Biomarker Analysis Services for Scleroderma
Drug R&D Solutions

Biomarker Analysis Services for Scleroderma

Inquiry

Protheragen offers specialized biomarker analysis services dedicated to advancing Scleroderma research and therapeutic development. Our comprehensive biomarker panel is designed to provide deep insights into the molecular and cellular mechanisms underlying Scleroderma, supporting the discovery and characterization of novel drug targets. All services are exclusively focused on drug discovery through preclinical development stages and do not include clinical diagnostic services.

Biomarker Discovery and Identification

Effective therapeutic intervention begins with the precise identification of disease-relevant biomarkers. At Protheragen, our biomarker discovery services are integral to the early stages of drug development, enabling the identification of molecular signatures associated with Scleroderma pathophysiology. We employ a systematic approach encompassing high-throughput screening, candidate selection, and rigorous validation to ensure the reliability and relevance of identified biomarkers. Our process includes the integration of experimental data and bioinformatics analyses to prioritize candidates with the greatest potential for therapeutic targeting.

Multi Omics: Our multi-omics approach leverages cutting-edge technologies across genomics, transcriptomics, proteomics, and metabolomics to provide a comprehensive understanding of biological systems in Scleroderma. By integrating data from DNA, RNA, protein, and metabolite analyses, we facilitate the identification of biomarkers at multiple molecular levels. This enables the elucidation of complex disease pathways, including immune activation, fibrosis, and extracellular matrix remodeling processes that are central to Scleroderma. Our approach supports the discovery of both established and novel biomarker candidates relevant to disease mechanisms.

Candidate Validation: We employ robust validation strategies to confirm the association of candidate biomarkers with Scleroderma pathophysiology. Our preliminary screening processes include quantitative and qualitative assessments across relevant biological samples and models. Criteria for prioritizing promising candidates include specificity to disease processes, reproducibility across independent datasets, and alignment with known or emerging Scleroderma pathways. This rigorous approach ensures that only the most relevant biomarker candidates advance to further stages of assay development.

Biomarker Assay Development and Validation

Diverse Technological Platforms: Protheragen provides custom assay development capabilities, adapting technological platforms to meet specific research requirements for Scleroderma. Our portfolio includes immunoassays, mass spectrometry, flow cytometry, molecular diagnostics, and advanced histopathology and imaging solutions. Each platform is selected and optimized based on the nature of the biomarker and the intended research application, ensuring precise and reliable measurement.

Immunoassays: We offer enzyme-linked immunosorbent assay (ELISA), chemiluminescent immunoassays, and multiplex immunoassay formats for sensitive and specific quantification of protein biomarkers.

Mass Spectrometry: Our LC-MS/MS platforms enable high-resolution, quantitative analysis of proteins, peptides, and metabolites relevant to Scleroderma research.

Flow Cytometry: We utilize flow cytometry for detailed phenotypic and functional characterization of immune cell populations and surface biomarker expression.

Molecular Diagnostics: Our molecular diagnostics capabilities include quantitative PCR, digital PCR, and nucleic acid amplification for the detection of gene expression and genetic variants.

Histopathology And Imaging: We provide advanced tissue-based analyses, including immunohistochemistry and imaging, to assess biomarker localization and distribution in disease-relevant tissues.

Rigorous Method Validation: All analytical methods undergo rigorous validation in accordance with established research guidelines. Validation parameters include sensitivity, specificity, accuracy, precision, linearity, and reproducibility. Comprehensive quality control measures are implemented throughout the process to ensure data integrity and reliability, supporting robust biomarker quantification for preclinical research applications.

Biomarker Quantitative and Qualitative Analysis

Our platforms support quantitative analysis of biomarker expression and activity across diverse biological matrices. We utilize standardized protocols and calibration strategies to ensure accurate and reproducible measurements, enabling comparative studies and longitudinal monitoring in Scleroderma research models.

Sample Analysis: We handle a broad spectrum of sample types, including serum, plasma, tissue lysates, and cell preparations relevant to Scleroderma research. Our analysis protocols are optimized for each matrix, incorporating stringent quality control checkpoints to maintain sample integrity and analytical accuracy throughout the workflow.

