CYP4F12 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
PBS with 0.1% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze-thaw cycles.
Lead Time
Typically, we can ship products within 1-3 business days after receiving your order. Delivery time may vary depending on the purchase method and location. Please consult your local distributor for specific delivery times.
Synonyms
CYP4F12; UNQ568/PRO1129; Cytochrome P450 4F12; CYPIVF12
Target Names
CYP4F12
Uniprot No.

Target Background

Function
CYP4F12 is a cytochrome P450 monooxygenase involved in the metabolism of endogenous polyunsaturated fatty acids (PUFAs). It functions by utilizing molecular oxygen to insert one oxygen atom into a substrate, while reducing the second oxygen atom into a water molecule. This process requires two electrons supplied by NADPH via cytochrome P450 reductase (CPR; NADPH-ferrihemoprotein reductase). CYP4F12 primarily catalyzes the hydroxylation of carbon hydrogen bonds, exhibiting a preference for the omega-2 position. It metabolizes (5Z,8Z,11Z,14Z)-eicosatetraenoic acid (arachidonate) to 18-hydroxy arachidonate. Additionally, CYP4F12 catalyzes the epoxidation of double bonds in PUFAs such as docosapentaenoic and docosahexaenoic acids. While CYP4F12 has low omega-hydroxylase activity towards leukotriene B4 and arachidonate, it plays a role in the metabolism of xenobiotics. Furthermore, it catalyzes the hydroxylation of the antihistamine drug ebastine.
Gene References Into Functions
  1. Studies suggest that human and marmoset P450 3A4/90 and 4F12 in livers or small intestines play significant roles in terfenadine t-butyl hydroxylation. Marmosets could serve as a model for humans during first-pass extraction of terfenadine and related substrates. PMID: 28436281
  2. Research indicates that HCV infection induces the expression of CYP4F12 protein, which binds to the HCV replication complex to facilitate viral replication. PMID: 26845356
  3. Evidence suggests that CYP4F8 and CYP4F12 catalyze epoxidation of 22:6n-3 and 22:5n-3, and CYP4F8 omega3-hydroxylation of 22:5n-6. PMID: 16112640
Database Links

HGNC: 18857

OMIM: 611485

KEGG: hsa:66002

STRING: 9606.ENSP00000321821

UniGene: Hs.131459

Protein Families
Cytochrome P450 family
Subcellular Location
Endoplasmic reticulum membrane. Microsome membrane.
Tissue Specificity
Expressed in small intestine, liver, colon and heart.

Q&A

What is CYP4F12 and what are its primary functions in normal tissues?

CYP4F12 (Cytochrome P450, Family 4, Subfamily F, Polypeptide 12) is a member of the cytochrome P450 enzyme family that plays crucial roles in metabolism and homeostasis. In normal tissues, CYP4F12 is expressed across multiple cell types, with particularly notable expression in certain epithelial tissues. Based on research findings, CYP4F12 appears to be involved in maintaining normal cellular adhesion and regulating cell migration processes, suggesting it plays a role in tissue integrity and wound healing under physiological conditions. The protein is notably expressed in non-cancerous tissues at higher levels than in their malignant counterparts, indicating a potential tumor-suppressive role in normal cellular functioning .

What applications is the CYP4F12 antibody most commonly used for in research?

The CYP4F12 antibody is primarily utilized in several key research applications:

  • Western Blotting (WB): For detecting and quantifying CYP4F12 protein expression in cell or tissue lysates

  • Enzyme-Linked Immunosorbent Assay (ELISA): For quantitative measurement of CYP4F12 levels in biological samples

  • Immunohistochemistry (IHC): For visualizing and localizing CYP4F12 expression in tissue sections

These applications are particularly valuable for researchers investigating CYP4F12's role in cancer biology, especially in head and neck squamous cell carcinoma (HNSC) and other malignancies where CYP4F12 expression is frequently dysregulated . The antibody's capability to function across multiple experimental platforms makes it versatile for comprehensive protein expression studies, from quantitative analysis to spatial localization within tissues.

How should researchers validate CYP4F12 antibody specificity before experimental use?

