Protheragen offers specialized biomarker analysis services tailored for Immunosuppression research and drug discovery, supporting the development of innovative therapeutics. Our comprehensive biomarker panel is designed to elucidate the underlying pathophysiology of immunosuppression, enabling a deeper understanding of disease mechanisms and therapeutic targets. All services are exclusively focused on drug discovery through preclinical development stages; we do not provide clinical diagnostic services.
Effective therapeutic intervention for immunosuppression begins with robust biomarker discovery and identification. At Protheragen, our biomarker discovery services are integral to the drug development pipeline, enabling the identification of molecular indicators that inform candidate selection and mechanism-of-action studies. We employ systematic screening and validation processes, including high-throughput screening, literature mining, and experimental validation, to ensure the relevance and reliability of identified biomarkers. Our approach supports the rational development of targeted therapeutics by linking molecular signatures to immunosuppressive states.
Multi Omics: Our multi-omics approach leverages cutting-edge technologies across genomics, transcriptomics, proteomics, and metabolomics to provide a comprehensive view of biological systems involved in immunosuppression. By integrating data from DNA, RNA, protein, and metabolite profiling, we identify and characterize biomarkers that reflect key disease pathways, such as cytokine signaling, immune cell activation, and regulatory networks. This holistic strategy enables the elucidation of complex molecular interactions and supports the discovery of novel targets relevant to immunosuppression.
Candidate Validation: Protheragen employs rigorous validation strategies to confirm the association of candidate biomarkers with immunosuppression pathophysiology. Our preliminary screening processes include reproducibility assessments, cross-platform comparisons, and functional studies to evaluate the biological significance of each candidate. Promising biomarkers are prioritized based on criteria such as specificity, sensitivity, biological relevance, and translational potential, ensuring that only the most informative markers advance through the pipeline.
Diverse Technological Platforms: We offer custom assay development capabilities, adapting technological platforms to meet specific project requirements. Our expertise spans a range of analytical systems, including immunoassays, mass spectrometry, flow cytometry, molecular diagnostics, and advanced imaging modalities. Each platform is selected and optimized based on target analyte characteristics, desired throughput, and sensitivity needs.
Immunoassays: We develop and implement a variety of immunoassays, including ELISA, chemiluminescent assays, and multiplex bead-based platforms, enabling sensitive and specific quantification of cytokines and other protein biomarkers.
Mass Spectrometry: Our LC-MS/MS capabilities facilitate high-resolution, quantitative analysis of proteins, peptides, and metabolites, supporting both targeted and untargeted biomarker discovery.
Flow Cytometry: We utilize flow cytometry for multiparametric analysis of immune cell populations, intracellular cytokine detection, and cell surface marker profiling relevant to immunosuppression.
Molecular Diagnostics: Our molecular diagnostic assays include quantitative PCR and digital PCR for gene expression analysis, as well as nucleic acid-based detection of genetic variants and transcriptomic signatures.
Histopathology And Imaging: Advanced histopathology and imaging techniques are employed for spatial localization and morphological assessment of biomarkers in tissue samples, providing context to molecular findings.
Rigorous Method Validation: All analytical methods undergo a rigorous validation process in accordance with relevant regulatory and industry guidelines. We assess performance characteristics such as accuracy, precision, sensitivity, specificity, linearity, and reproducibility. Stringent quality control measures are implemented throughout, including the use of reference standards, controls, and inter-assay comparisons to ensure data reliability and integrity.
We provide quantitative analysis capabilities across a range of biomarker types, enabling precise measurement of gene, protein, and metabolite levels. Our protocols are optimized for sensitivity and dynamic range, supporting both absolute and relative quantification. Data normalization and statistical analysis are integrated to ensure robust interpretation of results.
Sample Analysis: Protheragen handles a variety of sample types, including serum, plasma, tissue lysates, cell culture supernatants, and peripheral blood mononuclear cells. Each sample undergoes standardized processing and analysis protocols tailored to the biomarker of interest. Comprehensive quality measures, including sample integrity checks and process controls, are in place to maintain high standards throughout the analytical workflow.
