EPX Antibody

Shipped with Ice Packs
In Stock

Description

EPX Antibody Characteristics

EPX antibodies are designed to detect eosinophil peroxidase, a granule protein exclusive to eosinophils. Key features include:

Diagnostic and Research Applications

EPX antibodies are pivotal in both clinical diagnostics and mechanistic studies:

Key Applications

  • Immunohistochemistry (IHC): Detects eosinophil infiltration in tissues (e.g., EoE biopsies) .

  • Flow Cytometry: Identifies eosinophils in peripheral blood with high specificity (0.5–1 µg/10^6 cells) .

  • Western Blot (WB): Validates EPX expression in cell lysates (e.g., HEK293T, PC12) at ~81 kDa .

  • ELISA: Quantifies anti-EPX autoantibodies in serum, linked to refractory ECRS severity .

Clinical Relevance

  • Eosinophilic Mucin Decomposition: Neutralizing anti-EPX antibodies accelerates mucin breakdown and restores corticosteroid sensitivity in ECRS .

  • Biomarker Utility:

    • Sputum EPX: More sensitive than eosinophil counts for detecting airway inflammation in asthma (AUC = 0.85) .

    • Serum EPX: Correlates with sinus CT scores (r = 0.35, P = 0.009) and fractional exhaled nitric oxide (FeNO) in ECRS .

EPX in Disease Pathogenesis

Study FocusKey OutcomeSource
ECRS and AsthmaAnti-EPX IgG levels elevated in refractory cases; dupilumab reduces levels .
EoE DiagnosisCytosponge EPX staining correlates with eosinophil density (r = 0.82) .
Mepolizumab TherapyNormalizes serum EPX in 96% of asthma patients but sputum EPX in only 49% .

Mechanistic Insights

  • EPX antibodies enhance dsDNA release from eosinophils, perpetuating inflammation in eosinophilic mucin .

  • EPX-mediated tyrosine nitration inactivates PP2A, reducing glucocorticoid receptor nuclear translocation .

Future Directions

  • Therapeutic Targeting: Neutralizing EPX antibodies may offer novel treatments for steroid-resistant eosinophilic inflammation .

  • Biomarker Optimization: Standardizing sputum EPX thresholds could improve EoE and asthma monitoring .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Description

This polyclonal rabbit anti-EPX antibody is raised against a recombinant protein encompassing amino acids 140-300 of the human Eosinophil peroxidase protein. It is specifically designed to detect the human EPX protein. The antibody has undergone protein G purification, achieving a purity level of up to 95%. Its specificity has been validated through ELISA and IHC assays.

Eosinophil peroxidase (EPX) is a crucial enzyme primarily responsible for catalyzing the oxidation of halide ions, resulting in the generation of reactive oxygen species (ROS). These ROS play a vital role in the elimination of parasites and other pathogens. Beyond its role in host defense, EPX has been implicated in the pathogenesis of various diseases, including asthma, allergic rhinitis, and inflammatory bowel disease.

Form
Liquid
Lead Time
Generally, we are capable of dispatching the products within 1-3 working days after receiving your orders. Delivery timelines may vary depending on the chosen purchasing method or location. For specific delivery estimates, kindly consult your local distributors.
Synonyms
Eosinophil peroxidase [Precursor] antibody; Eosinophil peroxidase antibody; Eosinophil peroxidase heavy chain antibody; EPER antibody; EPO antibody; EPP antibody; EPX antibody; EPX PEN antibody; PERE antibody; PERE_HUMAN antibody
Target Names
EPX
Uniprot No.

