Protheragen offers specialized biomarker analysis services tailored for Graft-Versus-Host Disease (GVHD) research and drug discovery. Our comprehensive biomarker panel is designed to facilitate a deep understanding of GVHD pathophysiology, supporting the identification and characterization of molecular targets relevant to immune dysregulation and inflammation. All services are exclusively focused on preclinical therapeutic development and drug discovery, and do not include clinical diagnostic offerings.
Effective therapeutic intervention for GVHD begins with the discovery and identification of robust biomarkers that illuminate disease mechanisms and immune dysregulation. Protheragen’s biomarker discovery services leverage advanced screening platforms to identify candidate molecules associated with GVHD. Our iterative process includes high-throughput screening, in silico analyses, and experimental validation to ensure that selected biomarkers are relevant to disease biology and suitable for integration into drug development pipelines. Rigorous validation steps establish the biological significance and reproducibility of each candidate, supporting their application in preclinical models.
Multi Omics: Our multi-omics approach integrates genomics, transcriptomics, proteomics, and metabolomics to provide a holistic view of biological systems in GVHD. Utilizing next-generation sequencing, quantitative PCR, RNA-Seq, mass spectrometry, and comprehensive protein profiling, we identify DNA, RNA, protein, and metabolite biomarkers that reflect key immune and inflammatory pathways. This systems-level analysis enables the elucidation of complex disease networks, including cytokine signaling, immune cell activation, and inflammatory cascades central to GVHD pathophysiology.
Candidate Validation: Candidate validation at Protheragen involves a series of strategic assays and functional studies to confirm the association of identified biomarkers with GVHD pathophysiology. Preliminary screening includes quantitative assessment in relevant preclinical models, correlation with disease phenotypes, and evaluation of expression patterns in immune cell populations. Prioritization criteria include biological plausibility, reproducibility, dynamic range, and relevance to immune response modulation, ensuring that only the most promising candidates progress to further assay development.
Diverse Technological Platforms: We offer custom assay development across diverse technological platforms, adapting methodologies to meet specific research objectives for GVHD. Our capabilities extend from single-plex to multiplexed formats, supporting both exploratory and quantitative studies. Platforms are selected and optimized based on biomarker class (protein, nucleic acid, cell surface marker) and required sensitivity, facilitating seamless integration into preclinical research workflows.
Immunoassays: We develop and implement ELISA, chemiluminescent, and multiplex immunoassays for sensitive and specific quantification of cytokines, chemokines, and soluble receptors implicated in GVHD.
Mass Spectrometry: Our LC-MS/MS platforms enable quantitative proteomics and targeted metabolite profiling, supporting the identification and validation of protein and small molecule biomarkers.
Flow Cytometry: We use multiparametric flow cytometry to characterize immune cell subsets, activation markers, and surface receptor expression relevant to GVHD immune responses.
Molecular Diagnostics: We apply qPCR, digital PCR, and nucleic acid amplification techniques for the detection and quantification of gene expression and genetic variants associated with disease pathways.
Histopathology And Imaging: Our services include immunohistochemistry, in situ hybridization, and advanced imaging to localize and quantify biomarkers in tissue sections, supporting spatial mapping of disease processes.
Rigorous Method Validation: All assay methods are validated in accordance with established research guidelines, including assessment of accuracy, precision, specificity, sensitivity, linearity, and reproducibility. We implement stringent quality control measures, including the use of reference standards, controls, and inter-assay comparisons, to ensure robust and reliable performance of biomarker assays throughout preclinical development.
Protheragen provides quantitative analysis capabilities for a wide range of biomarker classes. Our platforms support absolute and relative quantification, enabling detailed profiling of biomarker dynamics in GVHD models. Data analysis includes normalization, statistical evaluation, and integration with multi-omics datasets to generate actionable insights for therapeutic research.
Sample Analysis: We process and analyze a variety of sample types relevant to GVHD research, including serum, plasma, tissue homogenates, and cell lysates. Standardized protocols govern sample handling, preparation, and storage to preserve biomarker integrity. Each analytical run is subject to rigorous quality assurance procedures, including sample tracking, duplicate analysis, and inclusion of negative and positive controls.