High Throughput Capabilities: Our high-throughput analytical platforms enable multiplexed biomarker analysis, increasing efficiency and data output while conserving valuable samples. Multiplexing strategies allow simultaneous quantification of multiple analytes in a single run, reducing sample consumption and turnaround time. Automated systems and scalable workflows further enhance throughput, supporting large-scale discovery and validation projects.
| Gene Target | Biological Function | Application as a Biomarker |
|---|---|---|
| ATP binding cassette subfamily B member 1 (ABCB1) | ATP binding cassette subfamily B member 1 (ABCB1), also known as P-glycoprotein (P-gp) or MDR1, is a transmembrane protein that functions as an ATP-dependent efflux pump. It is primarily involved in the transport of a wide variety of substrates, including xenobiotics, drugs, and endogenous compounds, across cellular membranes. ABCB1 is highly expressed in tissues with barrier functions, such as the intestinal epithelium, blood-brain barrier, liver, and kidney. Its activity contributes to the absorption, distribution, and excretion of many pharmacological agents by actively exporting them out of cells, thereby influencing drug bioavailability and resistance. ABCB1 plays a significant role in protecting tissues from toxic substances and in the pharmacokinetics of numerous therapeutic drugs. | ABCB1 expression and activity have been studied as biomarkers in several contexts, particularly in relation to multidrug resistance (MDR) in cancer. Elevated levels of ABCB1 in tumor cells are associated with reduced intracellular accumulation of chemotherapeutic agents, which can affect treatment response. Assessment of ABCB1 status has been used to investigate correlations with drug resistance, prognosis, and therapeutic outcomes in oncology. Additionally, ABCB1 polymorphisms and expression levels have been explored as indicators of individual variability in drug pharmacokinetics and toxicity in clinical pharmacogenetics. |
| C-X-C motif chemokine ligand 8 (CXCL8) | C-X-C motif chemokine ligand 8 (CXCL8), also known as interleukin-8 (IL-8), is a pro-inflammatory chemokine produced by various cell types, including macrophages, epithelial cells, and endothelial cells. CXCL8 primarily functions as a chemoattractant, directing the migration of neutrophils and other immune cells to sites of infection or tissue injury. It binds to G protein-coupled receptors CXCR1 and CXCR2 on target cells, triggering intracellular signaling pathways that promote cell adhesion, migration, and activation. In addition to its role in immune cell recruitment, CXCL8 can influence angiogenesis, cell proliferation, and modulation of the inflammatory response. | CXCL8 has been utilized as a biomarker in a variety of clinical and research contexts. Elevated levels of CXCL8 have been associated with inflammatory and infectious diseases, such as sepsis, rheumatoid arthritis, and chronic obstructive pulmonary disease (COPD). In oncology, increased CXCL8 expression has been observed in several tumor types and may correlate with disease progression, tumor angiogenesis, and response to therapy. Measurement of CXCL8 in biological fluids, such as serum, plasma, or bronchoalveolar lavage, has been applied to assess disease activity, monitor treatment response, and evaluate prognosis in both inflammatory and malignant conditions. |
| interferon gamma (IFNG) | Interferon gamma (IFNG) is a cytokine produced primarily by activated T cells and natural killer (NK) cells. It plays a central role in innate and adaptive immunity by activating macrophages, enhancing antigen presentation, and promoting the differentiation of T helper 1 (Th1) cells. IFNG induces the expression of major histocompatibility complex (MHC) class I and II molecules, stimulates the production of other pro-inflammatory cytokines, and contributes to the control of intracellular pathogens such as viruses, bacteria, and certain parasites. Additionally, IFNG modulates immune responses by influencing the activity and proliferation of various immune cells. | IFNG is used as a biomarker to assess cell-mediated immune responses, particularly in the context of infectious diseases and immune-related conditions. Its measurement is applied in assays such as the interferon gamma release assay (IGRA) for the detection of latent or active Mycobacterium tuberculosis infection. IFNG levels are also evaluated in monitoring immune activation, assessing immune competence, and investigating autoimmune or inflammatory disorders. Its expression can reflect immune status and response to immunomodulatory therapies in various clinical and research settings. |
