Protheragen offers specialized biomarker analysis services tailored for Food Allergy research and development, with a comprehensive biomarker panel designed to elucidate the complex pathophysiology underlying food-induced allergic responses. Our services are exclusively dedicated to supporting drug discovery and preclinical development initiatives, and do not encompass clinical diagnostic applications. By integrating advanced scientific methodologies, we empower researchers and pharmaceutical partners to accelerate the identification and characterization of novel therapeutic targets relevant to Food Allergy.
Effective therapeutic intervention in Food Allergy begins with the robust discovery and identification of relevant biomarkers. At Protheragen, our biomarker discovery services form the foundation of innovative drug development by systematically exploring molecular signatures associated with disease mechanisms. We employ a multi-step approach, starting with high-throughput screening of candidate genes, proteins, and regulatory RNAs, followed by rigorous validation using state-of-the-art analytical platforms. This process ensures that only the most promising biomarkers, with potential relevance to Food Allergy pathophysiology, advance for further investigation in preclinical models.
Multi Omics: Our biomarker discovery pipeline leverages cutting-edge -omics technologies, including genomics, transcriptomics, proteomics, and metabolomics, to provide a comprehensive view of biological systems involved in Food Allergy. By integrating data from DNA, RNA, protein, and metabolite analyses, we uncover complex molecular networks and signaling pathways implicated in allergic responses. This multi-omics strategy enables the identification of diverse biomarker classes, such as cytokines (e.g., interleukins), transcription factors, and non-coding RNAs, that may contribute to immune dysregulation, epithelial barrier function, and inflammatory processes central to Food Allergy. Key pathways investigated include Th2-mediated immunity, cytokine signaling, and epithelial-immune cell interactions.
Candidate Validation: Protheragen applies rigorous validation strategies to prioritize biomarker candidates with the strongest association to Food Allergy pathophysiology. Initial screening involves quantitative and qualitative assessments in relevant preclinical models, followed by orthogonal validation using independent analytical techniques. Candidates are evaluated based on their specificity, sensitivity, reproducibility, and biological plausibility. Only those meeting stringent criteria—such as robust differential expression in Food Allergy models and mechanistic links to allergic inflammation—are advanced for further assay development and functional characterization.
Diverse Technological Platforms: We offer custom biomarker assay development tailored to the unique requirements of Food Allergy research, utilizing a spectrum of advanced technological platforms. Our laboratory infrastructure supports the adaptation and optimization of assays for diverse sample types and analytical endpoints, ensuring compatibility with the latest immunoassay, molecular, and imaging technologies to address specific research objectives.
Immunoassays: We develop and implement enzyme-linked immunosorbent assays (ELISA), chemiluminescent immunoassays, and multiplex immunoassay platforms to enable sensitive and specific quantification of cytokines, chemokines, and other protein biomarkers relevant to Food Allergy.
Mass Spectrometry: Liquid chromatography-tandem mass spectrometry (LC-MS/MS) is employed for targeted and untargeted proteomic and metabolomic profiling, enabling high-resolution detection and quantitation of proteins, peptides, and metabolites involved in allergic responses.
Flow Cytometry: We utilize multiparametric flow cytometry to characterize immune cell subsets, assess surface and intracellular marker expression, and quantify cellular responses associated with Food Allergy models.
Molecular Diagnostics: Our capabilities include quantitative PCR (qPCR), digital PCR, and next-generation sequencing (NGS) for the detection and quantification of gene expression, microRNAs (e.g., MIR155), and genetic variants pertinent to allergic disease mechanisms.
Histopathology And Imaging: We offer immunohistochemistry and advanced imaging modalities to visualize and localize biomarker expression within tissue samples, supporting the spatial analysis of immune and epithelial cell interactions in Food Allergy.
Rigorous Method Validation: Each analytical method developed at Protheragen undergoes a rigorous validation process consistent with established guidelines for preclinical research. Performance characteristics such as accuracy, precision, sensitivity, specificity, linearity, and reproducibility are systematically assessed. Comprehensive quality control measures, including the use of internal standards, calibration controls, and replicate analyses, ensure data integrity and reliability throughout the biomarker analysis workflow.
Our quantitative analysis capabilities enable precise measurement of biomarker concentrations and expression levels across a wide dynamic range. Standardized protocols and calibration strategies are implemented to ensure reproducibility and comparability of results across different studies and sample types. Data analysis is supported by robust statistical and bioinformatic tools for interpretation and integration of multi-omics datasets.