High Throughput Capabilities: Our high-throughput analytical platforms enable multiplexed biomarker analysis, allowing simultaneous quantification of multiple targets in limited sample volumes. This approach enhances efficiency, conserves valuable preclinical samples, and accelerates data generation for comprehensive biomarker profiling in GVHD research.
| Gene Target | Biological Function | Application as a Biomarker |
|---|---|---|
| interferon gamma (IFNG) | Interferon gamma (IFNG) is a cytokine produced primarily by activated T lymphocytes and natural killer (NK) cells. It plays a central role in innate and adaptive immunity by promoting the activation of macrophages, enhancing antigen presentation through upregulation of major histocompatibility complex (MHC) molecules, and stimulating the differentiation of T helper 1 (Th1) cells. IFNG also regulates the expression of numerous genes involved in immune responses, inhibits viral replication, and modulates the activity of various immune cells. Its actions are critical for host defense against intracellular pathogens and for the orchestration of immune-mediated inflammation. | IFNG is utilized as a biomarker to assess cell-mediated immune responses, particularly in the context of infectious diseases such as tuberculosis, where interferon gamma release assays (IGRAs) measure IFNG production following antigen-specific stimulation. It is also measured in research and clinical studies to evaluate immune activation status, monitor responses to immunotherapies, and investigate immune dysregulation in conditions such as autoimmune diseases, cancer, and transplant rejection. |
| interleukin 1 beta (IL1B) | Interleukin 1 beta (IL1B) is a pro-inflammatory cytokine produced primarily by activated macrophages, as well as other cell types such as monocytes, dendritic cells, and epithelial cells. It is synthesized as an inactive precursor (pro-IL1B) and requires cleavage by caspase-1 within the inflammasome complex to become biologically active. IL1B plays a central role in the regulation of immune and inflammatory responses by promoting the expression of adhesion molecules on endothelial cells, stimulating the production of other cytokines and chemokines, and inducing fever through its action on the hypothalamus. It also contributes to the activation and recruitment of immune cells to sites of infection or injury, and influences processes such as cell proliferation, differentiation, and apoptosis. | IL1B is used as a biomarker to assess the presence and extent of inflammation in various biological contexts. Elevated levels of IL1B in blood, tissue, or other biological fluids have been associated with inflammatory and autoimmune diseases, such as rheumatoid arthritis, inflammatory bowel disease, and sepsis, as well as with certain infections and tissue injuries. Measurement of IL1B can provide information on the activation of the innate immune response and the inflammatory state of an individual. It is also utilized in research and clinical studies to monitor disease progression, response to therapy, and to investigate the pathophysiology of inflammatory conditions. |
| interleukin 1 receptor like 1 (IL1RL1) | Interleukin 1 receptor like 1 (IL1RL1), also known as ST2, is a member of the interleukin-1 receptor family. IL1RL1 exists in both membrane-bound and soluble isoforms. It primarily functions as the receptor for interleukin-33 (IL-33), mediating the biological effects of IL-33 by activating intracellular signaling pathways such as NF-κB and MAP kinases. This interaction leads to the modulation of immune responses, particularly by promoting T-helper type 2 (Th2) cell responses, and is involved in the regulation of inflammation, tissue repair, and homeostasis. IL1RL1 is expressed on various immune cells, including mast cells, eosinophils, basophils, and certain T cell subsets. | IL1RL1 has been studied as a biomarker in several clinical contexts. Elevated levels of its soluble form (sST2) in blood have been associated with cardiovascular diseases, particularly heart failure, where increased sST2 concentrations correlate with disease severity and prognosis. Additionally, IL1RL1 expression and circulating levels have been investigated in relation to inflammatory and allergic diseases, such as asthma, where they may reflect disease activity or severity. |
