Protheragen offers specialized biomarker analysis services dedicated to advancing uveitis research and therapeutic development. Our comprehensive biomarker panel is designed to elucidate the complex pathophysiology of uveitis, supporting drug discovery and preclinical development efforts. Please note that all our services are exclusively focused on research and preclinical drug development; we do not provide clinical diagnostic services.
Effective therapeutic intervention in uveitis begins with the precise discovery and identification of relevant biomarkers. At Protheragen, our biomarker discovery services form the cornerstone of preclinical drug development by enabling the identification of molecular indicators associated with disease mechanisms. We employ systematic screening and validation processes, including literature mining, high-throughput screening, and targeted validation, to pinpoint and confirm candidate biomarkers pertinent to uveitis pathology.
Multi Omics: Protheragen utilizes a multi-omics approach, integrating cutting-edge technologies such as genomics, transcriptomics, and proteomics to provide a comprehensive analysis of biological systems in uveitis. Our platforms enable the identification of DNA, RNA, protein, and metabolite biomarkers, offering a holistic view of the molecular networks underlying disease processes. This approach facilitates the investigation of key pathways involved in immune activation, inflammation, and tissue remodeling relevant to uveitis.
Candidate Validation: Our candidate validation strategies encompass rigorous experimental and computational methods to assess the association of biomarkers with uveitis pathophysiology. Preliminary screening involves quantitative and qualitative analyses to determine expression patterns, functional relevance, and disease linkage. Promising candidates are prioritized based on criteria such as specificity, sensitivity, biological plausibility, and translational potential for therapeutic development.
Diverse Technological Platforms: Protheragen provides custom assay development capabilities tailored to the specific requirements of uveitis research. Our technological platforms are adaptable and include immunoassays, mass spectrometry, flow cytometry, molecular diagnostics, and advanced histopathology and imaging systems. These platforms enable robust and reproducible measurement of biomarker candidates across diverse sample types.
Immunoassays: We offer enzyme-linked immunosorbent assays (ELISA), chemiluminescent immunoassays, and multiplex immunoassay platforms for sensitive and specific quantification of cytokines, chemokines, and other protein biomarkers.
Mass Spectrometry: Our LC-MS/MS workflows enable accurate, high-throughput quantification and characterization of proteins, peptides, and metabolites relevant to uveitis.
Flow Cytometry: We utilize multiparametric flow cytometry for detailed immunophenotyping and quantification of cell surface and intracellular biomarkers, supporting immune cell profiling in uveitis models.
Molecular Diagnostics: Our molecular assays, including qPCR and digital PCR, allow for precise detection and quantification of nucleic acid biomarkers at the DNA and RNA levels.
Histopathology And Imaging: We perform histopathological analyses and advanced imaging (e.g., immunohistochemistry, immunofluorescence) to localize and visualize biomarker expression in ocular tissues.
Rigorous Method Validation: All assay development and validation processes at Protheragen adhere to established guidelines to ensure data reliability and reproducibility. We evaluate critical performance characteristics such as specificity, sensitivity, linearity, accuracy, and precision. Stringent quality control measures are implemented throughout the workflow to maintain high standards and ensure the robustness of biomarker quantification.
Our quantitative analysis capabilities encompass sensitive and reproducible measurement of biomarker levels across multiple biological matrices. We leverage advanced analytical platforms to generate high-quality, quantitative data for biomarker discovery, validation, and preclinical research applications in uveitis.
Sample Analysis: Protheragen processes a wide range of sample types, including serum, plasma, ocular fluids, tissue lysates, and cell culture supernatants. Our standardized analysis protocols incorporate rigorous quality assurance steps, including sample tracking, internal controls, and replicates, to ensure the integrity and reliability of results.
