Protheragen offers specialized biomarker analysis services exclusively focused on supporting drug discovery and preclinical development for Eosinophilic Esophagitis (EoE). Our comprehensive biomarker panel is designed to advance the understanding of EoE pathophysiology, enabling the identification and characterization of molecular targets involved in disease mechanisms. Please note that all services are strictly for research and preclinical drug development purposes; we do not provide any clinical diagnostic services.
Effective therapeutic intervention for Eosinophilic Esophagitis begins with the precise discovery and identification of disease-relevant biomarkers. Protheragen’s biomarker discovery services leverage advanced screening and validation methodologies to identify molecular signatures associated with EoE. Our approach encompasses high-throughput screening of candidate molecules, followed by rigorous validation to ensure relevance to disease pathways. This process is integral to informing target selection, guiding drug development strategies, and elucidating mechanisms of action during preclinical research.
Multi Omics: Our multi-omics platform integrates genomics, transcriptomics, proteomics, and metabolomics to deliver a comprehensive analysis of biological systems implicated in Eosinophilic Esophagitis. By combining cutting-edge sequencing technologies, mass spectrometry, and advanced bioinformatics, we enable the identification of DNA, RNA, protein, and metabolite biomarkers. This approach facilitates an in-depth exploration of key disease pathways, such as Th2-mediated immune responses, eosinophil activation, cytokine signaling, and epithelial barrier dysfunction, which are central to EoE pathogenesis.
Candidate Validation: Protheragen employs robust validation strategies to assess candidate biomarkers for their association with Eosinophilic Esophagitis pathophysiology. Preliminary screening processes include quantitative and qualitative analyses across diverse sample types, ensuring reproducibility and relevance. Candidates are prioritized based on criteria such as differential expression profiles, biological function, involvement in EoE-relevant pathways, and potential for measurable impact in preclinical models. This systematic approach supports the selection of the most promising biomarkers for further assay development.
Diverse Technological Platforms: Our custom assay development capabilities span a wide range of technological platforms, allowing for adaptation to specific research requirements. We offer tailored solutions using immunoassays, mass spectrometry, flow cytometry, molecular diagnostics, and histopathology/imaging platforms. These platforms are optimized to accommodate diverse sample types and analytical needs, ensuring flexibility and precision in biomarker quantification and characterization.
Immunoassays: We develop and implement ELISA, chemiluminescent, and multiplex immunoassays for the quantitative detection of cytokines, chemokines, and other protein biomarkers associated with EoE.
Mass Spectrometry: Our LC-MS/MS platforms enable the sensitive and specific quantification of proteins, lipids, and metabolites relevant to EoE, supporting both targeted and untargeted analyses.
Flow Cytometry: We utilize flow cytometry for the phenotypic and functional characterization of immune cell populations, including eosinophils, Th2 lymphocytes, and other relevant subsets.
Molecular Diagnostics: We provide molecular assays for the detection and quantification of gene expression (mRNA, miRNA) and genetic variants associated with EoE pathogenesis.
Histopathology And Imaging: Our services include tissue-based analyses using immunohistochemistry, in situ hybridization, and advanced imaging techniques to localize and quantify biomarkers within esophageal tissue samples.
Rigorous Method Validation: All analytical methods undergo rigorous validation in accordance with established research guidelines. We assess performance characteristics including sensitivity, specificity, precision, linearity, and reproducibility. Comprehensive quality control measures are implemented throughout the process to ensure data integrity, reliability, and scientific robustness, supporting the translational potential of research findings.
Protheragen provides quantitative analysis capabilities for biomarker measurement across multiple platforms. Our protocols are designed to deliver accurate, reproducible, and sensitive quantification of biomarker levels in biological samples, supporting comparative studies and longitudinal analyses in preclinical research.
Sample Analysis: We handle a wide array of sample types, including blood, tissue, sputum, and cellular extracts relevant to Eosinophilic Esophagitis. Each sample undergoes standardized processing and analysis protocols, with stringent quality control at every step to ensure sample integrity and data reliability. Our workflows are tailored to maximize data yield while minimizing variability.