Proper validation of CYP4F12 antibody specificity requires a multi-step approach:

  • Positive and negative control samples: Use tissues or cell lines known to express high levels of CYP4F12 (such as BICR22, PE/CA-PJ15, SNU-1066, and CAL-33 cell lines) as positive controls and those with minimal expression (such as HSC-3, YD-15, PE/CA-PJ49, YD18, FaDu, PE/CA-PJ41, and SCC-9) as negative controls .

  • Western blot analysis: Confirm a single band of the expected molecular weight (approximately 59-60 kDa for CYP4F12).

  • Knockdown/overexpression validation: Compare antibody staining in cells with siRNA-mediated knockdown (using validated sequences like "GAAGCCAGCAUAUCCUCCATT" or "GAAGCCAGCATATCCTCCATT") versus cells overexpressing CYP4F12 via plasmid transfection .

  • Peptide competition assay: Pre-incubate the antibody with the immunizing peptide (such as recombinant Human Cytochrome P450 4F12 protein fragments) to confirm signal elimination in subsequent staining .

  • Cross-reactivity assessment: If working with multiple species, verify specificity across human, mouse, and rat samples as appropriate for the specific antibody clone being used .

This comprehensive validation workflow ensures experimental results accurately reflect true CYP4F12 expression patterns and reduces the risk of non-specific binding artifacts.

How can researchers effectively study CYP4F12's role in regulating cell migration and EMT in cancer models?

To effectively investigate CYP4F12's role in regulating cell migration and epithelial-mesenchymal transition (EMT) in cancer models, researchers should implement the following methodological approach:

  • Cell line selection: Choose appropriate HNSC cell lines with differential CYP4F12 expression. Based on published data, FaDu and SCC-9 cells exhibit low endogenous CYP4F12 expression, making them suitable for overexpression studies, while cells like BICR22 or CAL-33 with higher expression are appropriate for knockdown experiments .

  • Genetic manipulation techniques:

    • For overexpression: Transfect cells with CYP4F12 plasmid using Lipofectamine 3000 following established protocols (1.6 × 10^5 cells/mL for FaDu; 1.1 × 10^5 cells/mL for SCC-9 in 6-well plates) .

    • For knockdown: Utilize validated siRNAs targeting CYP4F12 (sequences: "GAAGCCAGCAUAUCCUCCATT" or "GAAGCCAGCATATCCTCCATT") .

  • Migration assays: Perform wound healing or transwell migration assays 48 hours post-transfection to assess the impact of CYP4F12 expression on cell motility.

  • Cell-matrix adhesion assessment: Utilize cell adhesion assay kits to quantify adhesion capacity changes following CYP4F12 manipulation. Follow established protocols with 1 × 10^5 cells per well in pre-coated 96-well plates, measuring adhesion rate as (OD-CYP4F12 − OD-CYP4F12-blank) / (OD-Control − OD-Control-blank) × 100% .

  • EMT marker analysis: Perform western blotting to analyze key EMT markers including:

    • E-cadherin (epithelial marker)

    • N-cadherin (mesenchymal marker)

    • Vimentin (mesenchymal marker)

    • α-catenin (cell adhesion protein)

  • Statistical validation: Apply appropriate statistical tests (Student's t-tests for continuous variables; Pearson's chi-square or Fisher's exact tests for categorical variables) with significance thresholds of p < 0.05, *p < 0.01, and **p < 0.001 .

This comprehensive approach enables researchers to establish causative relationships between CYP4F12 expression levels and cancer cell migration/EMT processes, providing mechanistic insights into CYP4F12's tumor-suppressive functions.

What are the key considerations when interpreting CYP4F12 expression data across different cancer types?

When interpreting CYP4F12 expression data across different cancer types, researchers should consider several critical factors:

  • Tissue-specific expression patterns: CYP4F12 demonstrates variable baseline expression across different tissues. Data from The Cancer Genome Atlas (TCGA) and Cancer Cell Line Encyclopedia (CCLE) databases show that CYP4F12 is downregulated in multiple cancer types compared to matched normal tissues, including breast invasive carcinoma (BRCA), cholangiocarcinoma (CHOL), colon adenocarcinoma (COAD), head and neck squamous cell carcinoma (HNSC), and several others .

