The ALOX12 antibody is a polyclonal or monoclonal antibody designed to detect and study the expression of arachidonate 12-lipoxygenase (ALOX12), an enzyme critical in lipid metabolism. ALOX12 catalyzes the oxidation of polyunsaturated fatty acids (PUFAs) like arachidonic acid and docosahexaenoic acid (DHA), producing bioactive lipids such as 12(S)-hydroperoxyeicosatetraenoic acid (12(S)-HpETE) and specialized pro-resolving mediators (SPMs) . These metabolites regulate inflammation, platelet activation, and immune responses, making the antibody indispensable in studying disease mechanisms and therapeutic interventions .
The ALOX12 antibody has been employed to study its therapeutic implications in MDS. AZA (5-azacytidine), a DNA hypomethylating agent, upregulates ALOX12 expression by demethylating its promoter region. Overexpression of ALOX12 in MDS cells reduces oxidative stress susceptibility and inhibits tumor progression, suggesting its role as a tumor suppressor .
In a murine model of lung IRI, ALOX12 and its metabolite 12-HETE were upregulated, contributing to endothelial ferroptosis. ALOX12 knockdown or inhibition with ML355 mitigated ferroptosis and neutrophil extracellular trap (NET) formation, highlighting its therapeutic potential in lung transplantation .
High ALOX12 expression in colorectal cancer (CRC) correlates with increased tumor immune infiltration and higher expression of immune checkpoints (e.g., PD-1, PD-L1, CTLA-4). This suggests that ALOX12 may predict responsiveness to immune checkpoint blockade (ICB) therapy .
This ALOX12 antibody targets an enzyme that catalyzes the regio- and stereo-specific incorporation of molecular oxygen into free and esterified polyunsaturated fatty acids. This process generates lipid hydroperoxides, which can be further reduced to corresponding hydroxy species. Specifically, it converts arachidonate ((5Z,8Z,11Z,14Z)-eicosatetraenoate) to the bioactive lipid (12S)-hydroperoxyeicosatetraenoate/(12S)-HPETE. (12S)-HPETE production regulates various biological processes, including platelet activation. The enzyme also catalyzes the epoxidation of polyunsaturated fatty acid double bonds, such as in (14S)-hydroperoxy-docosahexaenoate/(14S)-HPDHA, forming (13S,14S)-epoxy-DHA. Furthermore, it participates in the sequential oxidation of DHA ((4Z,7Z,10Z,13Z,16Z,19Z)-docosahexaenoate) to produce specialized pro-resolving mediators (SPMs) like resolvin D5 ((7S,17S)-diHPDHA) and (7S,14S)-diHPDHA. These SPMs actively downregulate immune responses and exhibit anti-aggregation properties with platelets. Additionally, ALOX12 transforms leukotriene A4 (LTA4) into the bioactive lipids lipoxin A4 (LXA4) and lipoxin B4 (LXB4) through a multi-step process. LXA4 and LXB4 are vasoactive, and LXA4 modulates neutrophil function. The enzyme can also peroxidize linoleate ((9Z,12Z)-octadecadienoate) to (13S)-hydroperoxyoctadecadienoate/(13S-HPODE). Given its roles in regulating vascular endothelial growth factor (VEGF) and integrin beta-1 expression (both implicated in tumorigenesis), ALOX12 exhibits protumorigenic properties. Its contribution to cell survival is significant, influencing both proliferation and apoptosis prevention in vascular smooth muscle cells.
The following studies highlight the diverse roles of ALOX12:
ALOX12, also known as 12S-LOX or Platelet-type lipoxygenase 12, is an enzyme that catalyzes the regio- and stereo-specific incorporation of molecular oxygen into polyunsaturated fatty acids, generating lipid hydroperoxides . Its primary function involves converting arachidonate ((5Z,8Z,11Z,14Z)-eicosatetraenoate) to (12S)-hydroperoxyeicosatetraenoate/(12S)-HPETE, a bioactive lipid that regulates various biological processes including platelet activation .
