The LTC4S Antibody, HRP conjugated is a rabbit-derived polyclonal antibody designed to detect Leukotriene C4 Synthase (LTC4S), a key enzyme in the biosynthesis of leukotrienes. HRP (Horseradish Peroxidase) conjugation enables enzymatic detection in assays such as Western blot (WB) and enzyme-linked immunosorbent assay (ELISA). Below is a detailed breakdown of its properties and applications.
Target Specificity:
The antibody binds to distinct regions of LTC4S, including the N-terminal domain (29–55 aa) and a central hydrophilic loop (36–51 aa), ensuring precise detection of the enzyme .
HRP Function:
HRP catalyzes oxidation reactions, enabling colorimetric or chemiluminescent detection in assays. This conjugation enhances sensitivity for low-abundance protein detection .
Cross-Reactivity:
While primarily validated for human LTC4S, some variants may show limited cross-reactivity with other GST enzymes (e.g., mGST-II), requiring careful validation .
Purpose: Quantify LTC4S protein levels in cell lysates or tissue homogenates.
Protocol:
Example:
In studies of bronchial mast cells, WB has confirmed LTC4S overexpression in asthma, correlating with leukotriene-mediated inflammation .
Purpose: Measure soluble LTC4S in biological fluids (e.g., serum, bronchoalveolar lavage).
Advantages:
Example:
ELISA using HRP-conjugated LTC4S antibodies has been employed to monitor enzyme activity in response to inflammatory stimuli .
Asthma:
Mast Cells vs. Eosinophils:
Bronchial mast cells exhibit 50-fold higher LTC4S immunoreactivity than eosinophils, as shown via immunohistochemistry and digital image analysis .
Therapeutic Implications:
Reduced LTC4S-positive mast cells correlate with clinical improvement in treated asthma patients .
Atherosclerosis:
Enzyme Complex Formation:
LTC4S interacts with 5-lipoxygenase (5-LO) via its second hydrophilic loop (aa 90–113) and with FLAP at the N-terminal hydrophobic region .
Regulation:
Phosphorylation at Ser-36 by RPS6KB1 inhibits LTC4S activity, modulating leukotriene synthesis .
Leukotriene C4 Synthase (LTC4S) is a critical enzyme that catalyzes the conjugation of leukotriene A4 with reduced glutathione to form leukotriene C4 . LTC4S belongs to the MAPEG (Membrane Associated Proteins in Eicosanoid and Glutathione metabolism) family and plays a pivotal role in the biosynthesis of cysteinyl leukotrienes, which are potent biological compounds derived from arachidonic acid . The significance of studying LTC4S stems from its involvement in inflammatory conditions, particularly bronchial asthma and anaphylaxis, making it an important target for immunological and pharmacological research . LTC4S localizes to the nuclear envelope and adjacent endoplasmic reticulum, providing insights into the subcellular organization of leukotriene biosynthesis pathways .
HRP-conjugated LTC4S antibodies primarily serve several key applications in research settings. These antibodies are extensively used in Enzyme-Linked Immunosorbent Assays (ELISA) for quantitative detection of LTC4S in various biological samples . They are also employed in Western Blotting (WB) procedures for protein detection and semi-quantitative analysis . HRP conjugation provides a significant advantage as it enables direct detection without requiring secondary antibodies, thereby simplifying experimental workflows and potentially reducing background noise. The conjugation to HRP allows for colorimetric or chemiluminescent detection methods, making these antibodies versatile tools in research investigating inflammatory pathways, respiratory diseases, and immunological responses .
Antibody specificity to different amino acid regions of LTC4S significantly influences experimental design considerations. LTC4S antibodies targeting specific epitopes, such as AA 36-51 or AA 29-55 from the N-terminal region, offer distinct advantages for different applications . When designing experiments, researchers must consider that:
Epitope accessibility may vary depending on protein conformation in different sample preparation methods
N-terminal targeting antibodies (such as AA 29-55) may be preferable for detecting full-length protein in Western blotting applications
The choice between different epitope-specific antibodies should align with the research question and whether specific domains or the entire protein need to be detected
Cross-reactivity profiles differ between antibodies targeting different regions, affecting species compatibility in comparative studies
Selecting the appropriate epitope-specific antibody ensures optimal sensitivity and specificity for the intended application, reducing the risk of false negatives or positives in experimental results.