High Throughput Capabilities: Our high-throughput analytical platforms, including multiplex immunoassays and automated mass spectrometry systems, enable efficient processing of large sample cohorts. These technologies maximize analytical efficiency, conserve valuable samples, and facilitate comprehensive biomarker profiling for uveitis research.
| Gene Target | Biological Function | Application as a Biomarker |
|---|---|---|
| interferon gamma (IFNG) | Interferon gamma (IFNG) is a cytokine primarily produced by activated T lymphocytes, particularly Th1 CD4+ T cells, CD8+ cytotoxic T cells, and natural killer (NK) cells. It plays a central role in innate and adaptive immunity by activating macrophages, enhancing antigen presentation through upregulation of major histocompatibility complex (MHC) molecules, and promoting the differentiation of naive T cells toward the Th1 phenotype. IFNG also has antiviral, immunoregulatory, and anti-proliferative effects, and is critical in orchestrating immune responses against intracellular pathogens such as viruses and certain bacteria. | IFNG is used as a biomarker to assess cellular immune responses, particularly in the context of infectious diseases, autoimmune disorders, and immunotherapy monitoring. Its measurement is employed in assays such as the interferon-gamma release assay (IGRA) for detecting latent Mycobacterium tuberculosis infection. Elevated or diminished IFNG levels in blood, tissue, or culture supernatants can indicate immune activation status, help in disease diagnosis, and provide information about immune system function in various clinical contexts. |
| interleukin 1 beta (IL1B) | Interleukin 1 beta (IL1B) is a pro-inflammatory cytokine produced primarily by activated macrophages, monocytes, and dendritic cells. It plays a central role in the regulation of immune and inflammatory responses. IL1B is synthesized as an inactive precursor (pro-IL1B) and is cleaved by caspase-1 to generate the active cytokine. Upon secretion, IL1B binds to the interleukin-1 receptor (IL-1R), initiating signaling cascades that lead to the expression of various inflammatory mediators, including prostaglandins, nitric oxide, and other cytokines. IL1B is involved in processes such as fever induction, leukocyte recruitment, and the activation of lymphocytes, and it contributes to the pathogenesis of a range of inflammatory and autoimmune conditions. | IL1B is measured in biological fluids such as serum, plasma, and synovial fluid as an indicator of inflammation. Elevated levels of IL1B have been associated with various inflammatory diseases, including rheumatoid arthritis, sepsis, and certain autoinflammatory syndromes. Its quantification can assist in assessing the degree of inflammation, monitoring disease activity, and evaluating response to anti-inflammatory therapies in clinical and research settings. |
| interleukin 17A (IL17A) | Interleukin 17A (IL17A) is a pro-inflammatory cytokine produced primarily by T helper 17 (Th17) cells, as well as other immune cell types such as γδ T cells, natural killer cells, and innate lymphoid cells. IL17A plays a key role in host defense against extracellular bacteria and fungi by promoting the recruitment and activation of neutrophils. It induces the expression of various chemokines, cytokines, and antimicrobial peptides in target cells, including epithelial and endothelial cells. IL17A is also involved in the regulation of tissue inflammation and has been implicated in the pathogenesis of several autoimmune and inflammatory diseases due to its ability to amplify inflammatory responses. | IL17A has been used as a biomarker to assess the presence and intensity of inflammatory and autoimmune responses. Elevated levels of IL17A have been observed in the blood, synovial fluid, and tissues of individuals with conditions such as psoriasis, rheumatoid arthritis, ankylosing spondylitis, and multiple sclerosis. Measurement of IL17A concentrations can provide information about immune activation status and disease activity in these contexts. |
| interleukin 2 receptor subunit alpha (IL2RA) | Interleukin 2 receptor subunit alpha (IL2RA), also known as CD25, is a component of the high-affinity receptor for interleukin-2 (IL-2). IL2RA is primarily expressed on activated T lymphocytes, regulatory T cells (Tregs), and some B cells. Upon binding to IL-2, IL2RA forms a receptor complex with IL2RB (CD122) and IL2RG (CD132), which initiates intracellular signaling pathways crucial for T cell proliferation, differentiation, and survival. IL2RA plays a pivotal role in the regulation of immune responses, particularly in the development and maintenance of immune tolerance through its expression on Tregs. | IL2RA is used as a biomarker to assess immune activation and regulatory T cell populations. Elevated levels of soluble IL2RA (sCD25) in serum or plasma have been associated with various conditions characterized by immune activation, such as autoimmune diseases (e.g., type 1 diabetes, multiple sclerosis), certain lymphoid malignancies (e.g., adult T-cell leukemia/lymphoma), and graft-versus-host disease. Measurement of IL2RA expression on cell surfaces is also utilized to identify and quantify regulatory T cells in immunological studies. |