High Throughput Capabilities: Our high-throughput analytical platforms enable the simultaneous analysis of multiple biomarkers using multiplexed assays. This approach increases efficiency, conserves valuable samples, and accelerates data acquisition, facilitating large-scale studies and comprehensive biomarker profiling in EoE research.
| Gene Target | Biological Function | Application as a Biomarker |
|---|---|---|
| C-C motif chemokine receptor 3 (CCR3) | C-C motif chemokine receptor 3 (CCR3) is a G protein-coupled receptor predominantly expressed on eosinophils, basophils, Th2 lymphocytes, and certain mast cells. CCR3 binds several chemokines, including eotaxin (CCL11), eotaxin-2 (CCL24), and eotaxin-3 (CCL26), among others. Upon ligand binding, CCR3 mediates chemotaxis and activation of these immune cells, facilitating their migration to sites of inflammation. This receptor plays a significant role in the regulation of allergic responses, inflammatory processes, and host defense by directing the trafficking and function of effector leukocytes. | CCR3 expression has been utilized as a biomarker for identifying and characterizing eosinophils and Th2-type immune responses in various conditions. Elevated CCR3 levels have been observed in tissues affected by allergic diseases such as asthma, allergic rhinitis, and atopic dermatitis. Measurement of CCR3 expression can aid in the assessment of eosinophil-associated inflammation and has been used in research to monitor disease activity, evaluate the efficacy of anti-inflammatory therapies, and distinguish between different immune cell populations in tissue samples. |
| arachidonate 15-lipoxygenase (ALOX15) | Arachidonate 15-lipoxygenase (ALOX15) is an enzyme that catalyzes the oxygenation of polyunsaturated fatty acids, primarily arachidonic acid, to generate 15-hydroperoxyeicosatetraenoic acid (15-HPETE). This enzymatic activity results in the production of various bioactive lipid mediators, including 15-hydroxyeicosatetraenoic acid (15-HETE) and specialized pro-resolving mediators such as lipoxins. ALOX15 is involved in the regulation of inflammation, cell differentiation, and apoptosis. It is expressed in several tissues, including eosinophils, reticulocytes, and airway epithelial cells, and plays roles in processes such as erythropoiesis and the resolution of inflammatory responses. | ALOX15 expression and activity have been investigated as potential biomarkers in a variety of contexts, including inflammatory diseases, asthma, atherosclerosis, and certain cancers. Changes in ALOX15 levels or activity in tissues or biological fluids have been associated with disease presence, severity, or progression. For example, elevated or altered ALOX15 expression has been reported in airway tissues of individuals with asthma and in atherosclerotic lesions. Additionally, ALOX15 has been studied as a marker for oxidative stress and inflammatory status in various pathological conditions. |
| interleukin 10 (IL10) | Interleukin 10 (IL-10) is a cytokine produced by a variety of immune cells, including T cells, B cells, macrophages, and dendritic cells. Its primary biological function is to limit and terminate inflammatory responses by inhibiting the synthesis of pro-inflammatory cytokines such as IFN-γ, IL-2, IL-3, TNF-α, and GM-CSF produced by cells like macrophages and helper T cells. IL-10 also enhances B cell survival, proliferation, and antibody production. It plays a crucial role in maintaining immune homeostasis and preventing tissue damage during immune responses by suppressing the activation and effector functions of various immune cells. | IL-10 is frequently measured in biological fluids to assess immune status and inflammation. Elevated IL-10 levels have been observed in various infectious, autoimmune, and inflammatory conditions, and are often associated with disease activity or severity. In clinical and research settings, IL-10 is used as a biomarker to monitor immune regulation, evaluate response to therapy, and characterize the immune profile in diseases such as sepsis, autoimmune disorders, and certain cancers. |
