Biomarker Analysis Services for Sjogren Syndrome
Drug R&D Solutions

Biomarker Analysis Services for Sjogren Syndrome

Inquiry

Protheragen offers specialized biomarker analysis services tailored for Sjogren Syndrome research and therapeutic development, with a focus exclusively on drug discovery through preclinical development stages. Our comprehensive biomarker panel is designed to advance the understanding of Sjogren Syndrome pathophysiology, supporting the identification and characterization of molecular targets relevant to immune dysregulation. Please note that our services are strictly limited to research and preclinical drug development; we do not provide clinical diagnostic services.

Biomarker Discovery and Identification

Effective therapeutic intervention for Sjogren Syndrome begins with robust biomarker discovery and identification. At Protheragen, our biomarker discovery services are integral to the drug development process, enabling the detection of molecular and cellular signatures associated with disease mechanisms. Our approach encompasses systematic screening of candidate biomarkers using high-throughput technologies, followed by rigorous validation to assess their relevance and reliability. Through iterative analysis and experimental validation, we ensure the selection of high-confidence biomarkers that inform preclinical therapeutic strategies.

Multi Omics: Our multi-omics platform leverages advanced genomics, transcriptomics, proteomics, and related -omics technologies to provide a holistic view of the biological systems underlying Sjogren Syndrome. By integrating DNA, RNA, protein, and metabolite profiling, we comprehensively characterize disease-associated pathways such as immune cell activation, cytokine signaling, and B-cell dysregulation. This systems-level approach facilitates the identification of molecular biomarkers that reflect the complexity of Sjogren Syndrome pathogenesis, supporting the development of targeted therapeutics.

Candidate Validation: Candidate biomarker validation at Protheragen involves a combination of in vitro, ex vivo, and in silico strategies to establish associations with Sjogren Syndrome pathophysiology. Preliminary screening processes include quantitative and qualitative assays to assess expression patterns, functional relevance, and disease linkage. Promising candidates are prioritized based on criteria such as specificity to disease mechanisms, robustness across sample types, and potential utility in monitoring therapeutic response. Our validation workflow ensures that only the most relevant biomarker candidates advance to subsequent stages of preclinical research.

Biomarker Assay Development and Validation

Diverse Technological Platforms: Protheragen develops custom biomarker assays across a range of technological platforms, adapting to the specific requirements of each research program. Our capabilities include the design and optimization of immunoassays, mass spectrometry-based workflows, flow cytometry panels, molecular diagnostic assays, and histopathological imaging protocols. Each platform is selected and configured to maximize sensitivity, specificity, and throughput for the intended biomarker targets.

Immunoassays: We utilize ELISA, chemiluminescent, and multiplex immunoassay formats for quantifying cytokines, cell surface markers, and soluble proteins relevant to Sjogren Syndrome.

Mass Spectrometry: Our LC-MS/MS workflows enable sensitive and specific detection of protein and peptide biomarkers, supporting both targeted and discovery-based analyses.

Flow Cytometry: Multiparametric flow cytometry is employed to phenotype immune cell populations, assess activation markers, and quantify cellular expression of key targets.

Molecular Diagnostics: We implement PCR-based and other nucleic acid amplification techniques for the detection and quantification of gene expression and genetic variants.

Histopathology And Imaging: Advanced histological staining and imaging methods are used to localize biomarker expression in tissue samples and evaluate cellular architecture.

Rigorous Method Validation: All analytical methods developed at Protheragen undergo rigorous validation according to established guidelines, including assessments of accuracy, precision, sensitivity, specificity, linearity, and reproducibility. Our quality control measures encompass the use of calibrated standards, controls, and replicates to ensure data integrity and reliability throughout the biomarker analysis process.

Biomarker Quantitative and Qualitative Analysis

Protheragen provides quantitative analysis capabilities for biomarker measurement across a variety of biological matrices. Our protocols are optimized for accurate quantification of low-abundance targets, enabling robust data generation for preclinical research applications.

