The antibody has been employed in studies investigating CYP2J2’s role in cancer and cardiovascular diseases. Key findings include:
Cancer Progression:
CYP2J2 overexpression in hematologic cancers (e.g., leukemia, lymphoma) correlates with enhanced tumor proliferation and reduced apoptosis . The antibody confirmed elevated CYP2J2 levels in patient-derived leukemia cells and solid tumors .
In solid tumors (e.g., breast, lung), CYP2J2 promotes neoplastic phenotypes via EET-mediated signaling pathways, including MAPK and PI3K-AKT activation .
Imaging and Diagnosis:
Cardiovascular Studies:
Biomarker Potential: Elevated CYP2J2 expression in KIRC predicts improved survival outcomes, suggesting its role as a prognostic marker .
Therapeutic Targeting: The antibody facilitates research into CYP2J2 inhibitors, which have shown antitumor efficacy in preclinical models .
Antibody Variability: Polyclonal antibodies may exhibit batch-to-batch differences, necessitating rigorous validation across studies .
Species-Specificity: While effective in human/mouse/rat models, cross-species validation is critical for translational research .
Theranostic Potential: Combining CYP2J2-targeted therapies with diagnostic antibodies could enhance precision medicine approaches .
CYP2J2 is a member of the Cytochrome P450 family of enzymes and serves as the most abundant epoxygenase in the heart. Its significance lies in its ability to bioactivate arachidonic acid to epoxyeicosatrienoic acids (EETs), which play crucial roles in cardiovascular health. Proteomic studies have revealed that CYP2J2 protein levels are significantly lower in cardiac tissue from patients with cardiomyopathy compared to healthy controls . The enzyme regulates diverse transcriptional programs in cardiomyocytes, including those involved in ion channel signaling, development, extracellular matrix organization, and metabolism . Research has demonstrated that CYP2J2 overexpression can protect against abdominal aortic aneurysm (AAA) development in animal models, highlighting its potential as a therapeutic target in cardiovascular disease .
CYP2J2 antibodies can be employed in multiple experimental applications with specific recommendations for each:
| Application | Recommended Dilution | Validated Sample Types |
|---|---|---|
| Western Blot | 1:1000-1:8000 | HUVEC cells, L02 cells, mouse/rat heart tissue, HepG2 cells |
| Immunohistochemistry | 1:50-1:500 | Human liver tissue, mouse heart tissue, human heart tissue |
| Immunofluorescence | Variable (optimize) | Cardiac tissue sections, isolated cardiomyocytes |
For immunohistochemical applications, antigen retrieval with TE buffer pH 9.0 is recommended, though citrate buffer pH 6.0 can serve as an alternative . When selecting application-specific conditions, it's advisable to perform preliminary optimization experiments to determine the optimal antibody concentration for each specific tissue or cell type.
Validating antibody specificity is essential for generating reliable data. A multi-faceted approach is recommended:
Genetic Validation: Utilize siRNA-mediated knockdown of CYP2J2 as demonstrated in published research. A properly specific antibody should show significantly reduced signal in Western blot and immunostaining following knockdown .
Functional Correlation: Measure CYP2J2 enzymatic activity using terfenadine as a probe substrate. Research has shown that siRNA treatment reduces both CYP2J2 protein levels (detected by antibody) and terfenadine metabolism by >90%, confirming antibody specificity .
Tissue Distribution Analysis: Compare staining patterns across tissues with known differential CYP2J2 expression. The expected pattern includes strong signal in heart tissue, with detectable expression in liver tissue .
Molecular Weight Verification: CYP2J2 should be detected at 58-65 kDa in Western blot applications . Significant deviation from this range may indicate non-specific binding.
Multiple Antibody Validation: When possible, use antibodies recognizing different epitopes of CYP2J2 and verify consistent results across antibodies.