ALOX12 plays multiple roles in cellular function:
Regulates inflammation through production of lipid mediators
Participates in the generation of specialized pro-resolving mediators (SPMs) like resolvin D5
Transforms leukotriene A4 into bioactive lipoxins (LXA4 and LXB4)
Influences cell survival by preventing apoptosis in vascular smooth muscle cells
Affects tumor progression through regulation of VEGF and integrin beta-1 expression
Cellular location studies indicate that ALOX12 is primarily located in the cytoplasm and cytosol, with membrane association stimulated by EGF . It is prominently expressed in vascular smooth muscle cells but can be found in various tissue types depending on physiological conditions .
Selecting the right ALOX12 antibody requires consideration of several technical factors:
Target specificity: Choose antibodies targeting specific epitopes based on your research focus. For instance, C-terminal antibodies (like those targeting the 618-650 amino acid region) offer good specificity for human ALOX12 .
Cross-reactivity profile: Consider species reactivity - the antibody described in the search results demonstrates reactivity with human, mouse, and rat ALOX12, making it versatile for comparative studies .
Application compatibility: Match the antibody to your intended application. The ALOX12 antibody described has been validated for Western Blot (WB), Immunofluorescence (IF), Flow Cytometry (FC), and Immunohistochemistry-Paraffin (IHC-P) .
Antibody format: Consider format based on your detection system. The described antibody is supplied in PBS with 0.09% sodium azide and purified through protein A column followed by peptide affinity purification .
| Application | Recommended Dilution | Notes |
|---|---|---|
| Immunofluorescence | 1:100 | For cellular localization studies |
| Western Blot | 1:1000 | For protein expression analysis |
| IHC-P | 1:10~50 | For tissue section analysis |
| Flow Cytometry | 1:10~50 | For single-cell analysis |
The choice between polyclonal (like the one described) and monoclonal antibodies depends on your need for broad epitope recognition versus high specificity for a single epitope .
Proper storage and handling of ALOX12 antibodies is crucial for maintaining their activity and specificity:
Short-term storage: Maintain refrigerated at 2-8°C for up to 2 weeks .
Long-term storage: For extended periods, store at -20°C in small aliquots to prevent freeze-thaw cycles that can degrade antibody quality .
Aliquoting strategy: Upon receipt, consider dividing the antibody into small working aliquots before freezing to minimize freeze-thaw cycles. Each freeze-thaw cycle can reduce antibody activity by approximately 10-15%.
Temperature transitions: When using refrigerated or frozen antibodies, allow them to equilibrate to room temperature (18-25°C) before opening to prevent condensation that could introduce contaminants .
Working dilutions: Prepare working dilutions on the day of use whenever possible. If storage of diluted antibody is necessary, store at 4°C and use within 24 hours for optimal performance.
Contamination prevention: Use sterile techniques when handling antibodies, and add sodium azide (0.02%-0.05%) to diluted antibodies if they must be stored for multiple days (though note that azide can interfere with some detection systems).
Following these guidelines will help ensure consistent and reliable antibody performance across experiments.
Proper experimental controls are essential for validating ALOX12 antibody results:
Cell lines or tissues known to express ALOX12 (such as vascular smooth muscle cells)
Recombinant ALOX12 protein at known concentrations
Previously validated samples with established ALOX12 expression patterns
Samples from ALOX12 knockout models
Cell lines with confirmed absence of ALOX12 expression
Primary antibody omission control
Isotype control (using non-specific rabbit IgG at the same concentration)
Peptide competition/blocking experiments using the immunizing peptide (618-650 amino acids for the C-terminal antibody described)
siRNA knockdown of ALOX12 to demonstrate reduced signal
Loading controls for Western blot (β-actin, GAPDH)
Tissue architecture/cellular morphology validation in IHC/IF applications
Standardization curves when using quantitative applications like ELISA
Include multiple controls in parallel with your experimental samples to ensure the observed signals truly represent ALOX12 detection rather than non-specific binding or technical artifacts.