Sample Analysis: Protheragen handles a variety of sample types, including blood, serum, plasma, tissue lysates, and cell culture supernatants, relevant to Food Allergy research. Each sample is processed according to standardized protocols that preserve biomarker integrity and minimize pre-analytical variability. Stringent quality control measures are enforced at every stage, from sample receipt through data reporting, to ensure the validity and traceability of all results.
High Throughput Capabilities: Our high-throughput analytical platforms enable multiplexed biomarker detection, allowing simultaneous analysis of multiple targets from limited sample volumes. This approach enhances efficiency, reduces turnaround times, and conserves valuable biological specimens. Automated workflows and scalable assay formats support large-scale preclinical studies and screening campaigns.
| Gene Target | Biological Function | Application as a Biomarker |
|---|---|---|
| KIT proto-oncogene, receptor tyrosine kinase (KIT) | The KIT proto-oncogene, receptor tyrosine kinase (KIT), encodes a transmembrane receptor that belongs to the type III receptor tyrosine kinase family. KIT is primarily activated by its ligand, stem cell factor (SCF), leading to receptor dimerization and autophosphorylation of intracellular tyrosine residues. This activation triggers multiple downstream signaling pathways, including the PI3K/AKT, RAS/MAPK, and JAK/STAT pathways, which regulate diverse cellular processes such as proliferation, differentiation, survival, and apoptosis. KIT plays essential roles in the development and maintenance of several cell types, notably hematopoietic stem cells, melanocytes, germ cells, and interstitial cells of Cajal. | KIT is used as a biomarker in various clinical and research settings, particularly in oncology and hematopathology. Immunohistochemical detection of KIT (CD117) is commonly employed to aid in the diagnosis of gastrointestinal stromal tumors (GISTs), where KIT expression is frequently observed. KIT mutations and expression are also assessed in certain subtypes of acute myeloid leukemia, mastocytosis, melanoma, and seminoma. The presence or absence of KIT expression, as well as specific KIT gene mutations, can provide diagnostic, prognostic, and therapeutic information in these contexts. |
| interleukin 10 (IL10) | Interleukin 10 (IL10) is an anti-inflammatory cytokine primarily produced by monocytes, regulatory T cells, and certain subsets of lymphocytes. IL10 plays a central role in limiting immune responses by inhibiting the synthesis of pro-inflammatory cytokines such as IFN-γ, IL-2, IL-3, TNF-α, and GM-CSF produced by activated macrophages and helper T cells. It also suppresses antigen presentation by downregulating the expression of major histocompatibility complex (MHC) class II molecules and co-stimulatory molecules on antigen-presenting cells. Through these mechanisms, IL10 helps maintain immune homeostasis and prevents excessive tissue damage during immune responses. | IL10 levels have been measured in biological fluids to assess immune status in various clinical contexts. Elevated or decreased IL10 concentrations have been reported in inflammatory and autoimmune diseases, infectious diseases, and certain cancers. In these settings, IL10 has been used to monitor disease activity, evaluate prognosis, and study response to therapy. Its measurement can provide information about the balance between pro- and anti-inflammatory signals in the body. |
| interleukin 13 (IL13) | Interleukin 13 (IL13) is a cytokine primarily produced by activated T helper type 2 (Th2) cells. It plays a key role in the regulation of immune responses, particularly in the modulation of inflammatory and allergic processes. IL13 influences the differentiation and proliferation of B cells, promotes immunoglobulin E (IgE) class switching, and stimulates the production of mucus by airway epithelial cells. It also affects macrophage activation and contributes to tissue remodeling and fibrosis. IL13 exerts its effects through binding to its receptor complex, which includes IL13 receptor alpha 1 (IL13RA1) and IL4 receptor alpha (IL4RA) subunits, leading to downstream signaling pathways such as STAT6 activation. | IL13 levels have been investigated as a biomarker in various conditions characterized by immune dysregulation, particularly in allergic diseases such as asthma and atopic dermatitis. Elevated IL13 expression or protein concentrations in blood, bronchoalveolar lavage fluid, or tissue samples have been associated with Th2-type inflammation and disease severity in these disorders. Measurement of IL13 may assist in identifying inflammatory phenotypes and monitoring responses to targeted therapies in clinical and research settings. |