| interleukin 10 (IL10) | Interleukin 10 (IL10) is an anti-inflammatory cytokine produced by a variety of immune cells, including T cells, B cells, monocytes, and macrophages. IL10 plays a critical role in limiting immune responses and preventing tissue damage by suppressing the expression of pro-inflammatory cytokines, chemokines, and antigen presentation. It inhibits the activation and effector functions of T cells, macrophages, and dendritic cells, thereby regulating immune homeostasis. IL10 achieves its effects primarily through the IL10 receptor, activating the JAK-STAT signaling pathway, which leads to transcriptional regulation of target genes involved in immune modulation. | IL10 levels are measured in biological fluids such as serum, plasma, or tissue samples as an indicator of immune status and inflammatory activity. Increased or decreased IL10 concentrations have been associated with various clinical conditions, including autoimmune diseases, infectious diseases, and cancer. In clinical research, IL10 is used to assess the balance between pro- and anti-inflammatory responses, monitor disease progression, and evaluate responses to therapeutic interventions targeting the immune system. |
| 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 other immune cells such as γδ T cells, natural killer (NK) cells, and innate lymphoid cells. IL17A plays a central role in the regulation of immune responses, particularly in host defense against extracellular bacteria and fungi. It exerts its effects by binding to the IL-17 receptor complex on target cells, leading to the induction of pro-inflammatory mediators, including cytokines, chemokines, and matrix metalloproteinases. This signaling promotes the recruitment and activation of neutrophils and other immune cells to sites of infection or inflammation. IL17A is also involved in the maintenance of mucosal barriers and has been implicated in the pathogenesis of various autoimmune and inflammatory diseases due to its potent pro-inflammatory properties. | IL17A has been investigated as a biomarker of immune activation and inflammation in several clinical contexts. Elevated levels of IL17A have been observed in the blood, tissues, or biological fluids of individuals with autoimmune diseases such as psoriasis, rheumatoid arthritis, ankylosing spondylitis, and multiple sclerosis, as well as in certain infectious and allergic conditions. Measurement of IL17A concentrations may provide information regarding disease activity, inflammatory status, or therapeutic response in these settings. Its application as a biomarker is primarily based on its association with the presence and severity of inflammatory and autoimmune processes. |
| interleukin 2 (IL2) | Interleukin 2 (IL2) is a cytokine primarily produced by activated CD4+ T lymphocytes and, to a lesser extent, by CD8+ T cells and natural killer (NK) cells. IL2 plays a central role in the immune system by promoting the proliferation, differentiation, and survival of T cells following antigen stimulation. It is essential for the development of regulatory T cells (Tregs), which help maintain immune tolerance and prevent autoimmunity. IL2 also enhances the cytotoxic activity of NK cells and supports B cell proliferation and antibody production. Through these actions, IL2 is a key regulator of immune responses and homeostasis. | IL2 levels in biological fluids, such as serum or plasma, have been used as an indicator of immune activation or dysregulation. Elevated IL2 concentrations have been observed in conditions characterized by immune activation, including certain autoimmune diseases, infections, and following immunotherapy. Measurement of IL2 can be utilized to monitor immune responses in clinical and research settings, such as assessing the efficacy of immunomodulatory treatments or tracking immune reconstitution after transplantation. Additionally, IL2 production by lymphocytes can be used in functional assays to evaluate T cell activation. |
| interleukin 2 receptor subunit alpha (IL2RA) | Interleukin 2 receptor subunit alpha (IL2RA), also known as CD25, is a component of the high-affinity interleukin-2 (IL-2) receptor complex. IL2RA combines with the beta (CD122) and gamma (CD132) subunits to form the trimeric IL-2 receptor, which is primarily expressed on activated T lymphocytes, regulatory T cells (Tregs), and certain B cells. The alpha subunit alone binds IL-2 with low affinity, but its association with the beta and gamma chains is required for high-affinity binding and subsequent signal transduction. IL2RA plays a critical role in regulating immune responses by mediating IL-2-driven proliferation, differentiation, and survival of lymphocytes, particularly in the maintenance and function of regulatory T cells, which are essential for immune tolerance. | IL2RA is utilized as a biomarker in several clinical and research contexts. Its expression on activated T cells and regulatory T cells is measured to assess immune activation and regulation. Soluble IL2RA (sCD25), which is released into the circulation upon shedding from cell surfaces, is quantified in serum or plasma as an indicator of immune system activation. Elevated levels of IL2RA or sCD25 have been associated with various conditions, including hematologic malignancies (such as adult T-cell leukemia/lymphoma), autoimmune diseases (such as type 1 diabetes and multiple sclerosis), and in the monitoring of transplant rejection. Measurement of IL2RA levels can aid in disease monitoring, prognosis, and assessment of therapeutic response. |
