Biomarker Analysis Services for Systemic Mastocytosis
Drug R&D Solutions

Biomarker Analysis Services for Systemic Mastocytosis

Inquiry

At Protheragen, we offer specialized biomarker analysis services dedicated to advancing research and therapeutic development for Systemic Mastocytosis. Our comprehensive biomarker panel is designed to facilitate a deep understanding of disease pathophysiology, supporting the discovery and evaluation of novel drug targets. All services are exclusively focused on drug discovery through preclinical development stages; we do not provide clinical diagnostic services.

Biomarker Discovery and Identification

The foundation of effective therapeutic intervention lies in the identification of precise and actionable biomarkers. Protheragen’s biomarker discovery services leverage state-of-the-art molecular screening and validation strategies to support drug development for Systemic Mastocytosis. Our approach includes the systematic screening of candidate genes, proteins, and cellular markers, followed by rigorous validation to confirm their relevance and utility in preclinical models. This process enables the identification of key molecular drivers and supports the rational design of targeted therapeutics.

Multi Omics: Our multi-omics approach integrates cutting-edge genomics, transcriptomics, and proteomics technologies to enable a comprehensive study of biological systems relevant to Systemic Mastocytosis. By analyzing DNA, RNA, protein, and metabolite profiles, we identify and characterize candidate biomarkers associated with disease-related pathways such as tyrosine kinase signaling, apoptosis regulation, and immune cell activation. This holistic strategy allows for the elucidation of complex molecular networks and the discovery of novel targets for therapeutic intervention.

Candidate Validation: Protheragen employs robust validation strategies to prioritize biomarker candidates associated with Systemic Mastocytosis pathophysiology. Preliminary screening includes functional assays, expression profiling, and pathway analysis to establish associations with disease mechanisms such as aberrant mast cell proliferation, tyrosine kinase activation, and immune regulation. Criteria for prioritizing promising candidates include biological relevance, reproducibility, and potential for assay development, ensuring that only the most informative markers advance for further study.

Biomarker Assay Development and Validation

Diverse Technological Platforms: We offer custom assay development capabilities across a diverse range of technological platforms, including immunoassays, mass spectrometry, flow cytometry, molecular diagnostics, and advanced histopathology and imaging. Our platforms are adaptable to specific research requirements, allowing for the sensitive and specific detection of biomarkers in a variety of sample types.

Immunoassays: We develop and optimize ELISA, chemiluminescent, and multiplex immunoassays for the sensitive quantification of protein biomarkers.

Mass Spectrometry: Our LC-MS/MS platforms enable precise and high-throughput analysis of proteins and metabolites relevant to Systemic Mastocytosis.

Flow Cytometry: We utilize flow cytometry for immunophenotyping and quantification of cell surface markers, supporting the identification of mast cell and eosinophil populations.

Molecular Diagnostics: Our molecular diagnostic assays include PCR-based and sequencing methods for the detection of gene mutations and expression changes.

Histopathology And Imaging: Advanced histological and imaging techniques are employed for spatial localization and quantification of biomarkers in tissue sections.

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

Biomarker Quantitative and Qualitative Analysis

Our quantitative analysis capabilities enable accurate measurement of biomarker levels across diverse sample matrices. We employ standardized protocols and calibration procedures to ensure reproducibility and comparability of results, supporting the robust assessment of candidate biomarkers during preclinical studies.

Sample Analysis: Protheragen handles a wide range of sample types, including cell lines, primary cells, and tissue specimens relevant to Systemic Mastocytosis research. Detailed analysis protocols are tailored to each sample type, incorporating stringent quality measures such as sample tracking, integrity assessment, and contamination control to ensure high-quality, reliable data.

High Throughput Capabilities: Our high-throughput analytical platforms support multiplexed biomarker analysis, enabling efficient processing of large sample cohorts. Multiplexing strategies conserve valuable samples, reduce assay time, and enhance data generation, accelerating the pace of preclinical research and discovery.