  • Cell line heterogeneity within cancer types: Even within HNSC, CYP4F12 expression varies significantly between cell lines. For example, BICR22, PE/CA-PJ15, SNU-1066, and CAL-33 show high expression, while HSC-3, YD-15, PE/CA-PJ49, YD18, FaDu, PE/CA-PJ41, and SCC-9 exhibit minimal expression . This heterogeneity must be accounted for when designing experiments and interpreting results.

  • Correlation with clinicopathological variables: When analyzing CYP4F12 expression in cancer, researchers should assess correlations with:

    • Tumor stage and grade

    • Patient demographics

    • Viral infection status (e.g., HPV in HNSC, where HPV-positive tumors show higher CYP4F12 expression)

    • Molecular subtypes of the cancer

    • Immune infiltration patterns

  • Technical considerations for detection:

    • Antibody selection based on the specific application (WB, ELISA, IHC)

    • Appropriate antibody concentration optimization

    • Selection of proper epitope regions (e.g., AA 265-524) based on research questions

    • Cross-reactivity assessment when studying multiple species

  • Data normalization and statistical approaches: Implement proper normalization techniques, particularly when comparing across different datasets (e.g., TCGA, GEO, CCLE), and use appropriate statistical methods for the specific analysis being performed.

By systematically addressing these considerations, researchers can generate more reliable interpretations of CYP4F12 expression patterns and their biological significance across cancer types.

How does CYP4F12 expression correlate with immune infiltration in tumor microenvironments?

CYP4F12 expression demonstrates significant correlations with immune infiltration patterns in tumor microenvironments, particularly in head and neck squamous cell carcinoma (HNSC). When investigating this relationship, researchers should focus on the following methodological approaches:

  • Immune correlation analysis: Divide patient cohorts into high and low CYP4F12 expression groups based on median expression values. Use computational packages such as ggstatsplot (v0.11.1) and pheatmap (v1.0.12) for immune correlation and immune cell biomarker analyses .

  • Immune score calculation: Employ the immunedeconv R package, which integrates six algorithms for comprehensive immune score evaluation. Specifically, use the TIMER algorithm to quantitatively compare immune scores between high and low CYP4F12 expression groups .

  • Immune checkpoint analysis: Extract and analyze expression values of key immune checkpoint genes including SIGLEC15, TIGIT, CD274 (PD-L1), HAVCR2, PDCD1 (PD-1), CTLA4, LAG3, and PDCD1LG2 (PD-L2) to assess correlations with CYP4F12 expression. Visualize these relationships using R packages GGplot2 and pheatmap .

  • Immunotherapy response prediction: Apply the Tumor Immunodeficiency and Exclusion (TIDE) algorithm to predict potential responses to immune checkpoint blockade (ICB) therapy based on CYP4F12 expression levels, providing insights into CYP4F12's value as a predictive biomarker for immunotherapy efficacy .

  • Validation in tissue samples: Confirm computational findings through multiplex immunohistochemistry on patient tumor samples, correlating CYP4F12 protein expression with immune cell infiltration markers to establish tissue-level validation of bioinformatic predictions.

This systematic approach enables researchers to comprehensively characterize the relationship between CYP4F12 expression and tumor immune microenvironment, potentially revealing new opportunities for targeted or combination immunotherapeutic strategies in CYP4F12-expressing cancers.

What are the optimal conditions for using CYP4F12 antibodies in western blotting experiments?

When utilizing CYP4F12 antibodies for western blotting experiments, researchers should consider the following optimized protocol to achieve reliable and reproducible results:

  • Sample preparation:

    • Extract total protein from tissues or cultured cells using RIPA buffer supplemented with protease inhibitors

    • Quantify protein concentration using BCA or Bradford assay

    • Load 20-30 μg of total protein per lane for cell lines; 40-50 μg for tissue samples

    • Include both positive controls (tissues/cells with known high CYP4F12 expression like BICR22 or CAL-33) and negative controls (low expression lines like FaDu or SCC-9)

  • Gel electrophoresis and transfer:

    • Separate proteins on 10% SDS-PAGE gels (CYP4F12 has a molecular weight of approximately 59-60 kDa)