LTC4S antibodies can detect the target protein across various biological sample types, enabling diverse research applications. Compatible sample types include serum, plasma, cell culture supernatants, and other biological fluids . When working with tissue samples, proper extraction and preparation protocols are essential to ensure epitope preservation and accessibility. For cell-based research, the A-549 cell line has been validated for LTC4S detection using Western blot analysis . Each sample type requires specific optimization of protocols, including dilution factors, blocking reagents, and incubation conditions. For quantitative applications using ELISA, researchers should be aware that the LTC4S ELISA kits employ a sandwich ELISA approach, where samples are pipetted into antibody-precoated wells, followed by detection with biotin-conjugated antibodies and streptavidin-HRP . This methodological awareness helps researchers select appropriate sample types and preparation techniques for their specific research questions.
Optimizing signal-to-noise ratios with HRP-conjugated LTC4S antibodies requires a systematic approach addressing multiple experimental parameters. Based on research experiences, the following methodological considerations are crucial:
First, antibody dilution optimization is essential, with documented working ranges for LTC4S antibodies typically between 1:500 to 1:2000 for Western blot applications . Researchers should perform dilution series experiments to identify the minimum antibody concentration that yields reproducible signals. Second, blocking optimization using 3-5% BSA or milk protein in TBS-T buffer helps minimize non-specific binding, though BSA may be preferable for phospho-specific applications. Third, implementing stringent washing protocols (4-5 washes of 5-10 minutes each) with TBS-T significantly reduces background signal.
For membrane handling, transferring proteins from 12-15% SDS-PAGE gels (considering LTC4S's calculated molecular weight of 17kDa) with optimized transfer conditions (typically 100V for 60-90 minutes) improves detection sensitivity . Additionally, chemiluminescent substrate selection should be based on the expected abundance of LTC4S in samples—standard ECL for abundant targets and high-sensitivity substrates for low-expression scenarios. Finally, optimizing exposure time during imaging prevents signal saturation while capturing specific bands, with incremental exposures (5, 15, 30, 60 seconds) recommended to identify the optimal detection window.
Addressing cross-reactivity concerns with LTC4S antibodies requires comprehensive validation strategies and careful experimental design. Despite manufacturer specifications, researchers should conduct independent validation when working with non-human samples. For instance, while certain LTC4S antibodies show broad cross-reactivity across species including human, cow, dog, guinea pig, horse, bat, monkey, pig, mouse, rat, rabbit, and zebrafish , the actual performance may vary.
Sequence homology analysis should precede experimental work, comparing the antibody's target epitope sequence across species of interest. For antibodies targeting AA 36-51 or AA 29-55, alignment analysis of these specific regions provides predictive insights into potential cross-reactivity . Validation protocols should include positive controls from each species alongside human samples for direct comparison. When cross-reactivity is observed but signal strength varies, researchers should adjust loading concentrations proportionally to normalize for detection sensitivity differences.
Alternative approaches include using multiple antibodies targeting different epitopes to confirm findings or employing species-specific secondary antibodies in non-conjugated primary antibody systems. For truly quantitative cross-species comparisons, standard curves using recombinant proteins from each species should be developed, allowing for calibrated measurements that account for epitope-antibody affinity differences across species.
The performance differential of LTC4S antibodies between native and denatured protein states represents a critical consideration for experimental design. LTC4S antibodies targeting amino acid regions 36-51 or 29-55 demonstrate distinct performance characteristics across different application contexts .