Target Background

Function
EPX plays a significant role in mediating tyrosine nitration of secondary granule proteins within mature resting eosinophils. Notably, it exhibits significant inhibitory activity against Mycobacterium tuberculosis H37Rv, achieving bacterial fragmentation and lysis.
Gene References Into Functions
  1. Peroxidase enzymes, including MPO and EPO, are believed to have a critical role in inhibiting RANKL-induced osteoclast differentiation at inflammatory sites associated with bone fractures and injuries. PMID: 27836774
  2. Both MPO and EPO have been causatively linked to breast cancer progression and are identified as potential therapeutic targets. Specific novel inhibitors targeting these enzymes could potentially reduce tumor growth and limit the occurrence of metastasis. PMID: 28260049
  3. A significant finding is the identification of EPO as a novel ligand for the HER2 receptor. Upon activation of HER2, EPO induces activation of FAK and subsequent activation of beta1-integrin via inside-out signaling. This complex cascade leads to downstream activation of ERK1/2 and sustained upregulation of both MUC4 and the HER2 receptor. PMID: 27519953
  4. EPO-mediated protein carbamylation is enhanced during allergen-induced asthma exacerbation. This process can modulate immune responses and trigger a cascade of inflammatory signals prevalent in asthma. PMID: 27587397
  5. A strong correlation exists between nasal and pharyngeal EPX levels and the eosinophil percentage of induced sputum in a given patient. PMID: 26645423
  6. Myeloperoxidase and eosinophil peroxidase are readily internalized by HUVEC cells where they promote cellular proliferation, migration, invasion, and stimulate angiogenesis both in vitro and in vivo. PMID: 26386352
  7. EPO exhibits a preferential role in signaling via a specific surface receptor, leading to neural plasticity. PMID: 24937179
  8. mRNA levels of eosinophil granule proteins, rather than sputum eosinophil%, may reflect airway hyperresponsiveness and airflow limitation. PMID: 24814827
  9. Eosinophil peroxidase in sputum serves as a unique biomarker of airway eosinophilia. PMID: 23931643
  10. This study investigates the prevalence of a common SNP in the eosinophil protein x/eosinophil-derived neurotoxin (EPX/EDN, RNase2) and its association with cellular contents of EPX/EDN and ECP. PMID: 24738159
  11. Polymorphisms of EPX and ECP have been linked to inflammatory bowel disease in an age and gender-dependent manner. PMID: 23197886
  12. Data suggests that eosinophil peroxidase (EPX)-based ELISA is the only eosinophil-specific assay. PMID: 22750539
  13. A mechanism for inducing ASIC-3 expression relevant to allergic rhinitis is proposed. This mechanism suggests that eosinophil peroxidase (EPO), acting via ERK1/2, induces the expression of ASIC-3 in epithelial cells. PMID: 22702502
  14. HER2 has been identified as a novel mediator of eosinophil peroxidase signaling. Eosinophil peroxidase, at noncytotoxic levels, can drive cell-cycle progression and proliferation. PMID: 21454806
  15. EPO-dependent oxidative damage may contribute to tissue injury in bisulfite-exacerbated eosinophilic inflammatory disorders. PMID: 20501663
  16. EPX induces surface alteration, killing, and lysis of Mycobacterium tuberculosis. PMID: 12540536
  17. Findings suggest that Pro358Leu in EPO is strongly involved in the development of cedar pollinosis. PMID: 14657871
  18. Polymorphisms of the eosinophil peroxidase gene may be associated with Japanese cedar pollinosis. PMID: 15316147
  19. This study analyzes the role of thiocyanate (SCN-) in modulating the catalytic activity of myeloperoxidase (MPO) and other members of the lactoperoxidase (LPO) and eosinophil peroxidase (EPO) families. PMID: 15894800
  20. The mechanisms by which eosinophil peroxidase oxidizes nitrite are reported. PMID: 16336215
  21. Post-translational tyrosine nitration of eosinophil granule toxins is mediated by eosinophil peroxidase. PMID: 18694936
  22. No variation in genes encoding eosinophil peroxidase has been observed in this German cohort in relation to Atopic dermatitis pathogenesis. PMID: 19014520
  23. Genetic variability in the EPO gene may contribute to the susceptibility to allergic rhinitis (or related phenotypes) in the Czech population. PMID: 19439985

Show More

Hide All

Database Links

HGNC: 3423

OMIM: 131399

KEGG: hsa:8288

STRING: 9606.ENSP00000225371

UniGene: Hs.279259

Involvement In Disease
Eosinophil peroxidase deficiency (EPXD)
Protein Families
Peroxidase family, XPO subfamily
Subcellular Location
Cytoplasmic granule. Note=Cytoplasmic granules of eosinophils.

Q&A

What is EPX and why is it important in research?

Eosinophil peroxidase (EPX) is an 81 kilodalton protein found in the secondary granules of eosinophils . It serves as a specific marker of eosinophil activation in both clinical patients and experimental mouse models of human disease . The importance of EPX in research stems from its role as a reliable biomarker for eosinophilic disorders, including eosinophilic airway inflammation, bronchial asthma, and eosinophilic chronic rhinosinusitis (ECRS) . Detection of extracellular EPX in biological samples provides researchers with a quantitative measure of eosinophil degranulation, which is crucial for understanding the pathophysiology of various inflammatory and allergic conditions. The development of specific antibodies against EPX has significantly enhanced our ability to detect and quantify eosinophil involvement in disease processes, making it an invaluable tool in immunological and pathological research.