High Throughput Capabilities: Protheragen's high-throughput analytical platforms support multiplexed biomarker analysis, enabling efficient evaluation of multiple targets from minimal sample volumes. This approach conserves valuable research material and accelerates data acquisition, facilitating comprehensive studies of biomarker panels in Scleroderma.

Key Biomarkers for Scleroderma Drug Development

Gene Target Biological Function Application as a Biomarker
CD80 molecule (CD80) CD80, also known as B7-1, is a transmembrane protein expressed primarily on antigen-presenting cells such as dendritic cells, B cells, and macrophages. Its primary biological function is to provide costimulatory signals necessary for T cell activation and survival. CD80 binds to CD28 and CTLA-4 (CD152) receptors on T cells. Interaction with CD28 delivers a positive costimulatory signal that promotes T cell proliferation, cytokine production, and survival. In contrast, binding to CTLA-4 transmits an inhibitory signal that downregulates immune responses. Through these interactions, CD80 plays a critical role in regulating adaptive immune responses and maintaining immune homeostasis. CD80 expression has been utilized as a biomarker for assessing the activation status of antigen-presenting cells and the immune microenvironment in various settings. In oncology, increased CD80 expression can be indicative of immune activation within tumors or lymphoid tissues. In transplantation, CD80 levels on donor or recipient cells have been studied in relation to graft rejection and tolerance. Additionally, CD80 is measured in autoimmune and infectious diseases to evaluate immune cell activation and disease progression.
CD86 molecule (CD86) CD86 (Cluster of Differentiation 86) is a type I transmembrane glycoprotein expressed primarily on antigen-presenting cells, including dendritic cells, macrophages, and B cells. It functions as a costimulatory molecule that interacts with CD28 and CTLA-4 receptors on T cells. The binding of CD86 to CD28 provides a necessary second signal for T cell activation and proliferation, while interaction with CTLA-4 delivers inhibitory signals to modulate immune responses. CD86 plays a critical role in the regulation of adaptive immunity, particularly in the initiation and modulation of T cell-mediated immune responses. CD86 expression is utilized as a biomarker to assess the activation status and functional state of antigen-presenting cells in various immunological contexts. It is commonly measured in studies of immune activation, inflammation, and immune-related diseases, including autoimmune disorders and cancers. In transplantation immunology, CD86 levels can be monitored to evaluate immune responses and potential rejection events. Additionally, CD86 is used in research to characterize dendritic cell maturation and to distinguish between different immune cell subsets.
cellular communication network factor 2 (CCN2) Cellular communication network factor 2 (CCN2), also known as connective tissue growth factor (CTGF), is a matricellular protein that plays a key role in various physiological processes. CCN2 is involved in cell adhesion, migration, proliferation, differentiation, and extracellular matrix production. It is particularly important in tissue repair, fibrosis, and development, where it mediates signaling between cells and the extracellular environment. CCN2 interacts with multiple growth factors, integrins, and proteoglycans, modulating pathways such as TGF-β and Wnt signaling. Its expression is tightly regulated and is induced in response to tissue injury, mechanical stress, and certain cytokines. CCN2 has been studied as a biomarker in several pathological conditions, most notably in fibrotic diseases, such as liver fibrosis, kidney fibrosis, and systemic sclerosis. Elevated levels of CCN2 have been observed in the serum, plasma, or tissue samples of affected individuals, and its expression correlates with the extent of fibrosis in some studies. Additionally, CCN2 has been investigated in the context of cancer, where its expression may be associated with tumor progression and stromal remodeling. Measurement of CCN2 levels is utilized in research settings to assess disease presence, progression, or response to therapy.
