tobacco smoke and assess tissue-specific pathology
Compare wild-type CYP2A13 with variant forms to assess functional significance of polymorphisms
Correlation of enzyme activity with expression patterns:
These complementary approaches provide robust evidence for the functional significance of CYP2A13 in specific tissues and its role in smoking-related disease pathogenesis.
When investigating CYP2A13 polymorphisms in disease association studies, researchers should consider:
Comprehensive genetic characterization:
Functional impact assessment:
Tissue-specific consequences:
Exposure stratification:
Interaction with other genetic factors:
Assess interactions with polymorphisms in other genes involved in carcinogen metabolism
Consider haplotype analysis rather than single-SNP association
Evaluate gene-gene interactions that might modify CYP2A13-associated risk
These methodological considerations enhance the rigor of disease association studies investigating CYP2A13 polymorphisms and improve the interpretability of findings.
Sample preparation significantly impacts CYP2A13 antibody detection, with tissue-specific considerations:
Fixation effects:
Formalin fixation: The standard 5-μm thick paraffin sections from formalin-fixed tissues require antigen retrieval for optimal CYP2A13 detection
Fresh-frozen tissue: May preserve antigenicity better but can compromise morphological integrity
Fixation duration: Prolonged fixation can cause excessive protein cross-linking, potentially masking CYP2A13 epitopes
Antigen retrieval optimization:
Tissue-specific challenges:
Pancreatic tissue: Contains high levels of proteolytic enzymes that can degrade antibodies or antigens if not properly inactivated
Lung tissue: May contain endogenous peroxidases requiring quenching steps if using HRP-based detection
Both tissues: May contain lipofuscin causing autofluorescence that interferes with FITC detection
Section thickness considerations:
Optimization of these parameters for each tissue type ensures reliable and reproducible CYP2A13 detection, critical for accurate comparative studies across different tissues or pathological states.
Distinguishing between highly similar CYP2A subfamily members requires specialized approaches:
Antibody-based discrimination:
Epitope selection: Use C-terminal peptide sequences that differ most between CYP2A13, CYP2A6, and CYP2A7
Validated antibodies: The antibody described in the literature was raised against amino acid residues 369-377 of CYP2A13, a region with maximal sequence divergence from other CYP2A proteins
Validation testing: Confirm antibody specificity against recombinant CYP2A proteins and liver microsomes containing CYP2A6
Combined protein and mRNA analysis:
Parallel assessment: Perform immunohistochemistry alongside in situ hybridization with isoform-specific probes
RT-PCR validation: Use isoform-specific primers to confirm expression of specific CYP2A genes
Quantitative comparison: Compare relative expression levels of different CYP2A members
Functional discrimination approaches:
Substrate selectivity: Use substrates with differential selectivity for CYP2A13 versus CYP2A6
Inhibitor profiling: Apply isoform-selective inhibitors to distinguish enzymatic activities
Correlation analysis: Compare activity profiles with protein expression patterns
Knockout/knockdown validation:
siRNA targeting: Use isoform-specific siRNAs to selectively suppress individual CYP2A members
Antibody validation: Confirm reduced staining following specific knockdown
This approach validates both antibody specificity and the identity of the detected protein
These methodological approaches provide robust discrimination between highly similar CYP2A subfamily members, essential for accurate characterization of their tissue-specific expression and roles in xenobiotic metabolism.
Integrating CYP2A13 antibody staining with complementary techniques provides comprehensive tissue analysis:
Multi-omics integration:
Laser capture microdissection of CYP2A13-positive cells followed by RNA-seq or proteomics
Spatial transcriptomics to correlate CYP2A13 protein expression with local gene expression profiles
Integration of CYP2A13 immunohistochemistry with metabolomics data from the same tissue regions
Advanced imaging approaches:
Multiplexed immunofluorescence: Combine CYP2A13 antibodies with markers for cell proliferation, apoptosis, and DNA damage
Mass cytometry imaging: Use metal-tagged antibodies for highly multiplexed spatial analysis
3D tissue reconstruction: Serial section immunostaining to create three-dimensional maps of CYP2A13 expression
Functional correlation studies:
Combine CYP2A13 immunohistochemistry with in situ activity assays
Correlate CYP2A13 expression with local NNK-DNA adduct formation
Map CYP2A13 expression in relation to histopathological changes in smokers' tissues
Clinical-pathological correlations:
Integrate CYP2A13 expression data with patient smoking history, genotype, and clinical outcomes
Create tissue microarrays from patient cohorts for high-throughput analysis
Correlate CYP2A13 expression patterns with response to therapy or disease progression
This integrated approach provides mechanistic insights beyond what can be achieved with CYP2A13 antibody staining alone, facilitating a deeper understanding of CYP2A13's role in disease pathogenesis.
Appropriate quantification methods for CYP2A13 immunofluorescence ensure reliable and reproducible data:
Cell-level quantification:
Positive cell counting: Determine the percentage of CYP2A13-positive cells within defined tissue regions
In pancreatic islets, this approach revealed that CYP2A13-expressing cells constitute a small portion of total islet cells, mainly located in the peripheral region
Signal intensity measurement: Quantify fluorescence intensity per cell using digital image analysis
Tissue-level quantification:
Region of interest (ROI) analysis: Define anatomical regions (e.g., islet periphery vs. center) and measure mean fluorescence intensity
Spatial distribution mapping: Create heat maps of CYP2A13 expression across tissue sections
Colocalization analysis: For dual-label studies, calculate Pearson's or Mander's coefficients to quantify colocalization with cell-type markers
Technical considerations:
Background correction: Subtract autofluorescence measured in negative control sections
Normalization: Use reference standards or housekeeping proteins for cross-sample normalization
Dynamic range optimization: Ensure image acquisition settings avoid signal saturation while maintaining sensitivity
Statistical analysis:
Apply appropriate statistical tests based on data distribution
For studies comparing genotypes or exposure groups, consider power analysis to determine required sample sizes
Account for biological variability by analyzing multiple sections per sample and multiple samples per condition
These quantification approaches provide objective measures of CYP2A13 expression patterns, facilitating meaningful comparisons across experimental conditions and between different studies.