Sample Analysis: We handle a wide range of sample types, including blood, serum, plasma, tissue biopsies, and cell lysates. Each sample is processed using standardized protocols to preserve biomarker integrity. Quality measures such as sample tracking, contamination prevention, and consistent processing conditions are strictly enforced to maintain analytical reliability.

High Throughput Capabilities: Our high-throughput analytical platforms support multiplexed biomarker analysis, allowing simultaneous measurement of multiple targets within limited sample volumes. This approach enhances efficiency, conserves valuable research specimens, and accelerates data acquisition for large-scale preclinical studies.

Key Biomarkers for Sjogren Syndrome Drug Development

Gene Target Biological Function Application as a Biomarker
CD80 molecule (CD80) CD80 (Cluster of Differentiation 80), 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 a costimulatory signal necessary for T cell activation and survival. CD80 binds to CD28 and CTLA-4 (CD152) receptors on T cells. Interaction with CD28 promotes T cell proliferation, cytokine production, and survival, while binding to CTLA-4 transmits inhibitory signals that regulate immune responses. Through these interactions, CD80 plays a critical role in modulating adaptive immune responses and maintaining immune homeostasis. CD80 expression has been utilized as a biomarker for assessing the activation state of antigen-presenting cells and evaluating immune responses. It is used in immunophenotyping to distinguish activated dendritic cells and B cells in various research and clinical settings. CD80 expression levels have also been investigated in the context of autoimmune diseases, transplant rejection, and certain malignancies, where altered expression may reflect changes in immune activation or regulation.
TNF receptor superfamily member 13C (TNFRSF13C) TNF receptor superfamily member 13C (TNFRSF13C), also known as B-cell-activating factor receptor (BAFF-R), is a transmembrane protein primarily expressed on B lymphocytes. It serves as the principal receptor for the cytokine BAFF (B-cell activating factor), which is critical for B cell survival, maturation, and homeostasis. Upon binding BAFF, TNFRSF13C transduces signals that promote B cell proliferation, differentiation, and resistance to apoptosis. This receptor is essential for the maintenance of peripheral B cell populations and the development of antibody responses. TNFRSF13C expression and genetic variants have been investigated as biomarkers in various immunological and hematological conditions. Its expression levels on B cells have been studied in the context of autoimmune diseases, such as systemic lupus erythematosus and rheumatoid arthritis, to assess B cell activation and disease activity. Additionally, alterations in TNFRSF13C have been explored in certain B cell malignancies and immunodeficiency disorders to provide information about disease status, prognosis, or therapeutic response.
TNF receptor superfamily member 17 (TNFRSF17) TNF receptor superfamily member 17 (TNFRSF17), also known as B-cell maturation antigen (BCMA), is a cell surface receptor that is predominantly expressed on mature B lymphocytes and plasma cells. It binds to the ligands B-cell activating factor (BAFF, also known as TNFSF13B) and a proliferation-inducing ligand (APRIL, also known as TNFSF13), both of which are members of the tumor necrosis factor (TNF) ligand family. TNFRSF17 signaling promotes B-cell survival, differentiation, and long-term maintenance of plasma cells by activating pathways such as NF-κB and MAPK. This receptor plays a critical role in humoral immunity by supporting the survival of antibody-producing cells. TNFRSF17 is utilized as a biomarker for the identification and quantification of plasma cells in both normal and pathological conditions. Its expression is frequently assessed in hematological malignancies, particularly in multiple myeloma, where TNFRSF17 is highly expressed on malignant plasma cells. Measurement of TNFRSF17 expression can aid in the diagnosis, monitoring of disease burden, and assessment of minimal residual disease in plasma cell disorders.