    • Transfer to PVDF membrane (preferred over nitrocellulose for enhanced protein retention)

    • Confirm transfer efficiency with reversible Ponceau S staining

  • Antibody incubation parameters:

    • Blocking: 5% non-fat dry milk in TBST, 1 hour at room temperature

    • Primary antibody: Use rabbit polyclonal anti-CYP4F12 antibody at 1:1000 dilution in 5% BSA/TBST, incubate overnight at 4°C

    • Washing: 3 × 10 minutes with TBST

    • Secondary antibody: Anti-rabbit IgG (H+L) (DyLight™ 800 4X PEG Conjugate) at 1:5000 dilution in 5% non-fat milk/TBST, incubate for 1 hour at room temperature

    • Final washing: 3 × 10 minutes with TBST

  • Detection and analysis:

    • Visualize using infrared fluorescence imaging systems (e.g., LI-COR Odyssey)

    • Include β-actin (Abcam, ab8226) as loading control

    • Quantify band intensity using ImageJ or similar software

    • Calculate relative CYP4F12 expression normalized to loading control

  • Troubleshooting tips:

    • For weak signals: Increase antibody concentration to 1:500 or extend primary antibody incubation to 48 hours at 4°C

    • For high background: Increase washing times or duration and ensure thorough blocking

    • For multiple bands: Validate antibody specificity using peptide competition assays

This optimized protocol ensures high specificity and sensitivity when detecting CYP4F12 in western blotting applications while minimizing background and non-specific binding issues.

How should researchers design experiments to investigate CYP4F12's potential as a diagnostic biomarker?

To rigorously investigate CYP4F12's potential as a diagnostic biomarker, researchers should implement a comprehensive experimental design with the following methodological components:

This systematic approach provides robust evidence for CYP4F12's potential as a diagnostic biomarker while addressing critical aspects of biomarker development including sensitivity, specificity, reproducibility, and clinical applicability.

What are the critical steps in optimizing immunohistochemistry protocols for CYP4F12 detection in tissue samples?

Optimizing immunohistochemistry (IHC) protocols for CYP4F12 detection in tissue samples requires careful attention to several critical parameters to ensure specific staining and reproducible results:

  • Tissue preparation and processing:

    • Fix tissue samples in 10% neutral buffered formalin for 24-48 hours

    • Process and embed in paraffin following standard protocols

    • Section at 4-5 μm thickness onto positively charged slides

    • Include known positive controls (tissues with documented CYP4F12 expression) and negative controls (omission of primary antibody)

  • Antigen retrieval optimization:

    • Compare heat-induced epitope retrieval methods:

      • Citrate buffer (pH 6.0): 20 minutes at 95-98°C

      • EDTA buffer (pH 9.0): 20 minutes at 95-98°C

    • Optimize retrieval time based on tissue type and fixation duration

    • Allow slides to cool slowly to room temperature (20-30 minutes)

  • Antibody selection and validation:

    • Choose antibodies targeting specific epitopes (such as AA 265-524) with validated specificity

    • Confirm cross-reactivity profile matches experimental species (human/mouse/rat)

    • Validate using positive and negative control tissues and cell lines with known CYP4F12 expression profiles

  • Primary antibody optimization:

    • Test dilution series (1:100, 1:250, 1:500, 1:1000) to determine optimal concentration

    • Compare overnight incubation at 4°C versus 1-hour incubation at room temperature

    • Evaluate different antibody diluents to minimize background staining

  • Detection system selection:

    • For low abundance targets: Use high-sensitivity detection systems (e.g., polymer-based or tyramide signal amplification)

    • For quantitative analysis: Consider chromogenic detection with 3,3'-diaminobenzidine (DAB)

    • For multiplexing: Implement fluorescent detection with appropriate controls for autofluorescence

  • Counterstaining and interpretation:

    • Use appropriate counterstains (hematoxylin for chromogenic IHC, nuclear stains for fluorescent IHC)

    • Establish clear scoring criteria (percentage of positive cells, staining intensity)

    • Implement digital image analysis when possible for quantitative assessment

  • Validation with alternative detection methods:

    • Confirm IHC findings with orthogonal techniques such as western blotting or qRT-PCR

    • Compare subcellular localization patterns across methodologies

By systematically optimizing these parameters, researchers can develop robust IHC protocols for reliable CYP4F12 detection in various tissue types, enabling accurate assessment of expression patterns in normal and pathological specimens.