| 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 by promoting the differentiation of naive T cells into Th2 cells and stimulating B cell proliferation, survival, and class switching to immunoglobulin E (IgE) and IgG1. IL4 also modulates the activity of macrophages, inhibiting pro-inflammatory functions and promoting alternative (M2) activation. Through these actions, IL4 is involved in the regulation of humoral immunity, allergic responses, and the suppression of certain inflammatory processes. | IL4 has been utilized as a biomarker in clinical and research settings to assess immune system activity, particularly in contexts involving allergic diseases, asthma, and certain autoimmune disorders. Elevated levels of IL4 in serum, plasma, or tissue samples have been associated with Th2-skewed immune responses, providing information about disease activity, severity, or response to therapy in conditions such as atopic dermatitis, allergic rhinitis, and asthma. Measurement of IL4 can also contribute to the characterization of immune profiles in various disease states. |
| interleukin 6 (IL6) | Interleukin 6 (IL6) is a multifunctional cytokine produced by a variety of cell types, including T cells, B cells, macrophages, fibroblasts, and endothelial cells. It plays a central role in the regulation of immune responses, inflammation, hematopoiesis, and the acute phase response. IL6 is involved in stimulating the production of acute-phase proteins in the liver, promoting the differentiation of B cells into antibody-producing plasma cells, and influencing the differentiation of T cells. It also affects metabolic, regenerative, and neural processes. IL6 exerts its effects through binding to the IL6 receptor (IL6R), triggering downstream signaling pathways such as the JAK/STAT pathway. | IL6 is widely measured in clinical and research settings as a biomarker of inflammation. Elevated levels of IL6 in serum or plasma have been associated with various inflammatory and infectious diseases, including sepsis, rheumatoid arthritis, and COVID-19. It is also monitored in some cancers and chronic diseases to assess disease activity, prognosis, or response to therapy. IL6 concentrations are used to help characterize the severity of inflammatory responses and to support differential diagnosis in conjunction with other clinical and laboratory findings. |
| tumor necrosis factor (TNF) | Tumor necrosis factor (TNF) is a pro-inflammatory cytokine primarily produced by activated macrophages, as well as other immune cells such as T lymphocytes and natural killer cells. TNF plays a central role in the regulation of immune responses, inflammation, and apoptosis. It mediates its effects by binding to two distinct receptors, TNFR1 and TNFR2, leading to the activation of downstream signaling pathways such as NF-κB and MAPK. TNF is involved in the induction of fever, recruitment of immune cells to sites of infection or injury, and modulation of cell proliferation, differentiation, and survival. It is also implicated in the pathogenesis of various inflammatory and autoimmune diseases. | TNF is measured in biological fluids, such as serum or plasma, as an indicator of systemic or localized inflammation. Elevated TNF levels have been associated with a range of conditions, including rheumatoid arthritis, inflammatory bowel disease, sepsis, and certain cancers. Assessment of TNF concentrations can provide information about disease activity, severity, or response to therapy in inflammatory and autoimmune disorders. TNF is also used in research settings to monitor the efficacy of anti-TNF therapies. |
Explore Research Opportunities with Protheragen. Our biomarker research services for Graft-Versus-Host Disease offer a comprehensive suite of exploratory and analytical capabilities, supporting the identification and characterization of research targets relevant to immune modulation and inflammation. All biomarkers described herein are research targets only and are not claimed as validated or mandatory for any application. Our focus remains on preclinical research stages, providing objective scientific support for therapeutic discovery and development.
We invite you to connect with Protheragen to discuss collaborative biomarker research for Graft-Versus-Host Disease. Our approach emphasizes exploratory science, objective data generation, and open knowledge exchange. Let’s advance the understanding of disease biology together through preclinical research partnerships.
Make Order
Experimental Scheme
Implementation
Conclusion