| interleukin 1 alpha (IL1A) | Interleukin 1 alpha (IL1A) is a pro-inflammatory cytokine belonging to the interleukin 1 family. It is primarily produced by activated macrophages, epithelial cells, and various other cell types in response to infection, injury, or immunological stimuli. IL1A acts as a key mediator of the inflammatory response, promoting the expression of adhesion molecules, chemokines, and other cytokines. It exerts its effects by binding to the interleukin 1 receptor type I (IL-1RI), leading to the activation of intracellular signaling pathways such as NF-κB and MAPKs. These pathways result in the transcription of genes involved in inflammation, immune cell recruitment, fever induction, and tissue remodeling. IL1A is also involved in the regulation of cell proliferation, differentiation, and apoptosis. | IL1A has been studied as a biomarker in various inflammatory and autoimmune conditions, including rheumatoid arthritis, systemic lupus erythematosus, and certain cancers. Elevated levels of IL1A in serum, plasma, or tissue samples have been associated with disease activity, severity, or prognosis in these contexts. Measurement of IL1A can be used in research settings to assess the degree of inflammation, monitor therapeutic response, or investigate the pathophysiology of inflammatory diseases. |
| interleukin 10 (IL10) | Interleukin 10 (IL10) is an anti-inflammatory cytokine produced by a variety of immune cells, including T cells, B cells, macrophages, dendritic cells, and certain subsets of regulatory T cells. IL10 primarily functions to limit and terminate inflammatory responses by inhibiting the synthesis of pro-inflammatory cytokines such as interferon-gamma (IFN-γ), interleukin-2 (IL2), and tumor necrosis factor-alpha (TNF-α). It also suppresses the antigen-presenting capacity of macrophages and dendritic cells, thereby reducing T cell activation. Through these mechanisms, IL10 plays a crucial role in maintaining immune homeostasis and preventing tissue damage during immune responses. | IL10 has been measured as a biomarker in various clinical and research settings to assess immune status and inflammatory activity. Elevated levels of IL10 in biological fluids, such as serum or plasma, have been observed in conditions characterized by immune activation or dysregulation, including infections, autoimmune diseases, and certain cancers. Its quantification has been utilized to monitor disease progression, evaluate the severity of inflammatory responses, and assess responses to therapeutic interventions. Additionally, IL10 levels are often included in cytokine profiling panels to provide insight into the balance between pro- and anti-inflammatory processes. |
| interleukin 2 (IL2) | Interleukin 2 (IL2) is a cytokine produced primarily by activated CD4+ T lymphocytes. It plays a central role in the regulation and proliferation of T cells, B cells, natural killer (NK) cells, and monocytes. IL2 is essential for the growth, differentiation, and survival of antigen-selected cytotoxic T cells and regulatory T cells (Tregs), thus contributing to the maintenance of immune tolerance and the modulation of immune responses. IL2 signaling occurs through the IL2 receptor complex, which activates downstream pathways such as the JAK-STAT pathway, influencing cell cycle progression and immune cell function. | IL2 has been utilized as a biomarker to assess immune activation and function in various clinical contexts. Measurement of IL2 levels in serum, plasma, or cell culture supernatants is used to evaluate T cell activation status, monitor immune responses during immunotherapy, and investigate immune dysregulation in conditions such as autoimmune diseases, infections, and cancer. Changes in IL2 expression or secretion can provide information about the efficacy of immunomodulatory treatments and the immunological status of patients. |
| interleukin 4 (IL4) | Interleukin 4 (IL4) is a cytokine produced primarily by activated T helper 2 (Th2) cells, as well as by mast cells, basophils, and eosinophils. IL4 plays a central role in the regulation of immune responses by promoting the differentiation of naive CD4+ T cells into Th2 cells. It stimulates B cell proliferation and differentiation, enhances immunoglobulin class switching to IgE and IgG1, and upregulates the expression of major histocompatibility complex (MHC) class II molecules. IL4 also inhibits the production of pro-inflammatory cytokines by macrophages and suppresses Th1-mediated immune responses, thereby contributing to the balance between humoral and cell-mediated immunity. | IL4 has been studied as a biomarker in various immunological and inflammatory conditions. Elevated levels of IL4 in serum, plasma, or tissue samples have been associated with allergic diseases such as asthma, atopic dermatitis, and allergic rhinitis, reflecting its role in promoting IgE-mediated immune responses. IL4 measurements have also been used in research to monitor immune status in autoimmune diseases, infections, and certain cancers, particularly those with a Th2-skewed immune environment. Its quantification can provide insights into the underlying immunopathology and disease activity in these contexts. |