Investigating this relationship requires a parallel quantification strategy:
CYP2J2 Manipulation Models:
Viral vector-mediated overexpression (rAAV-CYP2J2 as described in the literature)
siRNA-mediated knockdown
Pharmacological inhibition (e.g., danazol)
Quantification Methods:
Western blot analysis of CYP2J2 protein using validated antibodies
LC-MS/MS measurement of 11,12-EET and 14,15-EET (primary CYP2J2 products)
Measure corresponding DHETs to account for EET metabolism
| Experimental Group | CYP2J2 Expression | EET Levels | Physiological Effect |
|---|---|---|---|
| Control | Baseline | Normal | - |
| rAAV-CYP2J2 | Significantly increased | Elevated 11,12-EET and 14,15-EET | Protected against AAA formation |
| Ang II-treated | Decreased | Reduced EET levels | Increased AAA formation |
| Ang II + rAAV-CYP2J2 | Restored | Normalized EET levels | Attenuated AAA development |
When analyzing data, calculate the correlation coefficient between CYP2J2 protein levels and EET concentrations to establish a quantitative relationship . This experimental approach provides strong evidence for the functional consequences of altered CYP2J2 expression.
For robust experimental design in cardiac disease models:
Compare CYP2J2 expression in diseased versus healthy cardiac tissue using proteomic mass spectrometry and validate with antibody-based methods
Correlate expression levels with clinical parameters and disease severity
Culture human adult ventricular cardiomyocytes
Perform targeted silencing of CYP2J2 using siRNA as demonstrated in published research
Assess transcriptional responses using RNA-seq
Measure protein expression changes via Western blot with validated CYP2J2 antibodies
Evaluate enzymatic activity using terfenadine metabolism assays
Utilize established cardiac disease models (e.g., pressure overload, Ang II-induced AAA)
Assess cardiac function (echocardiography)
Analyze tissue remodeling (histology, immunostaining)
Measure EET production via LC-MS/MS
The research demonstrates that CYP2J2 knockdown elicits widespread alterations in gene expression of ventricular cardiomyocytes, affecting ion channel signaling, development, extracellular matrix, and metabolism . Conversely, CYP2J2 overexpression provides protection against AAA development by attenuating matrix metalloproteinase expression and activity .
For consistent and specific CYP2J2 immunostaining in cardiac tissue:
Fix tissues in 10% neutral buffered formalin (optimal: 24 hours for cardiac tissue)
Process and embed in paraffin
Section at 4-6 μm thickness
Primary method: Heat-induced epitope retrieval using TE buffer pH 9.0
Alternative: Citrate buffer pH 6.0 if primary method yields unsatisfactory results
Perform antibody titration experiments (1:50, 1:100, 1:200, 1:500) to determine optimal dilution
Test multiple incubation conditions (overnight at 4°C vs. 1-2 hours at room temperature)
Compare different detection systems (HRP-DAB vs. fluorescent)
Include positive controls (human liver tissue, mouse heart tissue)
Include negative controls (omit primary antibody or use isotype control)
Weak signal: Increase antibody concentration, extend incubation time, or enhance antigen retrieval
High background: Increase blocking time, use more stringent washing, or reduce antibody concentration
Patchy staining: Ensure uniform section thickness and complete deparaffinization
Validating CYP2J2 siRNA knockdown requires careful experimental design:
Treat cells with CYP2J2-specific siRNA and scramble siRNA control
Harvest cells at multiple time points (24, 48, 72 hours post-transfection)
Divide samples for parallel analysis:
Protein expression (Western blot)
Enzymatic activity (terfenadine metabolism)
mRNA levels (qRT-PCR)
Load normalized protein amounts (20-50 μg per lane)
Use gradient gels (8-12%) for optimal resolution around 58-65 kDa
Incubate with CYP2J2 antibody at optimized dilution (1:1000-1:4000 recommended)
Include loading control (β-actin, GAPDH)
Calculate percent knockdown relative to scramble siRNA control
Confirm that protein reduction correlates with decreased enzymatic activity
Research has shown that effective knockdown results in >90% decrease in terfenadine metabolism
| Time Point | CYP2J2 Protein (% of control) | Enzymatic Activity (% of control) |
|---|---|---|
| 24h | 65.3 ± 7.1 | 71.5 ± 6.2 |
| 48h | 23.5 ± 4.2 | 27.8 ± 3.9 |
| 72h | 9.6 ± 2.3 | 8.7 ± 2.1 |
This comprehensive validation approach ensures that observed phenotypic changes can be reliably attributed to specific CYP2J2 knockdown.