How should researchers interpret discrepancies between CYP4F12 mRNA and protein expression data?

When encountering discrepancies between CYP4F12 mRNA and protein expression data, researchers should implement a systematic analytical approach:

This comprehensive approach acknowledges that mRNA-protein discrepancies are biologically meaningful and can reveal important regulatory mechanisms controlling CYP4F12 expression, rather than simply representing technical artifacts that should be dismissed.

What methodological approaches are recommended for studying CYP4F12's role in epithelial-mesenchymal transition?

To rigorously investigate CYP4F12's role in epithelial-mesenchymal transition (EMT), researchers should implement a comprehensive methodological framework:

  • Cell model selection and manipulation:

    • Choose appropriate cell lines with differential CYP4F12 expression

    • Implement both gain-of-function (overexpression) and loss-of-function (siRNA knockdown) approaches

    • For overexpression: Transfect cells with CYP4F12 plasmid using optimized transfection conditions (e.g., Lipofectamine 3000 following manufacturer's protocol)

    • For knockdown: Utilize validated siRNA sequences targeting CYP4F12 (e.g., "GAAGCCAGCAUAUCCUCCATT" or "GAAGCCAGCATATCCTCCATT")

  • EMT marker analysis panel:

    • Epithelial markers: E-cadherin, α-catenin, ZO-1, claudins, occludin

    • Mesenchymal markers: N-cadherin, vimentin, fibronectin

    • EMT transcription factors: SNAIL, SLUG, ZEB1, ZEB2, TWIST

    • Evaluate these markers using:

      • Western blotting with validated antibodies (e.g., E-cadherin: Abcam ab40772, N-cadherin: Abcam ab76011, vimentin: Abcam ab92547, α-catenin: Abcam ab51032)

      • Immunofluorescence for subcellular localization

      • qRT-PCR for mRNA expression changes

  • Functional assays to assess EMT phenotypes:

    • Cell-matrix adhesion assays using standardized protocols (e.g., cell adhesion assay kit, measuring adhesion rate as (OD-CYP4F12 − OD-CYP4F12-blank)/(OD-Control − OD-Control-blank) × 100%)

    • Migration assays (wound healing, transwell)

    • Invasion assays (Matrigel-coated transwell)

    • Cell morphology assessment (phase-contrast microscopy)

  • Signaling pathway analysis:

    • Investigate canonical EMT-inducing pathways (TGF-β, Wnt/β-catenin, Notch)

    • Employ pathway inhibitors to determine which signaling cascade is affected by CYP4F12

    • Assess phosphorylation status of key signaling proteins via western blotting

  • In vivo validation:

    • Analyze patient samples for correlations between CYP4F12 expression and EMT markers

    • Consider xenograft models with CYP4F12-manipulated cell lines to assess metastatic potential

  • Mechanistic investigation:

    • Perform chromatin immunoprecipitation (ChIP) to identify potential interactions between CYP4F12 and EMT-related gene promoters

    • Conduct co-immunoprecipitation experiments to identify protein-protein interactions

    • Implement RNA-seq analysis to identify global transcriptional changes following CYP4F12 manipulation

This comprehensive methodological approach enables researchers to establish causative relationships between CYP4F12 expression and EMT processes, providing mechanistic insights into how CYP4F12 influences cancer cell behavior and potentially identifying novel therapeutic strategies targeting this pathway.

How can researchers effectively integrate CYP4F12 expression data with clinical outcomes for biomarker development?