TNF superfamily member 13b (TNFSF13B) TNF superfamily member 13b (TNFSF13B), also known as B-cell activating factor (BAFF), is a cytokine primarily involved in the regulation of B-cell development, survival, and differentiation. TNFSF13B binds to receptors such as BAFF-R (TNFRSF13C), TACI (TNFRSF13B), and BCMA (TNFRSF17) on B cells, promoting their maturation and proliferation. It plays a crucial role in maintaining peripheral B-cell homeostasis and is essential for the generation of antibody responses. Dysregulation of TNFSF13B expression has been associated with altered immune function and the development of autoimmune conditions. TNFSF13B has been studied as a biomarker in various clinical contexts, particularly in autoimmune diseases such as systemic lupus erythematosus (SLE) and rheumatoid arthritis, where elevated levels have been observed in patient serum. Its quantification may provide information on B-cell activity and immune system status. Additionally, TNFSF13B has been investigated as a potential biomarker in certain hematological malignancies, including B-cell lymphomas, where its expression may reflect disease presence or activity.
interleukin 17A (IL17A) Interleukin 17A (IL17A) is a pro-inflammatory cytokine primarily produced by a subset of T helper cells known as Th17 cells, as well as other immune cell types. IL17A plays a central role in mediating immune responses, particularly at mucosal barriers. It stimulates the production of other cytokines, chemokines, and antimicrobial peptides by various cell types, including epithelial and endothelial cells. This leads to the recruitment and activation of neutrophils and contributes to the defense against extracellular pathogens, such as bacteria and fungi. Additionally, IL17A is involved in the regulation of inflammatory processes and has been implicated in the maintenance of tissue homeostasis and the pathogenesis of several autoimmune and inflammatory diseases. IL17A is used as a biomarker to assess immune activation and inflammatory status in various clinical and research settings. Elevated levels of IL17A in biological fluids, such as serum or synovial fluid, have been associated with autoimmune and inflammatory conditions including psoriasis, rheumatoid arthritis, ankylosing spondylitis, and multiple sclerosis. Measurement of IL17A can aid in the evaluation of disease activity, monitoring of therapeutic response, and characterization of immune profiles in patients with immune-mediated disorders.
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 is expressed on the surface of activated T cells, regulatory T cells (Tregs), and certain other immune cell types. Its primary function is to bind IL-2, a cytokine critical for T cell proliferation, differentiation, and survival. By forming a high-affinity receptor complex with the beta (CD122) and gamma (CD132) subunits, IL2RA enables efficient IL-2 signaling, which is essential for the development and maintenance of immune tolerance and the regulation of immune responses. IL2RA is used as a biomarker for immune activation and regulatory T cell identification. Elevated levels of soluble IL2RA (sCD25) in serum or plasma have been observed in various conditions associated with immune activation, such as autoimmune diseases, certain hematological malignancies (e.g., adult T-cell leukemia/lymphoma), and graft-versus-host disease. Measurement of IL2RA expression on cell surfaces is also employed to identify and quantify regulatory T cells in immunological studies.
peptidylprolyl isomerase A (PPIA) Peptidylprolyl isomerase A (PPIA), also known as cyclophilin A, is an enzyme that catalyzes the cis-trans isomerization of proline residues in polypeptide chains. This activity facilitates proper protein folding and conformational changes, which are essential for protein function and stability. PPIA is ubiquitously expressed in human tissues and is involved in several cellular processes, including intracellular signaling, protein trafficking, and immune modulation. It also interacts with cyclosporin A, forming a complex that inhibits calcineurin and thereby suppresses T-cell activation. PPIA has been used as a reference or housekeeping gene in gene expression studies due to its relatively stable expression in many cell types. Additionally, elevated levels of PPIA have been observed in certain pathological conditions, such as inflammatory diseases, cardiovascular disorders, and some cancers. Its expression patterns have been investigated as a potential indicator of disease presence or progression, and PPIA levels in biological fluids have been explored in research as a non-invasive marker for tissue injury or disease activity.