| interleukin 13 (IL13) | Interleukin 13 (IL13) is a cytokine produced primarily by activated Th2-type CD4+ T lymphocytes, as well as by other immune cells including mast cells and basophils. IL13 plays a central role in the regulation of immune responses, particularly those involved in allergic inflammation and the defense against parasitic infections. It influences the differentiation and proliferation of B cells, enhances immunoglobulin E (IgE) production, and modulates the activity of macrophages. IL13 also contributes to the regulation of mucus production, airway hyperresponsiveness, and tissue remodeling processes, especially in the context of respiratory diseases. | IL13 has been studied as a biomarker in conditions characterized by Th2-mediated immune responses, such as asthma, allergic rhinitis, and certain atopic diseases. Elevated levels of IL13 in serum, sputum, or tissue samples have been associated with disease activity and severity in these disorders. Its measurement has been used in research to assess inflammatory status, monitor therapeutic responses, and investigate disease pathophysiology. |
| interleukin 15 (IL15) | Interleukin 15 (IL15) is a cytokine belonging to the four-alpha-helix bundle family, closely related to interleukin 2 (IL2) in structure and function. IL15 is primarily produced by mononuclear phagocytes, dendritic cells, and non-lymphoid tissues. It plays a critical role in the development, survival, and activation of natural killer (NK) cells and memory phenotype CD8+ T cells. IL15 signals through a receptor complex composed of IL15Rα, IL2/15Rβ (CD122), and the common γ chain (CD132), leading to the activation of JAK/STAT and other intracellular signaling pathways. IL15 is involved in immune homeostasis, promotes cytotoxic lymphocyte responses, and contributes to the regulation of inflammation and host defense against infections and tumors. | IL15 levels have been investigated as a biomarker in various clinical contexts, including immune-mediated diseases, infectious diseases, and cancer. Elevated IL15 concentrations in serum, plasma, or tissue samples have been associated with autoimmune conditions such as rheumatoid arthritis, systemic lupus erythematosus, and inflammatory bowel disease. In oncology, increased IL15 expression has been observed in certain hematologic malignancies and solid tumors, and it has been studied in the context of tumor immunology and response to immunotherapies. Additionally, IL15 has been explored as an indicator of immune activation in viral infections, such as HIV and influenza. Its measurement may provide information about immune status, disease activity, or therapeutic response in these settings. |
| interleukin 4 (IL4) | Interleukin 4 (IL4) is a cytokine produced primarily by activated T helper 2 (Th2) cells, mast cells, and basophils. It plays a central role in the regulation of immune responses, particularly in the differentiation of naive helper T cells (Th0) into Th2 cells. IL4 promotes the production and class switching of immunoglobulin E (IgE) and IgG1 by B cells, contributing to humoral immunity. It also inhibits the production of pro-inflammatory cytokines and supports the proliferation and survival of B cells, T cells, and mast cells. IL4 is involved in the modulation of allergic inflammation and is a key mediator in the pathogenesis of atopic diseases. | IL4 has been utilized as a biomarker in the context of allergic diseases, such as asthma, atopic dermatitis, and allergic rhinitis, due to its association with Th2-driven immune responses and elevated IgE levels. Measurement of IL4 levels in serum, plasma, or tissue samples can reflect the degree of type 2 immune activation. Additionally, IL4 has been studied as a biomarker in certain autoimmune diseases and in monitoring immune responses during infections and immunotherapies. Its application focuses on assessing immune status, disease activity, and response to treatment in relevant conditions. |
| interleukin 5 (IL5) | Interleukin 5 (IL5) is a cytokine primarily produced by activated T helper 2 (Th2) cells, mast cells, and eosinophils. Its main biological function is to regulate the growth, differentiation, recruitment, activation, and survival of eosinophils, a type of white blood cell involved in immune responses, particularly in allergic inflammation and defense against parasitic infections. IL5 exerts its effects by binding to the IL5 receptor, which is expressed predominantly on eosinophils and basophils, leading to downstream signaling that promotes eosinophil proliferation and activation. | IL5 has been utilized as a biomarker in conditions characterized by eosinophilic inflammation, such as asthma, eosinophilic esophagitis, and certain hypereosinophilic syndromes. Measurement of IL5 levels in blood, sputum, or tissue samples can reflect the degree of Th2-mediated immune activity and eosinophilic involvement. Its assessment can aid in the characterization of disease endotypes, monitoring of disease activity, and evaluation of responses to therapies targeting the IL5 pathway. |