What applications are EPX antibodies commonly used for?

EPX antibodies are utilized across a diverse range of experimental applications in research settings. Most commonly, these antibodies are employed in Western Blot (WB) analysis to detect and quantify EPX protein in tissue or cellular extracts . Enzyme-Linked Immunosorbent Assay (ELISA) represents another major application, allowing for highly sensitive quantification of EPX in biological fluids and tissue samples . Additionally, EPX antibodies are extensively used in immunohistochemistry (IHC) to visualize the presence and distribution of eosinophils in tissue sections, flow cytometry (FCM) for cellular analysis, and immunofluorescence (IF) for subcellular localization studies . The broad applicability of these antibodies makes them versatile tools for researchers investigating eosinophil biology across various experimental systems and disease models. Some specialized applications include investigating eosinophil degranulation in ex vivo cell cultures and examining the relationship between EPX and the formation of extracellular DNA traps in inflammatory conditions .

How should sample preparation be optimized for EPX antibody detection?

Optimal sample preparation for EPX antibody detection varies based on the specific application and tissue origin. For biological fluids such as bronchoalveolar lavage fluid or serum, samples should be promptly centrifuged at 1,300× g for 5 minutes, followed by a second centrifugation at 13,000× g for 5 minutes to generate cell-free and organelle-free supernatants . These supernatants should be stored at -80°C until analysis to preserve EPX integrity. For tissue extracts, gentle tissue homogenization in appropriate buffering solutions containing protease inhibitors is crucial to prevent degradation of the target protein. When preparing samples for Western blot analysis, non-reducing conditions may better preserve the epitopes recognized by some EPX antibodies, though this varies depending on the specific antibody clone used. For immunohistochemistry applications, proper fixation is critical—paraformaldehyde fixation followed by careful paraffin embedding typically yields optimal results for EPX detection in tissue sections. It is also important to note that eosinophil activation can be induced during sample processing, potentially leading to artificial degranulation; therefore, rapid processing at controlled temperatures and minimizing mechanical disruption are essential for obtaining accurate results.

How can EPX antibodies be used to develop sensitive detection assays?

Development of sensitive EPX detection assays requires careful optimization of multiple parameters to achieve maximum sensitivity and specificity. A full-factorial approach to ELISA development has proven most effective, as demonstrated in studies that achieved a 10-fold increase in sensitivity compared to traditional OPD-based peroxidase activity assays . Key optimization steps include careful selection of antibody pairs with complementary binding properties—typically utilizing one antibody as a capture reagent and a second, differently epitope-targeting antibody as a detection reagent. For instance, the clone MM25-429.1.1 has been successfully employed as a capture antibody in sandwich ELISA formats, while biotinylated MM25-82.2.1 serves effectively as a detection antibody . Biotinylation efficiency significantly impacts assay performance, with optimal results occurring when 8-12 molecules of biotin are conjugated per antibody molecule . Additional optimization factors include coating buffer composition, blocking agent selection, sample dilution parameters, and incubation conditions. The incorporation of streptavidin-conjugated enzyme reporters can further enhance sensitivity. For researchers developing such assays, preliminary cross-titration experiments with varying concentrations of capture and detection antibodies are essential to establish optimal working conditions and dynamic range parameters specific to their research applications.

What are the challenges in measuring EPX in complex biological samples?

Measuring EPX in complex biological matrices presents several technical challenges that researchers must address to obtain reliable results. First, eosinophil autoactivation during sample collection and processing can lead to artifactual EPX release, necessitating rapid sample handling and appropriate stabilizing buffers. Second, EPX can bind to extracellular matrix components and cellular debris, potentially masking epitopes and reducing detection efficiency—pre-treatment with appropriate dissociation buffers may be required to maximize recovery. Third, the presence of endogenous peroxidases in biological samples can interfere with activity-based EPX assays, requiring either selective inhibition of these interferents or the use of immunological detection methods. Fourth, in diseased tissues, the formation of EPX-containing immune complexes with anti-EPX autoantibodies can complicate accurate quantification, as seen in eosinophilic chronic rhinosinusitis where such complexes contribute to eosinophilic mucin formation . Finally, EPX stability varies considerably across different sample types, with degradation occurring more rapidly in protease-rich environments like inflammatory exudates. To overcome these challenges, researchers should consider multiple analytical approaches, implement appropriate control samples, and validate measurements using complementary techniques such as combining immunological detection with enzymatic activity assays when feasible.