Key Biomarkers for Systemic Mastocytosis Drug Development

Gene Target Biological Function Application as a Biomarker
BCL2 apoptosis regulator (BCL2) The BCL2 apoptosis regulator (BCL2) is a key member of the B-cell lymphoma 2 (BCL2) protein family. BCL2 is primarily localized to the outer mitochondrial membrane, where it functions as an anti-apoptotic protein. It inhibits the intrinsic (mitochondrial) pathway of apoptosis by binding and sequestering pro-apoptotic proteins such as BAX and BAK, thereby preventing mitochondrial outer membrane permeabilization and subsequent release of cytochrome c. Through this mechanism, BCL2 regulates cell survival and plays a crucial role in maintaining tissue homeostasis by controlling the balance between cell death and survival. BCL2 expression is frequently assessed as a biomarker in various malignancies, particularly in hematologic cancers such as follicular lymphoma and chronic lymphocytic leukemia. Its expression levels can aid in the classification and prognosis of lymphoid neoplasms. In some contexts, BCL2 immunohistochemistry is used to help distinguish between different subtypes of lymphoma. Additionally, BCL2 status may inform therapeutic decisions, especially in the context of targeted therapies that inhibit BCL2 function.
BCL2 like 1 (BCL2L1) BCL2 like 1 (BCL2L1) encodes proteins belonging to the BCL-2 family, which are critical regulators of apoptosis. The gene produces multiple isoforms through alternative splicing, most notably Bcl-xL (anti-apoptotic) and Bcl-xS (pro-apoptotic). Bcl-xL inhibits apoptosis by binding to and sequestering pro-apoptotic proteins such as BAX and BAK, thereby preventing mitochondrial outer membrane permeabilization and subsequent cytochrome c release. This function is essential for cell survival, tissue homeostasis, and regulation of cellular responses to stress. Dysregulation of BCL2L1 expression or function can disrupt the balance between cell survival and death, contributing to various pathological conditions. BCL2L1 expression levels have been investigated as a biomarker in several contexts, particularly in oncology. Elevated BCL2L1, especially the Bcl-xL isoform, has been associated with resistance to apoptosis in various malignancies, including hematological cancers and solid tumors. Its expression has been studied in relation to prognosis, response to chemotherapy, and potential for targeted therapeutic intervention. Additionally, BCL2L1 has been evaluated as part of gene expression panels to stratify risk or predict treatment outcomes in cancer patients.
Janus kinase 2 (JAK2) Janus kinase 2 (JAK2) is a non-receptor tyrosine kinase that plays a critical role in the signaling pathways of various cytokines and growth factors. Upon ligand binding to cytokine receptors, JAK2 becomes activated through trans-phosphorylation, leading to phosphorylation of specific tyrosine residues on the receptor. This creates docking sites for STAT (Signal Transducer and Activator of Transcription) proteins, which are subsequently phosphorylated by JAK2. Activated STAT proteins dimerize and translocate to the nucleus, where they regulate the transcription of target genes involved in cell proliferation, differentiation, apoptosis, and immune function. JAK2 is particularly important in hematopoiesis, mediating responses to erythropoietin, thrombopoietin, and other hematopoietic growth factors. JAK2 is used as a biomarker primarily in the context of myeloproliferative neoplasms (MPNs). The presence of the JAK2 V617F mutation is frequently detected in patients with polycythemia vera, essential thrombocythemia, and primary myelofibrosis. Detection of this mutation aids in the diagnosis and classification of these hematologic disorders. Additionally, JAK2 mutation analysis can assist in distinguishing MPNs from other causes of similar clinical presentations and may provide information relevant to prognosis and therapeutic decision-making.
KIT proto-oncogene, receptor tyrosine kinase (KIT) The KIT proto-oncogene, receptor tyrosine kinase (KIT), encodes a transmembrane receptor tyrosine kinase that is a member of the type III receptor tyrosine kinase family. KIT binds to its ligand, stem cell factor (SCF), leading to receptor dimerization, autophosphorylation, and activation of downstream signaling pathways such as PI3K/AKT, RAS/MAPK, and JAK/STAT. These signaling cascades regulate diverse cellular processes including proliferation, survival, differentiation, and migration. KIT plays critical roles in the development and maintenance of hematopoietic stem cells, melanocytes, germ cells, and interstitial cells of Cajal. KIT is utilized as a biomarker in several clinical and research contexts. Immunohistochemical detection of KIT protein is commonly used in the diagnosis of gastrointestinal stromal tumors (GISTs), where KIT expression is frequently observed. Mutations in the KIT gene are also associated with certain subtypes of acute myeloid leukemia, mastocytosis, and melanoma, and detection of these mutations can inform prognosis and potential therapeutic strategies. The presence or absence of KIT expression or mutation status is used to support differential diagnosis and guide targeted therapy decisions.