| interleukin 5 receptor subunit alpha (IL5RA) | Interleukin 5 receptor subunit alpha (IL5RA) encodes the alpha subunit of the interleukin-5 receptor complex. IL5RA is primarily expressed on the surface of eosinophils, as well as basophils and certain progenitor cells. Upon binding to its ligand, interleukin-5 (IL-5), IL5RA associates with the common beta subunit (CSF2RB) to form a functional receptor complex. This receptor mediates the activation, proliferation, differentiation, and survival of eosinophils in response to IL-5. Through these actions, IL5RA plays a central role in the regulation of eosinophilic inflammation and immune responses, particularly in the context of allergic diseases and parasitic infections. | IL5RA has been utilized as a biomarker for identifying and characterizing eosinophil populations in blood and tissue samples, due to its selective expression on eosinophils. Measurement of IL5RA expression levels can assist in the assessment of eosinophil activation and abundance, which is relevant in conditions such as asthma, hypereosinophilic syndrome, and other eosinophil-associated disorders. Additionally, IL5RA serves as a target for monoclonal antibody therapies designed to modulate eosinophil activity in certain inflammatory diseases. |
| prostaglandin D2 receptor 2 (PTGDR2) | Prostaglandin D2 receptor 2 (PTGDR2), also known as CRTH2, is a G protein-coupled receptor that binds prostaglandin D2 (PGD2). It is primarily expressed on Th2 lymphocytes, eosinophils, basophils, and certain innate lymphoid cells. Upon activation by PGD2, PTGDR2 mediates chemotaxis and activation of these immune cells, contributing to type 2 immune responses. This signaling pathway is involved in the regulation of allergic inflammation, including the recruitment of Th2 cells and eosinophils to sites of inflammation, and plays a role in the pathophysiology of allergic diseases such as asthma, allergic rhinitis, and atopic dermatitis. | PTGDR2 expression has been investigated as a biomarker for Th2-driven inflammatory conditions. Its presence on specific immune cell subsets and its upregulation in tissues affected by allergic inflammation have made it a candidate for identifying and characterizing Th2-type immune responses in diseases such as asthma and allergic rhinitis. Measurement of PTGDR2 expression or the frequency of PTGDR2-positive cells has been used in research settings to help distinguish Th2-associated phenotypes and to monitor disease activity or response to therapy in allergic and eosinophilic disorders. |
| thymic stromal lymphopoietin (TSLP) | Thymic stromal lymphopoietin (TSLP) is a cytokine primarily produced by epithelial cells, including those in the skin, lungs, and gastrointestinal tract. TSLP plays a key role in the regulation of immune responses, particularly at barrier surfaces. It acts on dendritic cells, promoting their maturation and enhancing their ability to prime naïve T cells towards a T helper 2 (Th2) phenotype. This cytokine also influences the activity of other immune cells, such as basophils, mast cells, and type 2 innate lymphoid cells (ILC2s), contributing to the orchestration of type 2 immune responses. Through these actions, TSLP is involved in the initiation and propagation of allergic inflammation and maintenance of mucosal homeostasis. | TSLP has been investigated as a biomarker in various allergic and inflammatory diseases, including asthma, atopic dermatitis, and eosinophilic esophagitis. Elevated levels of TSLP have been detected in affected tissues and biological fluids of individuals with these conditions, correlating with disease activity and severity in some studies. Measurement of TSLP expression or protein concentration is used in research settings to assess its involvement in disease pathogenesis, monitor response to therapy, and stratify patients in clinical studies. |
Explore Research Opportunities with Protheragen. Our biomarker research services offer advanced analytical platforms and scientific expertise for the exploratory investigation of molecular targets in Eosinophilic Esophagitis. All biomarkers discussed are research targets only; we do not claim any as validated or mandatory for any application. Our work is exclusively focused on preclinical research and early drug discovery, maintaining scientific objectivity and supporting your innovation in EoE research.
We invite you to connect with Protheragen to discuss collaborative opportunities in exploratory biomarker research for Eosinophilic Esophagitis. Let’s advance scientific understanding together through objective knowledge exchange and preclinical investigation.
Make Order
Experimental Scheme
Implementation
Conclusion