| interleukin 6 (IL6) | Interleukin 6 (IL6) is a multifunctional cytokine that plays a central role in immune response, inflammation, and hematopoiesis. It is produced by a variety of cell types, including T cells, B cells, macrophages, fibroblasts, and endothelial cells, in response to infections, tissue injuries, and other inflammatory stimuli. IL6 mediates its effects through binding to the IL6 receptor (IL6R) and the signal-transducing component gp130, activating downstream signaling pathways such as JAK/STAT, MAPK, and PI3K/Akt. Biologically, IL6 promotes the differentiation of B cells into plasma cells, stimulates the production of acute-phase proteins by hepatocytes, modulates T cell responses, and influences the balance between pro-inflammatory and anti-inflammatory processes. It is also involved in the regulation of metabolic, regenerative, and neural processes. | IL6 is widely measured as a biomarker of inflammation and immune activation in various clinical and research settings. Elevated IL6 levels in blood or other biological fluids are associated with a range of inflammatory, infectious, and autoimmune conditions, including sepsis, rheumatoid arthritis, and COVID-19. It is also monitored in certain cancers and chronic diseases to assess disease activity, progression, and response to therapy. IL6 concentrations can provide information on the severity of inflammatory responses and are used alongside other markers to support clinical decision-making. |
| stimulator of interferon response cGAMP interactor 1 (STING1) | Stimulator of interferon response cGAMP interactor 1 (STING1), also known as STING or TMEM173, is an endoplasmic reticulum-associated transmembrane protein that plays a central role in the innate immune response to cytosolic DNA. Upon detection of cytosolic double-stranded DNA by cyclic GMP-AMP synthase (cGAS), the second messenger cyclic GMP-AMP (cGAMP) is produced and binds to STING1. This binding triggers a conformational change in STING1, leading to its translocation from the endoplasmic reticulum to the Golgi apparatus. STING1 subsequently activates TANK-binding kinase 1 (TBK1) and interferon regulatory factor 3 (IRF3), resulting in the production of type I interferons and other pro-inflammatory cytokines. This pathway is important for host defense against viral and some bacterial pathogens, as well as for the regulation of autoimmunity and tumor immunity. | STING1 expression and activation status have been investigated as biomarkers in several contexts, including cancer, infectious diseases, and autoimmune disorders. In oncology, STING1 pathway activation has been associated with tumor-infiltrating immune cell responses and may correlate with prognosis or response to immunotherapies in certain cancers. In autoimmune diseases, altered STING1 activity has been observed in conditions such as systemic lupus erythematosus and STING-associated vasculopathy with onset in infancy (SAVI). Measurement of STING1 levels, mutations, or pathway activation markers in tissues or peripheral blood has been explored for disease characterization, stratification, and monitoring. |
| toll like receptor 2 (TLR2) | Toll-like receptor 2 (TLR2) is a pattern recognition receptor that plays a critical role in the innate immune system. It is expressed on the surface of various immune cells, including macrophages, dendritic cells, and neutrophils. TLR2 recognizes a broad range of pathogen-associated molecular patterns (PAMPs) derived from bacteria, fungi, and some viruses, such as lipoproteins, peptidoglycan, and lipoarabinomannan. Upon ligand binding, TLR2 forms heterodimers with either TLR1 or TLR6, initiating intracellular signaling cascades that result in the activation of nuclear factor-kappa B (NF-κB) and the production of pro-inflammatory cytokines. These responses contribute to the early detection and clearance of pathogens. | TLR2 expression and activation have been studied as biomarkers in various contexts, including infectious diseases, inflammatory disorders, and certain cancers. Altered TLR2 levels or signaling activity in blood or tissue samples may reflect immune activation or dysregulation. For example, increased TLR2 expression has been observed in sepsis, tuberculosis, and autoimmune diseases, and may correlate with disease presence or severity. In oncology, TLR2 has been investigated for its association with tumor microenvironment and prognosis. Measurement of TLR2 can be performed using techniques such as flow cytometry, immunohistochemistry, or quantitative PCR. |