Several approaches can be used for ALOX12 quantification, each with specific methodological considerations:
Include a concentration gradient of recombinant ALOX12 as a standard curve
Employ chemiluminescent or fluorescent detection systems
Use densitometry software for band intensity quantification
Always normalize to an appropriate loading control
Commercial sandwich ELISA kits for ALOX12 offer detection ranges of approximately 0.312-20 ng/mL
Ensure samples fall within the linear range of the standard curve
Consider using log-log graph plotting for more accurate interpretation
Multiply final concentration by dilution factor if samples were diluted
Follow detailed manufacturer protocols for washing and incubation times to minimize background
Use the recommended 1:10-50 dilution for flow cytometry applications
Include calibration beads with known quantities of fluorophores
Compare mean/median fluorescence intensity across samples
Use appropriate gating strategies to exclude debris and non-specific signals
Semi-quantitative scoring based on staining intensity
Digital image analysis using specialized software
Consider subcellular localization patterns (cytoplasmic, membrane-associated)
For all methods, validation across multiple techniques will strengthen confidence in your quantitative assessments of ALOX12 expression.
ALOX12's central function in lipid mediator synthesis can be investigated through several sophisticated approaches:
Use liquid chromatography-mass spectrometry (LC-MS/MS) to profile ALOX12-dependent metabolites, particularly (12S)-HPETE and downstream products
Complement antibody-based detection of ALOX12 with functional metabolomics to correlate enzyme presence with activity
Apply stable isotope labeling with arachidonic acid to track conversion to specific products
Measure ALOX12 enzymatic activity using purified enzyme and substrate conversion assays
Monitor oxygen consumption rates during catalysis using specialized electrodes
Utilize ALOX12 antibodies for immunoprecipitation followed by activity assessment of the purified protein complex
Investigate ALOX12's involvement in generating specialized pro-resolving mediators (SPMs) like resolvin D5 ((7S,17S)-diHPDHA) that regulate immune responses
Study the conversion of leukotriene A4 to lipoxins (LXA4 and LXB4)
Examine epoxidation reactions catalyzed by ALOX12 on substrates like (14S)-HPDHA
Use co-immunoprecipitation with ALOX12 antibodies to identify protein binding partners
Apply proximity ligation assays to visualize ALOX12 interactions with other pathway components in situ
Investigate membrane association dynamics, particularly in response to EGF stimulation
These methodologies can be combined with genetic manipulation (overexpression, knockdown, site-directed mutagenesis) of ALOX12 to comprehensively characterize its role in lipid mediator synthesis pathways.
Several technical challenges can affect ALOX12 antibody performance:
ALOX12 shares sequence homology with other lipoxygenase family members
Solution: Validate antibody specificity using ALOX12 knockout controls and peptide competition assays
Consider using antibodies targeting unique regions like the C-terminal domain (618-650 aa)
ALOX12 function can be regulated by phosphorylation and other modifications that might mask epitopes
Solution: Use multiple antibodies targeting different epitopes
Consider native versus denaturing conditions in your detection system
ALOX12 protein detection does not necessarily correlate with enzymatic activity
Solution: Complement antibody detection with functional assays measuring product formation
Investigate regulatory mechanisms affecting enzyme activity independent of expression
ALOX12 can shuttle between cytosolic and membrane-associated states
Solution: Use cell fractionation approaches combined with immunoblotting
Apply confocal microscopy with appropriate subcellular markers in IF applications
Formalin fixation can mask epitopes in IHC-P applications
Solution: Optimize antigen retrieval methods (heat-induced or enzymatic)
Test different fixation protocols for optimal epitope preservation
Semi-quantitative nature of many antibody-based techniques
Solution: Include standard curves with recombinant proteins
Use multiple methodologies (WB, ELISA, IF) to confirm expression patterns
Addressing these challenges requires rigorous experimental design and appropriate controls tailored to your specific research question and biological system.