To effectively integrate CYP4F12 expression data with clinical outcomes for biomarker development, researchers should implement the following methodological framework:

  • Cohort selection and data collection:

    • Design prospective studies with adequate sample size determined by power analysis

    • Collect comprehensive clinicopathological data including:

      • Demographic information (age, sex, ethnicity)

      • Tumor characteristics (stage, grade, molecular subtypes)

      • Treatment history (surgery, radiation, chemotherapy, immunotherapy)

      • Follow-up data (recurrence, progression, survival)

    • Establish clear inclusion/exclusion criteria to minimize confounding variables

  • Multi-platform CYP4F12 assessment:

    • Analyze CYP4F12 at both mRNA and protein levels:

      • RNA-seq or qRT-PCR for transcript quantification

      • IHC or western blotting for protein expression using validated antibodies

    • Implement standardized scoring systems:

      • For IHC: H-score or Allred scoring system

      • For mRNA: Normalized expression values (FPKM, TPM, or ΔCt)

  • Statistical analysis framework:

    • Stratify patients based on CYP4F12 expression (high vs. low using median or optimal cutoff)

    • Perform univariate analysis:

      • Kaplan-Meier survival curves with log-rank tests

      • Cox proportional hazards models for time-to-event outcomes

    • Conduct multivariate analysis:

      • Adjust for established prognostic factors

      • Test for interactions between CYP4F12 and other clinical variables

    • Assess predictive value:

      • Generate ROC curves and calculate AUCs to evaluate diagnostic potential

      • Determine sensitivity, specificity, positive predictive value, and negative predictive value

  • External validation strategy:

    • Validate findings in independent cohorts:

      • Public databases (TCGA, GEO)

      • Multi-institutional collaborations

    • Compare performance against established biomarkers

    • Assess consistency across different cancer types and subtypes

  • Translational potential assessment:

    • Evaluate practical implementation factors:

      • Assay reproducibility and inter-laboratory variability

      • Cost-effectiveness compared to existing biomarkers

      • Ease of integration into existing clinical workflows

    • Develop clear guidelines for CYP4F12 testing and interpretation

    • Consider combinatorial biomarker approaches incorporating CYP4F12

  • Therapeutic relevance exploration:

    • Correlate CYP4F12 expression with:

      • Response to specific therapeutic modalities

      • Immune infiltration patterns for immunotherapy prediction

      • Resistance mechanisms to conventional treatments

This comprehensive integration approach enables robust biomarker development, moving CYP4F12 from association studies toward clinically actionable biomarkers with clear implementation pathways for improving patient outcomes.

What are the key technical considerations when analyzing CYP4F12 in different experimental models?

When analyzing CYP4F12 in different experimental models, researchers must address several critical technical considerations to ensure robust and reproducible results:

  • Species-specific differences:

    • Sequence homology: Human CYP4F12 shows approximately 80-85% sequence homology with mouse and rat orthologs, requiring careful antibody selection for cross-species studies

    • Expression patterns: CYP4F12 expression profiles vary between species, necessitating proper baseline characterization in each model organism

    • Functional divergence: Enzymatic activity and substrate specificity may differ across species, requiring functional validation in each model

  • Cell line selection strategy:

    • Baseline expression profiling: Characterize endogenous CYP4F12 expression across multiple cell lines before selecting appropriate models (e.g., BICR22, PE/CA-PJ15, SNU-1066, and CAL-33 show high expression, while HSC-3, YD-15, FaDu, and SCC-9 show low expression)

    • Genetic background considerations: Account for molecular subtypes and genetic alterations in cell lines that might influence CYP4F12 function

    • Growth conditions: Standardize culture conditions, as CYP4F12 expression may be affected by serum levels, cell density, and passage number

  • Detection method optimization:

    • Antibody validation: For each experimental model, validate antibody specificity using positive/negative controls and knockdown/overexpression systems

    • For Western blotting: Optimize protein extraction methods (RIPA vs. NP-40 buffers), blocking agents (milk vs. BSA), and antibody dilutions specifically for each model system

    • For IHC/ICC: Adjust fixation protocols, antigen retrieval methods, and detection systems based on tissue/cell type

  • Genetic manipulation approaches:

    • Overexpression systems: Consider both transient transfection and stable expression systems, with appropriate empty vector controls

    • Knockdown strategies: Compare siRNA (for transient effects) versus shRNA or CRISPR-Cas9 (for stable manipulation)

    • Rescue experiments: Implement rescue experiments to confirm specificity of observed phenotypes

  • Model-specific quantification methods:

    • For in vitro models: Standard curve-based absolute quantification

    • For tissue samples: Consider tissue heterogeneity and implement microdissection when necessary