For researchers working with native protein conformations, antibodies targeting more exposed regions of the protein (often N-terminal domains) generally perform better than those targeting transmembrane or structurally constrained domains. When transitioning between applications requiring different protein states, validation experiments comparing antibody performance under native versus denaturing conditions are strongly recommended, as sensitivity and specificity profiles may differ substantially. Additionally, fixation methods in immunohistochemistry or immunofluorescence applications can significantly impact epitope accessibility, requiring protocol optimization specific to the antibody's target region.
Implementing multiplex detection systems with HRP-conjugated LTC4S antibodies requires careful methodological planning to address several technical challenges. The primary consideration is signal discrimination when multiple HRP-conjugated antibodies are used simultaneously. Since all HRP conjugates generate similar detection signals, researchers must employ sequential detection protocols with complete stripping between rounds or utilize spatial separation techniques.
For Western blot applications, a practical approach involves membrane cutting based on molecular weight markers, allowing separate incubation with different antibodies. LTC4S, with its calculated molecular weight of 17kDa , can be distinctly separated from most other proteins of interest. Alternative approaches include using antibodies from different host species conjugated to different reporter enzymes (e.g., HRP and alkaline phosphatase) with compatible substrates that generate distinguishable signals.
In ELISA-based multiplex systems, researchers should consider:
Cross-reactivity between detection antibodies and non-target analytes
Optimization of capture antibody concentrations to achieve comparable sensitivity across all targets
Development of balanced washing protocols that adequately remove non-specific binding without compromising specific signals
Careful selection of blocking reagents that minimize background without interfering with specific antibody-antigen interactions
When multiplexing involves fluorescent detection systems, sequential scanning at different wavelengths can circumvent signal overlap issues, provided appropriate fluorophores are selected with minimal spectral overlap.
Validating LTC4S antibody specificity requires implementing multiple orthogonal approaches. The foundational validation experiment involves positive and negative control samples. For LTC4S research, A-549 cells serve as a validated positive control based on documented expression patterns . Researchers should also include genetically modified cell lines with LTC4S knockdown or knockout for definitive negative controls.
A comprehensive validation protocol should include:
Western blot analysis confirming a single band at the expected molecular weight (17kDa)
Peptide competition assays where pre-incubation with the immunizing peptide should abolish specific signals
Immunoprecipitation followed by mass spectrometry to confirm the identity of the pulled-down protein
Correlation of protein detection with known expression patterns across different cell types or tissues
Side-by-side comparison with multiple antibodies targeting different epitopes of LTC4S
For immunohistochemical applications, validation should include comparison with in situ hybridization data for LTC4S mRNA expression. Additionally, researchers should be aware that antibodies targeting amino acids 36-51 might exhibit different specificity profiles than those targeting amino acids 29-55, necessitating epitope-specific validation . Quantitative PCR correlation with protein detection levels provides another layer of validation, particularly when examining differential expression across experimental conditions.
Transitioning between different detection systems with HRP-conjugated LTC4S antibodies requires systematic protocol adjustments to maintain optimal performance. When shifting from Western blot to ELISA applications, researchers must recalibrate antibody concentrations, as optimal dilutions for Western blotting (1:500-1:2000) typically differ from those for ELISA systems.
For Western blot to immunohistochemistry transitions:
Fixation optimization becomes critical, with paraformaldehyde generally preserving epitopes recognized by antibodies targeting amino acids 36-51 or 29-55
Antigen retrieval methods must be validated, with citrate buffer (pH 6.0) often providing good results for membrane protein epitopes
Incubation times typically require extension (overnight at 4°C) compared to Western blot protocols
Peroxidase blocking steps become essential to eliminate endogenous peroxidase activity in tissue samples
When moving from qualitative to quantitative applications:
Standard curves using recombinant LTC4S protein must be established
Signal development timing requires strict standardization
Multiple technical replicates become necessary for statistical validity
Calibration controls should be included in each experimental run
For fluorescence-based applications, substrate selection shifts from HRP chromogenic or chemiluminescent substrates to fluorescent tyramide signal amplification systems, requiring optimization of amplification timing to prevent signal saturation while maintaining sensitivity.