ALOX12 has been implicated in cancer biology through several mechanisms that can be investigated using specific methodologies:
ALOX12 regulates vascular endothelial growth factor (VEGF) expression, an angiogenic factor involved in tumor survival and metastasis
Methodology: Use ALOX12 antibodies in chromatin immunoprecipitation (ChIP) studies to investigate transcriptional regulation
Correlate ALOX12 expression with VEGF levels in tumor tissues using dual immunostaining approaches
ALOX12 affects integrin beta-1 expression, known to influence tumor cell migration and proliferation
Methodology: Employ scratch assays, transwell migration, and invasion assays in cells with modulated ALOX12 expression
Use live-cell imaging with fluorescently labeled ALOX12 antibodies to track protein localization during migration
ALOX12 plays a role in preventing apoptosis in vascular smooth muscle cells
Methodology: Assess apoptotic markers (caspase activation, PARP cleavage) in relation to ALOX12 expression
Apply flow cytometry with dual staining for ALOX12 and apoptotic markers
ALOX12-derived metabolites may influence tumor microenvironment
Methodology: Perform targeted lipidomics on tumor samples
Correlate metabolite profiles with ALOX12 expression patterns
Analyze ALOX12 expression in patient samples using the validated antibody dilutions for IHC-P (1:10-50)
Correlate expression patterns with clinical outcomes, staging, and therapy response
Develop tissue microarrays for high-throughput analysis
| Application | Methodology | Key Parameters |
|---|---|---|
| Tumorigenic potential | Colony formation assays | ALOX12 expression vs. colony number/size |
| Angiogenesis | Tube formation assay | Correlation with VEGF expression |
| Metastatic capacity | Tail vein injection models | ALOX12 inhibition/overexpression |
| Drug sensitivity | Cytotoxicity assays | ALOX12 expression vs. therapy response |
These research approaches can help unravel the complex relationships between ALOX12 enzymatic activity, lipid mediator production, and cancer progression pathways.
ALOX12's involvement in inflammation and immunity can be investigated through these methodological approaches:
ALOX12 participates in generating specialized pro-resolving mediators (SPMs) that actively down-regulate immune responses
Methodology: Use LC-MS/MS to identify and quantify SPMs in different inflammatory phases
Correlate SPM levels with ALOX12 expression and activity using specific antibodies
ALOX12-derived metabolites affect platelet activation and aggregation
Methodology: Perform platelet-leukocyte aggregation assays while manipulating ALOX12 expression/activity
Apply flow cytometry with ALOX12 antibodies (dilution 1:10-50) to correlate expression with functional outcomes
Lipoxin A4 (LXA4), produced through ALOX12 activity, may regulate neutrophil function
Methodology: Assess neutrophil chemotaxis, phagocytosis, and NETosis in relation to ALOX12 expression
Use immunofluorescence (dilution 1:100) to visualize ALOX12 localization during neutrophil activation
ALOX12 metabolites may influence macrophage phenotype switching
Methodology: Analyze M1/M2 marker expression in relation to ALOX12 levels
Apply single-cell approaches to correlate ALOX12 with macrophage subpopulations
ALOX12 activity may modulate cytokine production patterns
Methodology: Perform multiplex cytokine assays in systems with varied ALOX12 expression
Use ELISA-based ALOX12 quantification alongside cytokine measurements
Apply ALOX12 antibodies for tissue analysis in models of acute and chronic inflammation
Methodology: Use the recommended IHC-P dilutions (1:10-50) to assess expression during different inflammatory phases
Compare wild-type with ALOX12-deficient systems to establish causality
When designing these experiments, consider temporal dynamics, as ALOX12's role may shift between pro-inflammatory and pro-resolving functions depending on the inflammatory phase and tissue microenvironment.
When facing inconsistent ALOX12 antibody performance, implement this systematic troubleshooting approach:
Verify antibody quality through Western blotting against recombinant ALOX12
Confirm recognition of the expected molecular weight (~75.7 kDa)
Test against positive control samples with known ALOX12 expression
For protein extraction, evaluate different lysis buffers that preserve ALOX12 structure
Consider native versus denaturing conditions based on your application
For tissue samples, optimize fixation protocols (duration, fixative type) for IHC applications
Standardize antibody incubation times and temperatures
For ELISA applications, ensure consistent washing procedures as incomplete washing affects precision
Control environmental humidity (ideally <60%) which can affect performance of immunoassays
Test a range of antibody dilutions around the recommended values:
Implement antigen retrieval techniques for fixed tissues (heat-induced or enzymatic)
Test multiple retrieval buffers (citrate, EDTA, Tris) at different pH values
Adjust permeabilization conditions for cellular applications
Confirm findings using complementary techniques (e.g., validate IHC results with Western blot)
Consider orthogonal approaches like mRNA quantification via qPCR
Use genetic manipulation (siRNA knockdown) to confirm antibody specificity
Record and compare lot numbers across experiments
Maintain consistent sources of primary and secondary antibodies
Create internal reference samples that can be used across experiments
By systematically addressing these variables, you can identify the source of inconsistency and establish reliable protocols for ALOX12 detection across your experimental systems.