How do EPX antibodies contribute to understanding eosinophil-mediated pathology?

EPX antibodies have become instrumental in elucidating the mechanisms of eosinophil-mediated pathology across various disease states. By enabling precise tracking of eosinophil activation and degranulation in tissues, these antibodies have revealed that eosinophil peroxidase plays multifaceted roles beyond its enzymatic activity. In eosinophilic airway inflammation, immunofluorescence studies using anti-EPX antibodies have demonstrated that EPX contributes to tissue damage through direct cytotoxicity to epithelial cells and promotes hyperreactivity through nerve cell interactions . More recent investigations have uncovered that EPX participates in the formation of extracellular DNA traps, with anti-EPX antibodies helping to visualize these structures in inflammatory tissues . Particularly significant is the discovery that anti-EPX autoantibodies can form in patients with chronic eosinophilic conditions, creating immune complexes that resist clearance and perpetuate inflammation . Research utilizing EPX antibodies has further established correlations between tissue EPX deposition patterns and disease severity metrics, including CT scores in eosinophilic rhinosinusitis and exhaled nitric oxide levels in asthma . These findings have directly influenced therapeutic approaches, with studies showing that treatments like dupilumab can decrease anti-EPX antibody levels, correlating with clinical improvement . The methodological advance of EPX antibody-based assays has thus transformed our understanding of eosinophil biology from simple effector cells to orchestrators of complex inflammatory cascades.

What is the relationship between anti-EPX autoantibodies and disease severity?

Recent research has revealed significant correlations between anti-EPX autoantibodies and disease severity in eosinophilic disorders. Studies focused on eosinophilic chronic rhinosinusitis (ECRS) have demonstrated that serum levels of anti-EPX antibodies positively correlate with sinus computed tomography scores, providing a quantitative relationship between autoantibody presence and objective disease parameters . Additionally, these autoantibody levels show a strong positive correlation with fractionated exhaled nitric oxide (FeNO) measurements, suggesting their potential relevance as biomarkers for both upper and lower airway eosinophilic inflammation . Patients classified as having refractory ECRS consistently display significantly higher serum levels of anti-EPX antibodies compared to non-refractory cases, indicating a potential mechanistic role in treatment resistance . At the molecular level, immunoglobulins isolated from mucin supernatants of ECRS patients enhance double-stranded DNA (dsDNA) release from eosinophils, potentially contributing to inflammatory exacerbation . Conversely, neutralization of anti-EPX antibodies has been shown to inhibit dsDNA release and accelerate mucin decomposition in experimental models, suggesting potential therapeutic implications . These findings collectively point to anti-EPX autoantibodies functioning not merely as disease markers but as active contributors to pathological processes, particularly in the context of treatment-resistant eosinophilic disorders. The relationship appears bidirectional, with successful therapeutic interventions such as dupilumab treatment resulting in decreased serum levels of these autoantibodies .

What factors influence the optimization of EPX-based sandwich ELISA?

Optimizing EPX-based sandwich ELISA requires meticulous attention to multiple technical parameters to achieve maximum sensitivity and specificity. The selection of complementary antibody pairs is paramount—ideal pairs should recognize distinct, non-overlapping epitopes on the EPX molecule to prevent competitive binding . The capture antibody concentration significantly impacts assay performance, with 2μg/ml of anti-EPX monoclonal antibody (such as clone MM25-429.1.1) in appropriate coating solution typically yielding optimal results when incubated at 4°C overnight . Blocking conditions must be carefully optimized to minimize background while maintaining signal intensity, with 30-minute room temperature incubation in blocking solution generally providing adequate non-specific binding prevention . Sample incubation parameters affect both sensitivity and throughput, with 1.5-hour room temperature incubation without shaking often representing an optimal balance . The detection system must be calibrated specifically for EPX detection, with biotinylated detection antibodies (such as clone MM25-82.2.1) combined with streptavidin-conjugated enzymes proving particularly effective . Assay validation should include assessment of linearity, recovery, precision, and spike controls to ensure reliable quantification across the dynamic range. Temperature stability during all assay steps is essential for reproducibility, and batch-to-batch consistency in antibody preparations (particularly biotinylation efficiency) must be monitored to maintain consistent assay performance over time. Implementation of these optimization strategies can yield EPX detection assays with significantly enhanced sensitivity compared to traditional peroxidase activity measurements.

How should researchers approach EPX antibody validation for specific applications?