| toll like receptor 4 (TLR4) | Toll-like receptor 4 (TLR4) is a pattern recognition receptor that plays a central role in the innate immune system. It is primarily expressed on the surface of immune cells such as macrophages, dendritic cells, and neutrophils. TLR4 recognizes pathogen-associated molecular patterns (PAMPs), most notably lipopolysaccharide (LPS) from Gram-negative bacteria. Upon ligand binding, TLR4 initiates intracellular signaling cascades that activate transcription factors such as NF-κB, leading to the production of pro-inflammatory cytokines and type I interferons. This response is critical for the early detection and control of microbial infections and also contributes to the regulation of adaptive immune responses. | TLR4 expression and signaling activity have been investigated as biomarkers in various clinical contexts. Elevated TLR4 levels or activity have been associated with inflammatory and infectious diseases, including sepsis, atherosclerosis, and certain autoimmune disorders. Measurement of TLR4 expression or related signaling molecules in blood or tissue samples has been used in research studies to assess immune activation, disease severity, or prognosis. Additionally, TLR4 has been studied as a potential indicator of response to specific therapies, such as immunomodulatory agents. |
| tumor necrosis factor (TNF) | Tumor necrosis factor (TNF) is a pro-inflammatory cytokine primarily produced by activated macrophages, but also by a variety of other cell types including lymphocytes, natural killer cells, and endothelial cells. TNF plays a central role in the regulation of immune responses, inflammation, and apoptosis. It mediates its effects through binding to two receptors, TNFR1 and TNFR2, leading to the activation of signaling pathways such as NF-κB and MAPK. TNF is involved in the defense against infections, the induction of fever, the regulation of cell survival and death, and the modulation of immune cell activity. Dysregulation of TNF production or signaling has been associated with the pathogenesis of various inflammatory and autoimmune diseases. | TNF levels are measured in biological fluids, such as serum or plasma, to assess inflammatory activity in various clinical contexts. Elevated TNF concentrations have been observed in diseases characterized by chronic inflammation, including rheumatoid arthritis, inflammatory bowel disease, psoriasis, and sepsis. Measurement of TNF can aid in monitoring disease activity, evaluating response to anti-TNF therapies, and providing information about the inflammatory status of patients. Its application as a biomarker is based on its established role in mediating and reflecting inflammatory processes. |
| vascular endothelial growth factor A (VEGFA) | Vascular endothelial growth factor A (VEGFA) is a key signaling protein involved in the regulation of angiogenesis, the process by which new blood vessels form from pre-existing vessels. VEGFA primarily acts on endothelial cells, promoting their proliferation, migration, and survival. It binds to specific tyrosine kinase receptors, mainly VEGFR-1 (FLT1) and VEGFR-2 (KDR/FLK1), initiating intracellular signaling cascades that lead to increased vascular permeability and the growth of new capillaries. VEGFA plays essential roles in embryonic development, wound healing, and the female reproductive cycle. Dysregulation of VEGFA expression is associated with pathological angiogenesis in diseases such as cancer, diabetic retinopathy, and age-related macular degeneration. | VEGFA is commonly measured in biological fluids and tissues as an indicator of angiogenic activity. Elevated levels of VEGFA have been observed in various malignancies and are associated with tumor progression and metastasis. In oncology, VEGFA is used to assess tumor angiogenesis and may inform prognosis or therapeutic response, particularly in the context of anti-angiogenic therapies. Additionally, VEGFA levels are evaluated in ocular fluids for conditions characterized by abnormal blood vessel growth, such as diabetic retinopathy and age-related macular degeneration. Its measurement is also explored in cardiovascular and inflammatory diseases where altered angiogenesis is implicated. |
Explore Research Opportunities with Protheragen. Our biomarker research services for uveitis are designed to support drug discovery and preclinical development through comprehensive, exploratory analysis of immune, inflammatory, and angiogenic pathways. All biomarkers discussed are research targets only and are not claimed as validated or mandatory markers. Our work is focused exclusively on preclinical research, and we maintain scientific objectivity throughout all stages of partnership.
We invite you to engage with Protheragen for collaborative discussions on biomarker research in uveitis. Our work is exploratory and focused on advancing scientific knowledge through partnership. Let us work together to drive innovation in preclinical biomarker discovery and analysis.
Make Order
Experimental Scheme
Implementation
Conclusion