Epitope masking represents a significant challenge in LTC4S detection that can lead to false negative results or reduced signal intensity. As a membrane-associated protein involved in leukotriene biosynthesis, LTC4S exists in complex with other proteins and within membrane structures that can sterically hinder antibody access to specific epitopes .
Several biological factors contribute to epitope masking:
Protein-protein interactions: LTC4S functions within the MAPEG family and interacts with other proteins in the leukotriene biosynthesis pathway, potentially obscuring epitopes in native conditions
Post-translational modifications: These can alter epitope accessibility or recognition, particularly for antibodies targeting regions susceptible to phosphorylation or glycosylation
Membrane integration: The association of LTC4S with the nuclear envelope and endoplasmic reticulum can limit accessibility of certain epitopes in incompletely solubilized samples
Conformational states: Different functional states of LTC4S may expose or conceal specific epitopes
To address epitope masking challenges, researchers should implement modified sample preparation techniques including more rigorous detergent-based extraction (using combinations of non-ionic and ionic detergents), heat-induced epitope retrieval for fixed samples, and enzymatic treatments to remove interfering molecular structures. Additionally, comparing antibodies targeting different epitopes (e.g., N-terminal AA 29-55 versus AA 36-51) can help identify regions more susceptible to masking in specific experimental contexts .
Designing quantitative assays for LTC4S detection in clinical research environments demands stringent methodological considerations to ensure reproducibility and clinical relevance. The foundation of reliable quantification begins with proper assay validation, including determination of the lower limit of detection, dynamic range, precision (intra- and inter-assay variability), and accuracy through spike-recovery experiments.
For sandwich ELISA approaches, researchers should note that human LTC4S ELISA kits employ a specific methodology where the target protein is captured by pre-coated antibodies, followed by detection with biotin-conjugated antibodies and visualization via streptavidin-HRP systems . Sample collection and handling standardization is crucial, with consideration for:
Consistent collection timing relative to clinical parameters (e.g., symptom presentation, medication administration)
Standardized processing intervals to prevent degradation or artifactual changes in LTC4S levels
Appropriate anticoagulants for plasma samples that don't interfere with antibody-antigen interactions
Storage conditions validation to ensure stability during clinical sample banking
Reference range establishment requires large cohorts of healthy individuals stratified by relevant demographic factors. In multiplex clinical assays, potential cross-reactivity with other leukotrienes or structurally similar molecules must be thoroughly evaluated . For translational applications, correlation of LTC4S levels with established clinical markers or outcomes provides essential validation of the assay's clinical utility.
Optimal dilution ranges for LTC4S antibodies vary significantly by application type, target abundance, and detection system. Based on validated research protocols, the following dilution ranges provide starting points for experimental optimization:
It's important to note that these ranges should be considered starting points for titration experiments. Researchers should systematically test multiple dilutions within and beyond these ranges to identify optimal conditions for their specific samples and detection systems. For quantitative applications, standard curves using recombinant LTC4S at known concentrations should be prepared with each dilution to determine which provides the optimal balance between sensitivity and signal linearity. Additionally, when working with samples from species other than human, validation experiments comparing dilution efficiency across species are recommended due to potential differences in antibody affinity .
When confronting weak or absent signals in LTC4S detection, researchers should implement a systematic troubleshooting approach addressing all potential failure points in the experimental workflow. Begin by verifying target presence through parallel detection methods—if possible, use RT-qPCR to confirm LTC4S mRNA expression in your samples, particularly when working with cell lines or tissues not previously validated for LTC4S expression.