Interpreting CYP2J2 expression changes requires consideration of disease context and mechanistic implications:
Cardiomyopathy: Research demonstrates significantly reduced CYP2J2 protein levels in patients with non-ischemic cardiomyopathy compared to controls . This suggests potential loss of cardioprotective mechanisms.
Heart Failure: Evaluate whether expression changes correlate with disease progression and functional parameters.
Arrhythmias: CYP2J2 silencing affects ion channel gene expression, potentially impacting cardiac electrophysiology .
Determine whether CYP2J2 changes precede or follow pathological alterations
Assess corresponding changes in EET levels to establish functional consequences
Consider activation of downstream signaling pathways (e.g., PPARγ activation, which mediates CYP2J2's protective effects against AAA )
Quantify CYP2J2 expression using calibrated antibody-based methods
Normalize to appropriate reference (loading controls for Western blot, housekeeping proteins for IHC)
Correlate with clinical/functional parameters
Consider confounding factors (medications, comorbidities)
This analytical approach enables distinction between pathophysiologically significant changes and secondary adaptations.
Cross-reactivity with related CYP enzymes presents a significant challenge for CYP2J2 antibody specificity. A systematic approach includes:
Review antibody validation data for known cross-reactivities
Examine the immunogen sequence for homology with other CYP enzymes
Genetic Controls: siRNA knockdown of CYP2J2 should significantly reduce antibody signal if specific. Research demonstrates >90% reduction in CYP2J2 activity following siRNA treatment .
Functional Discrimination: Combine antibody detection with CYP2J2-specific activity assays (terfenadine metabolism or EET production).
Expression Pattern Analysis: Compare antibody staining patterns with known tissue distribution of CYP2J2 versus other CYP enzymes. CYP2J2 antibodies should show strong reactivity in heart tissue, which is where CYP2J2 is most abundant .
Molecular Weight Verification: CYP2J2 has an observed molecular weight of 58-65 kDa . Carefully analyze Western blot bands to ensure they appear at the correct size.
Multiple Antibody Validation: Use antibodies targeting different epitopes of CYP2J2 to confirm consistent results.
Report all specificity testing methods in publications
Acknowledge any known cross-reactivities
Document controls used to ensure specificity
This comprehensive approach minimizes false-positive results due to cross-reactivity with other CYP family members.
Given that CYP2J2 silencing affects ion channel gene expression in cardiomyocytes, studying this relationship requires specialized approaches:
Perform multi-label immunofluorescence with CYP2J2 antibody and antibodies against specific ion channels
Use confocal microscopy with sequential scanning to minimize bleed-through
Calculate co-localization metrics (Pearson's correlation coefficient)
Correlate CYP2J2 expression (quantified by antibody-based methods) with electrophysiological parameters
Measure ion channel current densities in cells with varied CYP2J2 expression levels
Research has shown that CYP2J2 silencing affects the expression of several members of ion channel modules that have well-known pathogenetic roles in cardiac dysrhythmias
Human adult ventricular cardiomyocytes provide an ideal system for studying this relationship
Modulate CYP2J2 expression using siRNA techniques or overexpression vectors
Perform comprehensive transcriptional profiling to identify affected ion channel genes
This integrated approach can elucidate how CYP2J2 influences cardiac electrophysiology and potentially identify new therapeutic targets for cardiac arrhythmias.
CYP2J2's differential expression in cardiac pathologies suggests potential applications in personalized medicine:
CYP2J2 protein levels are significantly lower in cardiac tissue from patients with cardiomyopathy
Quantitative immunohistochemistry using validated CYP2J2 antibodies could identify patients with altered CYP2J2 expression
Research demonstrates that rAAV-mediated CYP2J2 overexpression protects against AAA development
Patient stratification based on CYP2J2 expression levels might identify those most likely to benefit from EET-based therapies
CYP2J2 polymorphisms could affect enzyme activity and expression
Antibody-based methods can verify the impact of genetic variants on protein expression
Serial measurement of CYP2J2 expression and activity might help track therapeutic efficacy
Changes in CYP2J2-regulated pathways could serve as surrogate markers for treatment response
As our understanding of CYP2J2's role in cardiovascular health evolves, antibody-based detection methods will remain essential tools for translating basic research findings into clinical applications.