platelet derived growth factor receptor alpha (PDGFRA) Platelet derived growth factor receptor alpha (PDGFRA) is a cell surface tyrosine kinase receptor that binds members of the platelet-derived growth factor (PDGF) family. Upon ligand binding, PDGFRA undergoes dimerization and autophosphorylation, activating multiple downstream signaling pathways including the PI3K-AKT, RAS-MAPK, and PLCγ pathways. These signaling cascades regulate cellular processes such as proliferation, differentiation, migration, and survival. PDGFRA is expressed in various cell types, particularly mesenchymal cells, and plays an essential role in embryonic development, tissue repair, and maintenance of connective tissues. PDGFRA has been utilized as a biomarker in several clinical and research contexts. Its expression and mutational status are assessed in gastrointestinal stromal tumors (GIST), where activating mutations in PDGFRA can inform diagnosis and guide targeted therapy selection. PDGFRA alterations have also been evaluated in other malignancies, such as gliomas, where amplification or overexpression may be associated with tumor classification and prognosis. Immunohistochemical detection of PDGFRA protein and molecular testing for PDGFRA gene mutations are commonly applied in these settings.
platelet derived growth factor receptor beta (PDGFRB) Platelet derived growth factor receptor beta (PDGFRB) is a cell surface tyrosine kinase receptor that primarily binds members of the platelet-derived growth factor (PDGF) family. Upon ligand binding, PDGFRB dimerizes and undergoes autophosphorylation, activating downstream signaling pathways such as the PI3K-AKT, RAS-MAPK, and PLCγ pathways. These cascades regulate a variety of cellular processes including proliferation, migration, differentiation, and survival. PDGFRB is especially important in the development and maintenance of vascular smooth muscle cells, pericytes, and mesenchymal cells, playing a crucial role in angiogenesis, vascular stability, and tissue repair. PDGFRB expression and activation have been studied as biomarkers in several clinical contexts. In oncology, aberrant PDGFRB signaling has been associated with certain malignancies, including gastrointestinal stromal tumors (GIST), myeloproliferative neoplasms, and some sarcomas, where PDGFRB gene rearrangements or overexpression may be present. PDGFRB is also evaluated in the diagnosis and classification of hematologic disorders such as chronic myelomonocytic leukemia (CMML) and atypical chronic myeloid leukemia (aCML), particularly in cases with PDGFRB gene fusions. Additionally, its expression in stromal and vascular cells is used in research settings to assess angiogenesis and tissue remodeling in various diseases.
sialic acid binding Ig like lectin 8 (SIGLEC8) Sialic acid binding Ig like lectin 8 (SIGLEC8) is a member of the SIGLEC family of cell surface receptors, which are part of the immunoglobulin superfamily. SIGLEC8 is predominantly expressed on eosinophils, mast cells, and, to a lesser extent, basophils. It functions as an inhibitory receptor by recognizing sialylated glycans, specifically 6'-sulfo-sialyl Lewis X motifs, on glycoproteins. Engagement of SIGLEC8 on eosinophils induces apoptosis, while its ligation on mast cells inhibits mediator release. Through these mechanisms, SIGLEC8 plays a regulatory role in modulating immune cell survival and activation, particularly in the context of allergic inflammation and immune homeostasis. SIGLEC8 expression is utilized as a cell surface marker to identify and characterize human eosinophils and mast cells in blood and tissue samples. Its selective expression profile allows for the discrimination of these cell types in immunophenotyping assays, such as flow cytometry and immunohistochemistry. In research settings, SIGLEC8 has been applied to study the involvement of eosinophils and mast cells in allergic diseases, asthma, and certain eosinophilic disorders.

Partner with Protheragen to Advance Innovation

Explore Research Opportunities with Protheragen. Our biomarker research services for Systemic Mastocytosis encompass comprehensive discovery, validation, and assay development capabilities, tailored for exploratory and research-focused applications in preclinical drug development. Please note that all biomarkers discussed are research targets only; we do not claim any as validated or mandatory for Systemic Mastocytosis. Our work is exclusively focused on supporting preclinical research, maintaining scientific objectivity and flexibility to advance your therapeutic discovery programs.

We invite you to connect with Protheragen to discuss collaborative opportunities in exploratory biomarker research for Systemic Mastocytosis. Our team is committed to scientific collaboration and knowledge exchange, supporting objective and innovative approaches to preclinical research.

HOW WE WORK

Make Order

Make Order

Experimental Scheme

Experimental Scheme

Implementation

Implementation

Conclusion

Conclusion
Make an Inquiry