signal transducer and activator of transcription 1 (STAT1) Signal transducer and activator of transcription 1 (STAT1) is a member of the STAT protein family. STAT1 functions as a transcription factor that is activated primarily in response to cytokines and growth factors, most notably interferons (IFNs). Upon stimulation, STAT1 is phosphorylated by receptor-associated kinases, dimerizes, and translocates to the nucleus, where it binds to specific DNA sequences to regulate the expression of target genes. STAT1 plays a critical role in mediating cellular responses to type I and type II interferons, contributing to antiviral defense, immune modulation, cell growth inhibition, and apoptosis. Its activity is essential for host defense mechanisms and the regulation of immune responses. STAT1 expression and activation status have been investigated as biomarkers in various contexts, including infectious diseases, autoimmune disorders, and cancer. In clinical and research settings, STAT1 levels or phosphorylation state may be measured to assess interferon signaling pathway activity, immune system activation, or cellular responses to therapy. Altered STAT1 expression or activation has been associated with disease states such as chronic viral infections, interferonopathies, and certain malignancies, where it may provide information on disease mechanisms, prognosis, or therapeutic response.
toll like receptor 9 (TLR9) Toll-like receptor 9 (TLR9) is a member of the Toll-like receptor family, which plays a critical role in the innate immune system. TLR9 is primarily expressed in plasmacytoid dendritic cells, B cells, and certain other immune cell types. It is localized to endosomal compartments within these cells. TLR9 recognizes unmethylated CpG motifs commonly found in bacterial and viral DNA, but rare in vertebrate genomes. Upon binding to CpG DNA, TLR9 initiates a signaling cascade that activates transcription factors such as NF-κB and IRF7, leading to the production of type I interferons and pro-inflammatory cytokines. This response contributes to the detection and clearance of pathogens, bridging innate and adaptive immunity. TLR9 expression and activity have been investigated as biomarkers in various clinical contexts. In infectious diseases, altered TLR9 levels may reflect immune activation in response to microbial DNA. In oncology, TLR9 expression in tumor cells or the tumor microenvironment has been studied in relation to tumor progression and immune infiltration. Additionally, in autoimmune diseases such as systemic lupus erythematosus, aberrant TLR9 signaling is associated with disease activity. Measurement of TLR9 expression or activation status in tissues or peripheral blood can provide information on immune system status and disease processes.
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 exerts its effects by binding to two receptors, TNFR1 and TNFR2, which activate multiple intracellular signaling pathways, including NF-κB and MAPK pathways. These pathways mediate the expression of genes involved in inflammation, cell proliferation, differentiation, and cell death. TNF is involved in host defense against infections, the pathogenesis of autoimmune diseases, and the regulation of tissue homeostasis. TNF levels have been measured in biological fluids such as serum, plasma, and synovial fluid as an indicator of inflammatory activity. Elevated TNF concentrations have been observed in various conditions, including rheumatoid arthritis, inflammatory bowel disease, sepsis, and certain cancers. As such, TNF has been used to assess disease activity, monitor response to anti-TNF therapies, and investigate the inflammatory status in clinical and research settings.

Partner with Protheragen to Advance Innovation

Explore Research Opportunities with Protheragen. Our biomarker research services and capabilities are designed for exploratory and preclinical research into the molecular underpinnings of Sjogren Syndrome. The biomarkers discussed are research targets only and are not presented as validated or mandatory for therapeutic development. We focus exclusively on preclinical research stages, maintaining scientific objectivity and a commitment to rigorous, hypothesis-driven investigation.

We invite you to discuss collaborative opportunities in biomarker research for Sjogren Syndrome. Our approach emphasizes the exploratory nature of biomarker discovery, supporting scientific collaboration and knowledge exchange in a professional and objective manner.

HOW WE WORK

Make Order

Make Order

Experimental Scheme

Experimental Scheme

Implementation

Implementation

Conclusion

Conclusion
Make an Inquiry