| interleukin 6 (IL6) | Interleukin 6 (IL6) is a multifunctional cytokine produced by a variety of cell types, including T cells, B cells, macrophages, fibroblasts, endothelial cells, and others in response to infections, tissue injury, and other inflammatory stimuli. IL6 plays a central role in the regulation of immune responses, acute phase reactions, hematopoiesis, and inflammation. It signals primarily through the IL6 receptor complex, leading to the activation of intracellular pathways such as JAK/STAT, MAPK, and PI3K/Akt. IL6 promotes the differentiation of B cells into antibody-producing plasma cells, influences T cell differentiation, and stimulates the production of acute-phase proteins by hepatocytes. Additionally, IL6 is involved in the regulation of metabolic, regenerative, and neural processes. | IL6 is widely used as a biomarker in clinical and research settings to assess the presence and severity of inflammation. Elevated IL6 levels in blood, serum, or plasma have been associated with various inflammatory conditions, including infections, autoimmune diseases, and chronic inflammatory disorders. IL6 concentrations are also measured to monitor disease progression and response to therapy in conditions such as sepsis, rheumatoid arthritis, and certain cancers. Its role as an indicator of systemic inflammation makes it useful in evaluating acute phase responses and in the context of cytokine release syndromes. |
| nitric oxide synthase 2 (NOS2) | Nitric oxide synthase 2 (NOS2), also known as inducible nitric oxide synthase (iNOS), is an enzyme responsible for the high-output production of nitric oxide (NO) from L-arginine in response to pro-inflammatory stimuli. NOS2 is primarily expressed in activated macrophages and other immune cells following exposure to cytokines such as interferon-gamma and tumor necrosis factor-alpha, as well as bacterial products like lipopolysaccharide. The nitric oxide produced by NOS2 serves as a key effector molecule in host defense, contributing to the antimicrobial and cytotoxic activity of immune cells. Additionally, NOS2-derived NO plays roles in modulating vascular tone, inflammation, and cellular signaling pathways. | NOS2 expression and activity have been investigated as biomarkers of inflammation and immune activation in various pathological conditions. Elevated NOS2 levels have been observed in diseases characterized by chronic inflammation, such as sepsis, autoimmune disorders, and certain cancers. Measurement of NOS2 mRNA, protein, or enzymatic activity in tissues or bodily fluids has been utilized in research settings to assess the extent of inflammatory responses or to monitor disease progression. NOS2 has also been studied as a potential indicator of macrophage activation status in infectious and inflammatory diseases. |
| tumor necrosis factor (TNF) | Tumor necrosis factor (TNF) is a pro-inflammatory cytokine primarily produced by activated macrophages, as well as by other cell types such as lymphocytes, natural killer cells, and endothelial cells. TNF plays a central role in the regulation of immune responses, inflammation, and apoptosis. It is involved in the activation of nuclear factor-kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathways, leading to the expression of genes that mediate inflammation, cell proliferation, differentiation, and survival. TNF also contributes to the recruitment and activation of immune cells at sites of infection or injury and is implicated in the pathogenesis of various inflammatory and autoimmune diseases. | TNF is measured in biological fluids such as serum, plasma, or synovial fluid to assess the presence and degree of inflammation. Elevated TNF levels have been observed in conditions such as rheumatoid arthritis, inflammatory bowel disease, sepsis, and certain cancers. Its quantification is used in research and clinical studies to monitor disease activity, response to therapy, and to investigate the underlying mechanisms of inflammatory and immune-mediated disorders. |
Explore Research Opportunities with Protheragen. Our biomarker research services offer comprehensive analytical capabilities for the discovery and characterization of molecular targets relevant to immunosuppression. We emphasize the exploratory and research-focused nature of our work, supporting drug discovery and preclinical development. Please note that all biomarkers discussed are research targets only; we do not claim any as validated or mandatory for any application. Our services are dedicated to preclinical research stages, and we maintain scientific objectivity throughout all collaborations.
We invite you to connect with Protheragen to discuss collaborative opportunities in biomarker research for immunosuppression. Our focus is on scientific exploration and knowledge exchange, supporting preclinical research and discovery. Let’s advance the understanding of immunosuppression together through objective and innovative research.
Make Order
Experimental Scheme
Implementation
Conclusion