Rigorous validation of EPX antibodies for specific research applications requires a systematic, multi-parameter approach to ensure reliable and reproducible results. Begin with specificity assessment using positive and negative control samples—eosinophil-rich tissues from wild-type animals versus EPX knockout models provide ideal validation materials . Western blot analysis should confirm single-band recognition at the expected molecular weight of approximately 81 kDa . For immunohistochemical applications, co-localization studies with established eosinophil markers (such as major basic protein) can verify specific staining patterns. When developing new assay formats, cross-reactivity testing against related peroxidases (myeloperoxidase, lactoperoxidase) is essential to confirm specificity within the peroxidase family. Sensitivity assessment should include limit of detection and quantification determinations using serial dilutions of purified EPX protein. For functional applications, researchers should verify that antibody binding does not significantly alter EPX enzymatic activity unless such inhibition is the experimental goal. Application-specific validation is crucial—antibodies that perform well in ELISA may not necessarily work in immunohistochemistry or flow cytometry. When validating for clinical sample analysis, assessment across diverse pathological specimens is recommended to account for matrix effects and potential interfering substances. Finally, epitope mapping or similar approaches can provide valuable information about the binding characteristics of specific antibody clones, informing optimal pairing strategies for sandwich assays and improving interpretation of experimental results.

How can EPX antibodies be used to study eosinophil degranulation in vitro?

Studying eosinophil degranulation in vitro using EPX antibodies enables detailed investigation of activation mechanisms and potential therapeutic interventions. A standardized protocol involves isolating eosinophils (typically from peripheral blood or from IL-5 transgenic mouse models ) and suspending cells at concentrations of 1×10^6 cells/ml in appropriate buffer, before stimulation with degranulation-inducing agents such as PAF-C18 (50ng/ml) or PAF-C18 plus ionomycin (1μM) . Following incubation periods (typically 6 hours at 37°C in 5% CO2), samples should be centrifuged at 1,300×g for 5 minutes, with supernatants subsequently centrifuged at 13,000×g to generate cell-free fractions suitable for EPX detection . EPX antibodies can then be employed in sandwich ELISA format to quantify released EPX as a direct measure of degranulation. For more detailed mechanistic studies, immunofluorescence approaches using anti-EPX antibodies can visualize the dynamic process of granule mobilization and release, particularly when combined with confocal microscopy techniques. Flow cytometric analyses with permeabilized versus non-permeabilized conditions can distinguish between surface-bound and intracellular EPX pools. Live-cell imaging techniques incorporating fluorescently labeled anti-EPX antibody fragments can capture real-time degranulation events. When evaluating potential inhibitors of eosinophil activation, dose-response relationships can be established by measuring EPX release across concentration gradients. Finally, comparing EPX release with other granule proteins (major basic protein, eosinophil cationic protein) provides a more comprehensive assessment of differential granule release patterns in response to various stimuli, offering insights into the heterogeneity of degranulation responses under different pathophysiological conditions.

How are EPX antibodies being used in therapeutic development research?

EPX antibodies are increasingly playing crucial roles in therapeutic development research, functioning as both research tools and potential therapeutic agents themselves. As research tools, these antibodies enable high-throughput screening assays to identify compounds that inhibit EPX release or enzymatic activity, providing platforms for discovering novel anti-eosinophilic drugs. Recent studies have demonstrated that neutralizing anti-EPX antibodies can directly inhibit double-stranded DNA release from eosinophils and accelerate the decomposition of pathological mucin formations, suggesting potential therapeutic applications in eosinophilic chronic rhinosinusitis and other eosinophilic disorders . In clinical development pipelines, monitoring EPX levels in biological samples using specific antibodies serves as a biomarker for assessing the efficacy of biological therapeutics targeting eosinophil-mediated diseases. For instance, dupilumab treatment has been shown to decrease serum levels of anti-EPX antibodies, correlating with clinical improvement in patients with refractory eosinophilic conditions . The development of humanized monoclonal antibodies against EPX itself represents an emerging therapeutic strategy, potentially neutralizing the harmful effects of extracellular EPX in various inflammatory conditions. Beyond direct targeting, EPX antibodies are facilitating research into the relationship between EPX and other disease mediators, revealing potential combination therapy approaches. As companion diagnostics, EPX antibody-based assays may help identify patient subpopulations most likely to respond to specific anti-eosinophilic therapies, advancing precision medicine approaches to treating these complex immunological disorders.

What are the emerging roles of EPX in non-classical eosinophil functions?