fibroblast growth factor 2 (FGF2) Fibroblast growth factor 2 (FGF2), also known as basic FGF, is a member of the fibroblast growth factor family. FGF2 is a multifunctional growth factor involved in a wide range of biological processes, including cell proliferation, differentiation, survival, and migration. It is particularly important in embryonic development, angiogenesis (formation of new blood vessels), wound healing, and tissue regeneration. FGF2 exerts its effects by binding to specific fibroblast growth factor receptors (FGFRs) on the cell surface, activating downstream signaling pathways such as MAPK, PI3K/AKT, and PLCγ. These pathways regulate gene expression and cellular responses essential for development and tissue maintenance. FGF2 has been studied as a biomarker in various clinical contexts. Elevated levels of FGF2 in tissue, serum, or plasma have been associated with certain malignancies, including glioblastoma, breast cancer, and prostate cancer, where it may reflect increased angiogenic activity or tumor progression. Additionally, FGF2 expression has been investigated in the context of cardiovascular diseases, such as atherosclerosis and myocardial infarction, due to its role in vascular remodeling and repair. Measurement of FGF2 levels can aid in disease characterization, prognosis, or monitoring of therapeutic response in these settings.
glycogen synthase kinase 3 beta (GSK3B) Glycogen synthase kinase 3 beta (GSK3B) is a serine/threonine protein kinase that plays a key role in numerous cellular processes. GSK3B is involved in the regulation of glycogen metabolism by phosphorylating and inhibiting glycogen synthase, the enzyme responsible for converting glucose to glycogen. Beyond its role in metabolism, GSK3B participates in various signaling pathways, including Wnt/β-catenin, insulin, and PI3K/Akt pathways. It regulates cellular functions such as proliferation, differentiation, apoptosis, and gene expression. GSK3B activity is tightly controlled by upstream kinases and is inhibited by phosphorylation at Ser9, which is mediated by insulin and growth factors. Dysregulation of GSK3B has been implicated in the pathogenesis of several diseases, including neurodegenerative disorders, diabetes, and cancer. GSK3B has been investigated as a biomarker in multiple contexts due to its involvement in diverse signaling pathways and disease processes. Altered expression or activity of GSK3B has been reported in neurodegenerative diseases such as Alzheimer's disease, where increased GSK3B activity is associated with tau hyperphosphorylation and neurofibrillary tangle formation. In oncology, aberrant GSK3B signaling has been observed in various cancers, and its expression or phosphorylation status has been studied in relation to tumor progression and prognosis. Additionally, GSK3B has been examined as a potential pharmacodynamic biomarker for monitoring the effects of therapeutic agents targeting relevant signaling pathways. Its utility as a biomarker depends on the specific disease context and the methods used for its detection and quantification.
histamine receptor H4 (HRH4) Histamine receptor H4 (HRH4) is a member of the G protein-coupled receptor family that binds histamine, a biogenic amine involved in immune regulation and inflammatory responses. HRH4 is primarily expressed in hematopoietic cells, including eosinophils, mast cells, basophils, dendritic cells, and T cells. Upon activation by histamine, HRH4 mediates chemotaxis, calcium mobilization, and cytokine production, contributing to the modulation of immune cell migration and activation. HRH4 is implicated in the regulation of inflammatory and allergic responses, as well as in the pathophysiology of various immune-mediated diseases. Expression levels and genetic variants of HRH4 have been investigated in relation to several inflammatory and immune-related conditions, such as allergic diseases, asthma, inflammatory bowel disease, and certain cancers. HRH4 has been studied as a potential biomarker for disease presence, progression, or response to therapy in these contexts. Its expression patterns in tissues or peripheral blood, as well as polymorphisms in the HRH4 gene, are assessed for their association with disease susceptibility or clinical outcomes.
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 cell types such as γδ T cells and innate lymphoid cells. IL17A plays a central role in the regulation of immune responses, particularly in the defense against extracellular bacteria and fungi. It exerts its effects by binding to the IL-17 receptor complex on various cell types, inducing the production of chemokines, cytokines, and antimicrobial peptides. These mediators promote the recruitment and activation of neutrophils and other immune cells to sites of infection or inflammation. IL17A is also involved in the pathogenesis of several autoimmune and inflammatory diseases due to its ability to sustain chronic inflammation. IL17A has been investigated as a biomarker in a range of clinical contexts, including autoimmune and inflammatory disorders such as psoriasis, rheumatoid arthritis, ankylosing spondylitis, and inflammatory bowel disease. Elevated levels of IL17A in serum, plasma, or tissue samples have been associated with disease activity, severity, or progression in these conditions. Measurement of IL17A may therefore provide information relevant to disease characterization, monitoring of inflammatory status, and assessment of therapeutic response in research and clinical settings.