| interleukin 17A (IL17A) | Interleukin 17A (IL17A) is a pro-inflammatory cytokine produced primarily by a subset of T helper cells known as Th17 cells, as well as by other immune cells such as γδ T cells and innate lymphoid cells. IL17A plays a critical role in host defense against extracellular bacteria and fungi by promoting the recruitment and activation of neutrophils and stimulating the production of antimicrobial peptides. It acts by binding to the IL-17 receptor complex on various cell types, leading to the induction of pro-inflammatory cytokines, chemokines, and matrix metalloproteinases. IL17A is involved in the regulation of immune responses at mucosal surfaces and contributes to the maintenance of tissue integrity during infection and inflammation. | IL17A has been utilized as a biomarker to assess inflammatory activity in various immune-mediated and autoimmune conditions. Elevated levels of IL17A in serum, plasma, or tissue samples have been associated with diseases such as psoriasis, rheumatoid arthritis, ankylosing spondylitis, and multiple sclerosis. Measurement of IL17A can aid in evaluating disease presence, activity, and response to therapy in these contexts. Its detection is also used in research to characterize Th17-mediated immune responses and to monitor the effects of immunomodulatory treatments targeting the IL-17 pathway. |
| interleukin 2 (IL2) | Interleukin 2 (IL2) is a cytokine produced primarily by activated CD4+ T lymphocytes. It plays a central role in the regulation of immune responses by promoting the proliferation, differentiation, and survival of T cells, including both effector and regulatory T cell subsets. IL2 also supports the growth and activity of natural killer (NK) cells and influences B cell function. Its signaling is mediated through the IL2 receptor, which is composed of three subunits (alpha, beta, and gamma chains) and activates downstream pathways that are critical for immune cell homeostasis and tolerance. | IL2 has been utilized as a biomarker to assess immune activation and function, particularly in contexts such as transplantation, autoimmune diseases, infectious diseases, and cancer immunotherapy. Measurement of IL2 levels in serum, plasma, or cell culture supernatants can provide information about T cell activation status and immune response dynamics. Changes in IL2 expression or secretion have been associated with disease activity, prognosis, and response to immunomodulatory therapies in various clinical settings. |
| interleukin 4 (IL4) | Interleukin 4 (IL4) is a cytokine produced primarily by activated T helper 2 (Th2) cells, mast cells, and basophils. IL4 plays a central role in the regulation of immune responses, particularly in the differentiation of naive CD4+ T cells into Th2 cells. It promotes B cell proliferation, survival, and class switching to immunoglobulin E (IgE) and IgG1. IL4 also inhibits the production of pro-inflammatory cytokines and supports the alternative activation of macrophages. Through these actions, IL4 is involved in humoral immunity, allergic responses, and the modulation of inflammation. | IL4 has been studied as a biomarker in various contexts, including allergic diseases such as asthma, atopic dermatitis, and allergic rhinitis, where elevated levels are often observed. It is also investigated in autoimmune conditions and certain cancers to assess immune status and disease progression. Measurement of IL4 in serum, plasma, or tissue samples can provide information about Th2-mediated immune activity and may assist in disease characterization, monitoring of treatment response, and stratification of disease subtypes. |
| microRNA 155 (MIR155) | microRNA 155 (MIR155) is a small non-coding RNA molecule that plays a critical role in the regulation of gene expression at the post-transcriptional level. MIR155 is processed from the MIR155 host gene (MIR155HG, also known as BIC) and is highly conserved among vertebrates. It is predominantly expressed in cells of the immune system, including B cells, T cells, macrophages, and dendritic cells. MIR155 modulates various biological processes such as immune response, inflammation, hematopoiesis, and cell differentiation by targeting messenger RNAs (mRNAs) for degradation or translational repression. Its regulatory targets include genes involved in cytokine production, cell proliferation, and apoptosis, positioning MIR155 as a key modulator in both innate and adaptive immunity. | MIR155 has been studied as a biomarker in a range of pathological conditions, particularly in oncology and immunology. Elevated levels of MIR155 have been observed in several types of cancers, including lymphomas, breast cancer, and lung cancer, as well as in inflammatory and autoimmune diseases such as rheumatoid arthritis and multiple sclerosis. Its expression profile in tissues, blood, or other body fluids has been investigated for potential use in disease diagnosis, prognosis, and monitoring of disease progression or response to therapy. The measurement of MIR155 levels is typically performed using quantitative PCR or next-generation sequencing techniques. |