Recent research utilizing specialized EPX antibodies has unveiled several non-classical roles of eosinophil peroxidase beyond its traditional cytotoxic functions. EPX has been implicated in the formation of extracellular DNA traps, structures that contribute to microbial containment but also potentially exacerbate tissue damage in chronic inflammatory conditions . Immunofluorescence studies with anti-EPX antibodies have revealed that EPX integrates into these DNA networks, enhancing their stability and antimicrobial properties. Additionally, research has uncovered that EPX can function as an autoantigenic target, with anti-EPX autoantibodies detected in patients with eosinophilic disorders . These autoantibodies appear to form immune complexes with EPX, potentially prolonging inflammatory responses and contributing to treatment resistance in conditions like eosinophilic chronic rhinosinusitis . EPX has also been shown to modulate the extracellular matrix composition through interactions with proteoglycans and structural proteins, potentially influencing tissue remodeling processes in chronic inflammatory conditions. There is emerging evidence that EPX may have immunomodulatory effects on various cell types, including dendritic cells and T lymphocytes, suggesting a role in shaping adaptive immune responses. Furthermore, recent studies indicate potential involvement of EPX in thrombotic processes, linking eosinophil activation with cardiovascular complications in hypereosinophilic syndromes and related disorders. The development of highly specific EPX antibodies has been instrumental in revealing these non-canonical functions, transforming our understanding of eosinophil biology from simple effector cells to multifunctional orchestrators of complex immunological processes.

How do different antibody clones compare in detecting various forms of EPX?

Different anti-EPX antibody clones exhibit varying capacities to detect distinct forms of EPX across experimental platforms, significantly impacting research outcomes. Monoclonal antibodies developed against specific epitopes may preferentially recognize native versus denatured forms of EPX, making some clones more suitable for applications like immunohistochemistry (where protein conformation is preserved) versus Western blotting (where proteins are typically denatured). For instance, clone MM25-429.1.1 has demonstrated superior performance as a capture antibody in sandwich ELISA formats, while clone MM25-82.2.1 functions optimally as a detection antibody when appropriately biotinylated . Some antibody clones preferentially bind to EPX in its enzymatically active conformation, while others can detect both active and inactive forms, a distinction particularly relevant when investigating the functional consequences of EPX release in inflammatory environments. In clinical samples, EPX may exist in complex with other proteins or partially degraded by proteases, potentially masking epitopes recognized by certain antibody clones. Antibodies targeting different regions of the EPX molecule (N-terminal, C-terminal, or internal domains) demonstrate varying abilities to detect EPX in the context of complex formation with anti-EPX autoantibodies, a phenomenon particularly relevant in eosinophilic chronic rhinosinusitis research . The glycosylation status of EPX can also influence antibody binding, with some clones demonstrating sensitivity to these post-translational modifications. For comprehensive EPX characterization, researchers should consider employing multiple antibody clones targeting different epitopes, particularly when investigating novel tissue types or disease conditions where EPX conformation or modification status may be altered.

What technological advances are improving EPX antibody-based detection systems?

Technological innovations are substantially enhancing the performance and utility of EPX antibody-based detection systems across multiple research domains. Digital ELISA platforms, employing single-molecule detection principles, have dramatically improved sensitivity limits for EPX quantification, potentially enabling detection of eosinophil activation at much earlier disease stages than previously possible. Multiplex immunoassay formats now allow simultaneous measurement of EPX alongside other inflammatory mediators in small sample volumes, providing more comprehensive inflammatory profiles while conserving precious research specimens. Advances in recombinant antibody engineering have yielded high-affinity anti-EPX antibody fragments with improved tissue penetration properties for imaging applications and enhanced stability for long-term storage. The integration of microfluidic systems with EPX antibody-based detection has enabled rapid, point-of-care assessment of eosinophil activation status, potentially translating research tools into clinical diagnostics. Automation of antibody-based assays has improved standardization across laboratories while reducing labor intensity and human error. Surface plasmon resonance and biolayer interferometry technologies now provide real-time, label-free analysis of EPX-antibody interactions, yielding valuable kinetic data about binding affinities under physiologically relevant conditions. Imaging mass cytometry incorporating anti-EPX antibodies allows highly multiplexed tissue analysis, revealing spatial relationships between eosinophils and other cellular components in inflammatory microenvironments. Finally, computational approaches to antibody design are accelerating the development of next-generation anti-EPX antibodies with enhanced specificity, reduced cross-reactivity, and optimized binding characteristics for specific research applications, promising continued improvements in our ability to detect and characterize this important biomarker of eosinophil activation.

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.