    • For in vivo models: Account for organ-specific expression patterns and potential compensatory mechanisms

  • Environmental and experimental variables:

    • Timing considerations: Standardize harvest times as CYP4F12 may exhibit circadian regulation

    • Drug treatments: Account for potential induction or suppression of CYP4F12 by experimental compounds

    • Microenvironmental factors: Consider oxygen tension, pH, and cell-cell interactions that may affect CYP4F12 expression

By systematically addressing these technical considerations, researchers can develop robust protocols for analyzing CYP4F12 across diverse experimental models, enhancing data reproducibility and facilitating meaningful cross-model comparisons.

How can researchers troubleshoot common issues encountered in CYP4F12 antibody-based experiments?

When troubleshooting common issues in CYP4F12 antibody-based experiments, researchers should implement a systematic approach to identify and resolve technical challenges:

Weak or No Signal in Western Blotting

Potential causes and solutions:

  • Insufficient protein loading:

    • Increase protein amount (30-50 μg per lane)

    • Verify protein concentration using multiple measurement methods (BCA, Bradford)

  • Ineffective protein transfer:

    • Optimize transfer conditions (time, voltage, buffer composition)

    • Confirm transfer efficiency with reversible staining (Ponceau S)

    • Consider semi-dry vs. wet transfer based on CYP4F12's molecular weight (~59-60 kDa)

  • Suboptimal antibody concentration:

    • Test concentration series (1:250 to 1:2000)

    • Extend primary antibody incubation (overnight at 4°C)

    • Try different antibody clones or those targeting different epitopes (AA 265-524 vs. others)

  • Degraded protein:

    • Use fresh samples with complete protease inhibitor cocktail

    • Avoid repeated freeze-thaw cycles of protein lysates

High Background in Immunohistochemistry

Potential causes and solutions:

  • Insufficient blocking:

    • Extend blocking time (2-3 hours)

    • Test alternative blocking agents (5% BSA, 10% normal serum, commercial blockers)

  • Non-specific antibody binding:

    • Pre-absorb antibody with tissue powder

    • Increase wash duration and number of washes

    • Optimize antibody dilution (typically start with 1:100 to 1:500)

  • Endogenous peroxidase activity:

    • Implement effective peroxidase quenching (3% H₂O₂, 10-15 minutes)

    • For fluorescent detection, use appropriate autofluorescence quenching protocols

  • Tissue fixation issues:

    • Optimize fixation time and conditions

    • Compare different antigen retrieval methods (citrate pH 6.0 vs. EDTA pH 9.0)

Multiple Bands in Western Blotting

Potential causes and solutions:

  • Cross-reactivity with related proteins:

    • Verify antibody specificity through peptide competition assays

    • Test antibodies targeting different epitopes of CYP4F12

    • Compare results with gene knockdown controls

  • Protein degradation products:

    • Use freshly prepared samples with complete protease inhibitors

    • Reduce sample processing time and temperature

  • Post-translational modifications:

    • Run additional experiments to identify modifications (phosphorylation, glycosylation)

    • Use specific enzymatic treatments to remove modifications and confirm band identity

Inconsistent Results Between Experiments

Potential causes and solutions:

  • Antibody lot variation:

    • Record lot numbers and test new lots against previous standards

    • Create internal reference standards for normalization between experiments

  • Cell/tissue heterogeneity:

    • Standardize cell confluency in culture experiments

    • Implement tissue microdissection for heterogeneous samples

  • Protocol drift:

    • Develop detailed standard operating procedures

    • Control technical variables (incubation times, temperatures, buffer compositions)

Discrepancies Between Antibody-Based and mRNA Results

Potential causes and solutions:

  • Post-transcriptional regulation:

    • Implement time-course experiments to detect temporal disconnects

    • Investigate potential microRNA regulation

  • Antibody specificity issues:

    • Validate with multiple antibodies targeting different epitopes

    • Confirm with orthogonal methods (mass spectrometry)

    • Implement genetic validation through overexpression or knockdown

By systematically applying these troubleshooting approaches, researchers can overcome common technical challenges in CYP4F12 antibody experiments, leading to more consistent and reliable results across various experimental platforms.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.