For Western blot applications showing weak signals:
Increase protein loading (up to 50-100μg total protein may be necessary for low-abundance targets)
Reduce antibody dilution incrementally (from 1:2000 to 1:1000 to 1:500)
Extend primary antibody incubation (overnight at 4°C instead of 1-2 hours at room temperature)
Switch to high-sensitivity detection substrates designed for low-abundance proteins
Optimize transfer conditions for low molecular weight proteins (17kDa for LTC4S)
For ELISA applications with suboptimal results:
Evaluate sample preparation methods—inappropriate storage or freeze-thaw cycles can degrade target proteins
Test different blocking reagents to improve signal-to-noise ratio
Extend incubation times for sample binding and detection antibody steps
Ensure all reagents are at room temperature before use as recommended in protocols
Consider sample concentration techniques for dilute samples
If signal remains problematic after these adjustments, epitope accessibility issues may be present. Consider different sample preparation methods, alternative antibodies targeting different epitopes of LTC4S, or enzymatic treatments to expose masked epitopes.
Valid interpretation of LTC4S antibody experimental results depends on implementing a comprehensive panel of controls addressing specificity, technical variation, and biological context. The following controls are considered essential:
Antibody Specificity Controls:
Positive control samples: Validated cell lines known to express LTC4S, such as A-549 cells
Negative control samples: Cell lines with confirmed low/no expression or CRISPR-mediated LTC4S knockout
Isotype controls: Irrelevant antibodies of the same isotype (IgG) and host species (rabbit) to assess non-specific binding
Peptide competition: Pre-incubation of antibody with immunizing peptide should abolish specific signal
Technical Controls:
Loading controls: For Western blot, housekeeping proteins (β-actin, GAPDH) for normalization
Standard curves: Recombinant LTC4S protein standards for quantitative applications
Dilution linearity: Serial dilutions of positive samples to confirm signal proportionality
Replicate samples: Technical replicates to assess method precision
Biological Context Controls:
Treatment controls: Samples with confirmed upregulation or downregulation of LTC4S (e.g., inflammatory stimulation)
Cross-species validation: When using antibodies across species, include human samples as reference points
Time-course samples: For studies examining temporal regulation of LTC4S
For sandwich ELISA applications specifically, additional controls should include blank wells (no sample), substrate-only wells (to assess HRP substrate stability), and dilution buffer controls to establish background signal baselines . Documentation of lot numbers and systematic recording of all experimental conditions ensures reproducibility and facilitates troubleshooting if inconsistencies arise.
Researchers should anticipate several key differences when comparing results obtained using different LTC4S antibody clones, particularly between those targeting distinct epitopes such as AA 36-51 versus AA 29-55 . These differences stem from fundamental variations in epitope recognition, accessibility, and antibody characteristics.
Signal Intensity Variations:
Different antibody clones typically exhibit varying affinities for their target epitopes, resulting in different signal intensities even when detecting identical amounts of LTC4S protein. Antibodies targeting the AA 36-51 region may demonstrate different sensitivity profiles compared to those targeting AA 29-55, necessitating clone-specific optimization of concentrations and incubation conditions .
Detection in Different Sample Types:
Epitope accessibility varies across sample preparation methods. For instance, certain fixation protocols may better preserve epitopes in the N-terminal region (AA 29-55) while potentially masking others. This results in clone-dependent performance across different applications (Western blot vs. immunohistochemistry) .
Cross-Reactivity Profiles:
Each antibody clone demonstrates a unique cross-reactivity pattern across species. Some LTC4S antibodies show broad reactivity across multiple species including human, cow, dog, and others, while some are more restricted to human samples . This differential cross-reactivity stems from evolutionary conservation patterns of specific epitope regions.
Post-Translational Modification Sensitivity:
Antibodies targeting different regions may exhibit variable sensitivity to post-translational modifications. Epitopes containing or adjacent to modification sites may show reduced binding when those modifications are present, creating discrepancies between results obtained with different clones.
When transitioning between antibody clones, researchers should perform side-by-side comparisons using identical samples and conditions to establish correlation factors, allowing for more accurate interpretation of historical data collected with different antibodies.