| signal transducer and activator of transcription 5A (STAT5A) | Signal transducer and activator of transcription 5A (STAT5A) is a member of the STAT family of transcription factors. STAT5A is activated by phosphorylation in response to cytokines and growth factors, such as interleukins, erythropoietin, prolactin, and growth hormone. Upon activation, STAT5A dimerizes and translocates to the nucleus, where it binds to specific DNA sequences to regulate the transcription of target genes. STAT5A plays critical roles in cell proliferation, differentiation, apoptosis, and immune function. It is particularly important in hematopoietic cell development, mammary gland development, and the regulation of immune responses. | STAT5A expression and activation status have been studied as biomarkers in various contexts, including hematological malignancies, breast cancer, and immune-related disorders. In certain leukemias and lymphomas, aberrant STAT5A activation has been associated with disease progression and prognosis. In breast tissue, STAT5A has been examined in relation to differentiation status and tumor subtype. Additionally, STAT5A activity has been evaluated as an indicator of cytokine signaling pathway engagement in immune cells. |
| signal transducer and activator of transcription 6 (STAT6) | Signal transducer and activator of transcription 6 (STAT6) is a member of the STAT family of transcription factors. STAT6 is primarily activated by interleukin-4 (IL-4) and interleukin-13 (IL-13) through the Janus kinase (JAK)-STAT signaling pathway. Upon cytokine stimulation, STAT6 becomes phosphorylated, dimerizes, and translocates to the nucleus, where it regulates the expression of genes involved in immune responses. STAT6 plays a key role in the differentiation of T helper 2 (Th2) cells, immunoglobulin class switching to IgE in B cells, and the regulation of genes associated with allergic inflammation and anti-parasitic responses. | STAT6 has been utilized in diagnostic pathology, particularly as an immunohistochemical marker for the identification of solitary fibrous tumors (SFT). Nuclear expression of STAT6, resulting from NAB2-STAT6 gene fusions, is highly characteristic of SFT and aids in distinguishing these tumors from histologic mimics. Additionally, STAT6 expression and activation patterns have been studied in the context of allergic diseases, asthma, and certain malignancies to provide insights into disease mechanisms and cellular phenotypes. |
| thymic stromal lymphopoietin (TSLP) | Thymic stromal lymphopoietin (TSLP) is a cytokine primarily produced by epithelial cells, including those in the skin, lungs, and gut. TSLP plays a central role in the regulation of immune responses, particularly at barrier surfaces. It is involved in the maturation and activation of dendritic cells, which in turn promote the differentiation of naïve T cells toward the T helper 2 (Th2) lineage. TSLP also influences other immune cells, such as basophils, mast cells, and group 2 innate lymphoid cells (ILC2s), facilitating the production of Th2-type cytokines. Through these actions, TSLP contributes to the initiation and maintenance of allergic inflammation and is implicated in the pathogenesis of various atopic and inflammatory diseases. | TSLP has been studied as a biomarker in several disease contexts, particularly in allergic and inflammatory conditions. Elevated levels of TSLP have been detected in tissue and biological fluids from individuals with diseases such as atopic dermatitis, asthma, and eosinophilic esophagitis. Measurement of TSLP expression or concentration can provide information about the presence and activity of type 2 immune responses, and may be used in research and clinical studies to assess disease severity, monitor response to therapy, or stratify patient populations. |
Explore Research Opportunities with Protheragen. Our biomarker research services provide comprehensive support for Food Allergy drug discovery and preclinical development, leveraging advanced technologies and a robust panel of candidate biomarkers. Please note that all biomarkers discussed are for research purposes only; we do not claim any as validated or mandatory for Food Allergy studies. Our focus remains on the exploratory and investigative stages of preclinical research, maintaining strict scientific objectivity in all collaborations.
We invite you to engage with Protheragen to discuss collaborative biomarker research in Food Allergy. Our approach emphasizes scientific exploration and knowledge exchange, without making claims regarding biomarker validation or necessity. Connect with us to advance the understanding of Food Allergy mechanisms and drive innovation in preclinical research.
Make Order
Experimental Scheme
Implementation
Conclusion