transglutaminase 2 (TGM2) Transglutaminase 2 (TGM2) is an enzyme belonging to the transglutaminase family, which catalyzes the formation of covalent cross-links between proteins through transamidation of glutamine and lysine residues. TGM2 exhibits multifunctional properties, including protein cross-linking, GTPase activity, protein disulfide isomerase activity, and involvement in cell signaling. It plays roles in a variety of physiological processes such as apoptosis, extracellular matrix stabilization, wound healing, and cell adhesion. TGM2 is widely expressed in many tissues and is localized both intracellularly and extracellularly. TGM2 expression and activity have been studied as potential biomarkers in several pathological conditions. Increased levels of TGM2 have been observed in various cancers, including pancreatic, colorectal, and breast cancer, where it has been associated with tumor progression, metastasis, and drug resistance. Elevated TGM2 is also seen in certain neurodegenerative diseases and autoimmune disorders such as celiac disease, where autoantibodies against TGM2 are used in serological testing. Its altered expression in disease states supports its utility in disease detection, prognosis, and monitoring.
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 exerts its biological effects by binding to two distinct receptors, TNFR1 and TNFR2, leading to activation of downstream signaling pathways such as NF-κB and MAPK. These pathways mediate processes including cell survival, differentiation, and programmed cell death. TNF is involved in host defense mechanisms against infections, but dysregulated TNF production is implicated in the pathogenesis of several inflammatory and autoimmune diseases. TNF is measured in biological fluids such as serum, plasma, and synovial fluid to assess inflammatory activity in various clinical contexts. Elevated TNF levels have been associated with chronic inflammatory diseases such as rheumatoid arthritis, inflammatory bowel disease, and psoriasis. In addition, TNF concentrations may be monitored in infectious diseases and sepsis to evaluate the extent of systemic inflammation. Measurement of TNF can aid in the characterization of disease activity and may be used in conjunction with other clinical and laboratory findings.
tyrosine kinase 2 (TYK2) Tyrosine kinase 2 (TYK2) is a member of the Janus kinase (JAK) family of non-receptor tyrosine kinases. TYK2 plays a critical role in mediating signal transduction for a variety of cytokine receptors, including those for type I interferons (IFN-α/β), interleukin-12 (IL-12), and interleukin-23 (IL-23). Upon cytokine binding to their respective receptors, TYK2 becomes activated through phosphorylation and subsequently initiates downstream signaling cascades, particularly the JAK-STAT (signal transducer and activator of transcription) pathway. This leads to the transcription of genes involved in immune regulation, inflammation, and host defense. TYK2 is thus essential for the development and function of both innate and adaptive immune responses. TYK2 expression and genetic variants have been investigated as biomarkers in various disease contexts, particularly autoimmune and inflammatory disorders. Altered TYK2 activity or mutations have been associated with susceptibility to conditions such as systemic lupus erythematosus, multiple sclerosis, psoriasis, and certain primary immunodeficiencies. In addition, TYK2 has been studied in the context of cancer and infectious diseases. Its expression levels, functional status, or the presence of specific polymorphisms may provide information relevant to disease risk, prognosis, or therapeutic response, especially in settings where JAK-STAT signaling is implicated.

Partner with Protheragen to Advance Innovation

Explore Research Opportunities with Protheragen. Our biomarker research services for Scleroderma provide a robust platform for exploratory studies and preclinical drug discovery. We offer comprehensive capabilities in biomarker identification, validation, and assay development, leveraging advanced analytical technologies. Please note that all biomarkers discussed are research targets only; we do not claim any biomarkers as validated or mandatory for Scleroderma research or therapy. Our services are strictly intended for preclinical research and maintain scientific objectivity throughout the process.

We invite you to connect with Protheragen to discuss exploratory biomarker research for Scleroderma. Our team is committed to scientific collaboration and the exchange of knowledge, supporting your research objectives with technical expertise and a focus on preclinical discovery. Let’s advance the understanding of Scleroderma together through objective, data-driven research.

HOW WE WORK

Make Order

Make Order

Experimental Scheme

Experimental Scheme

Implementation

Implementation

Conclusion

Conclusion
Make an Inquiry