Emerging technologies are significantly expanding the capabilities and applications of LTC4S antibodies in research settings. Single-cell proteomics techniques are increasingly integrating antibody-based detection methods to analyze LTC4S expression at the individual cell level, revealing heterogeneity within seemingly homogeneous cell populations. This approach is particularly valuable for understanding the differential involvement of LTC4S in inflammatory responses across various immune cell subsets.
Multiplexed imaging technologies, including cyclic immunofluorescence and mass cytometry imaging, now allow for simultaneous detection of LTC4S alongside dozens of other proteins within intact tissue architecture. This contextual analysis provides unprecedented insights into the spatial relationships between LTC4S-expressing cells and their microenvironment in inflammatory diseases . These techniques benefit from the specificity of antibodies targeting defined epitopes such as AA 36-51 or AA 29-55 .
Proximity ligation assays utilizing LTC4S antibodies enable in situ visualization of protein-protein interactions involving LTC4S, offering functional insights beyond mere expression analysis. This application is particularly relevant given LTC4S's role in the MAPEG family and its interactions within the leukotriene biosynthesis pathway . Additionally, advances in microfluidic-based detection systems are improving sensitivity and throughput of LTC4S quantification in limited clinical samples, potentially enabling point-of-care diagnostics for inflammatory conditions where LTC4S plays a critical role .
Current LTC4S antibody technologies face several limitations that constrain their research applications. Epitope accessibility represents a persistent challenge, particularly for membrane-associated proteins like LTC4S that reside in the nuclear envelope and endoplasmic reticulum . Many antibodies struggle to access conformational epitopes in native protein states, limiting applications requiring intact protein structures. Potential solutions include developing antibodies against more accessible regions or implementing modified sample preparation techniques that better expose target epitopes while preserving native conformations.
Cross-reactivity issues present another significant limitation. While some LTC4S antibodies demonstrate reactivity across multiple species , true cross-species quantitative comparisons remain challenging due to varying affinities for orthologous epitopes. Development of antibodies targeting perfectly conserved epitopes or species-specific antibody panels would address this limitation.
The dynamic range limitations of current detection systems also constrain LTC4S quantification, particularly in samples with extremely high or low expression levels. Signal amplification technologies like tyramide signal amplification or quantum dot-based detection offer potential solutions for low-abundance scenarios, while calibrated detection systems could extend the upper range of quantification.
Batch-to-batch variability in polyclonal antibody production introduces reliability concerns for longitudinal studies. Transitioning to recombinant antibody technologies with defined sequences would ensure consistent performance across production batches. Finally, current antibodies typically recognize total LTC4S protein without distinguishing between active and inactive forms. Development of conformation-specific antibodies that selectively detect the catalytically active state would significantly advance functional studies of LTC4S in inflammatory pathways.
Integration of LTC4S antibody-based assays into multi-omics research frameworks offers powerful opportunities for comprehensive understanding of inflammatory pathways and disease mechanisms. Strategic implementation involves several key approaches:
For proteogenomic integration, researchers can correlate LTC4S protein levels detected via antibody-based methods with corresponding mRNA expression data from RNA-seq or microarray analyses. This correlation identifies potential post-transcriptional regulation mechanisms affecting LTC4S expression in different physiological or pathological states. Discrepancies between protein and mRNA levels provide insights into regulatory mechanisms specific to LTC4S.
In metabolomic integration approaches, quantification of LTC4S protein using specific antibodies targeting AA 36-51 or AA 29-55 can be directly correlated with measurements of leukotriene C4 and related eicosanoids in the same samples. This protein-metabolite relationship analysis reveals functional consequences of LTC4S expression variations and potential rate-limiting steps in the leukotriene biosynthesis pathway.
Sequential antibody-based cell isolation followed by multi-omics analysis enables deep characterization of LTC4S-expressing cell populations. This workflow involves initial isolation of LTC4S-positive cells using antibody-based methods, followed by comprehensive analysis of these isolated populations through RNA-seq, ATAC-seq, and metabolomics, revealing the broader functional context of LTC4S-expressing cells.