Protheragen offers specialized biomarker analysis services dedicated to advancing Alopecia research and therapeutic development. Our comprehensive biomarker panel is designed to provide deep insights into the molecular and cellular mechanisms underlying Alopecia, supporting drug discovery and preclinical development. Please note that all Protheragen services are exclusively focused on research applications in drug discovery through preclinical development stages and do not include any clinical diagnostic services.
The foundation of effective therapeutic intervention for Alopecia lies in the precise discovery and identification of relevant biomarkers. At Protheragen, our biomarker discovery services leverage advanced screening methodologies to identify molecular targets associated with disease mechanisms. Through systematic screening and rigorous validation, we ensure that potential biomarkers are robustly characterized, enabling their integration into early-stage drug development pipelines. Our approach supports the identification of key molecular signatures that inform target selection and therapeutic strategy.
Multi Omics: Protheragen employs a cutting-edge multi-omics approach, integrating genomics, transcriptomics, proteomics, and metabolomics technologies to comprehensively study biological systems relevant to Alopecia. This enables the identification of DNA, RNA, protein, and metabolite biomarkers associated with disease pathways such as Wnt/β-catenin signaling, growth factor signaling (e.g., FGF5, IGF1R), inflammatory cytokines (e.g., IL17A, TNF), and androgen metabolism (e.g., SRD5A2). By utilizing high-throughput sequencing, quantitative proteomics, and molecular profiling, we provide a holistic view of the molecular landscape driving Alopecia pathophysiology.
Candidate Validation: Our candidate validation and prioritization strategies involve multi-tiered approaches including in vitro and in vivo models, molecular assays, and bioinformatics analyses. We assess the association of candidate biomarkers with Alopecia pathophysiology by evaluating their expression patterns, functional relevance, and involvement in disease-related pathways. Preliminary screening processes filter candidates based on reproducibility, specificity, and biological plausibility. Promising biomarkers are prioritized according to criteria such as differential expression, pathway involvement, and translational potential for therapeutic development.
Diverse Technological Platforms: Protheragen offers custom assay development capabilities tailored to Alopecia research needs. Our platforms are adaptable for diverse biomarker classes and sample types, ensuring robust and sensitive detection. We utilize a range of analytical platforms including immunoassays, mass spectrometry, flow cytometry, molecular diagnostics, and advanced histopathology and imaging systems. This flexibility allows us to address specific research requirements and optimize assay performance for each biomarker target.
Immunoassays: We develop and implement ELISA, chemiluminescent, and multiplex immunoassays for sensitive and quantitative measurement of protein biomarkers.
Mass Spectrometry: Our LC-MS/MS workflows provide high-resolution, quantitative analysis of proteins, peptides, and small molecules relevant to Alopecia.
Flow Cytometry: We utilize flow cytometry for cell-based biomarker detection, enabling multiparametric analysis of immune and cellular markers.
Molecular Diagnostics: Our molecular diagnostic assays include quantitative PCR, digital PCR, and next-generation sequencing for DNA and RNA biomarker analysis.
Histopathology And Imaging: We offer advanced histopathology and imaging solutions for spatial localization and visualization of biomarker expression in tissue samples.
Rigorous Method Validation: All analytical methods undergo a rigorous validation process in accordance with established research guidelines. Performance characteristics such as sensitivity, specificity, linearity, reproducibility, and robustness are thoroughly evaluated. Comprehensive quality control measures are implemented throughout assay development and execution to ensure data integrity and reliability, supporting robust preclinical research outcomes.
Our quantitative analysis capabilities enable precise measurement of biomarker levels across a range of sample types. Utilizing validated protocols and calibrated instrumentation, we provide accurate, reproducible, and sensitive quantification of DNA, RNA, protein, and metabolite biomarkers. This quantitative approach supports data-driven decision-making in Alopecia drug discovery and preclinical research.
Sample Analysis: Protheragen processes a variety of sample types including blood, serum, plasma, tissue biopsies, and cultured cells. Detailed sample handling and analysis protocols are implemented to preserve biomarker integrity and minimize pre-analytical variability. Our laboratory adheres to stringent quality measures, including standardized operating procedures and internal controls, to ensure reliable and reproducible results for exploratory biomarker research.
High Throughput Capabilities: We offer high-throughput analytical platforms capable of multiplexed biomarker analysis, enabling the simultaneous assessment of multiple targets from limited sample volumes. This approach enhances research efficiency, conserves valuable samples, and accelerates the generation of comprehensive biomarker profiles. Our systems are optimized for scalability, supporting both small-scale exploratory studies and larger preclinical research projects.
| Gene Target | Biological Function | Application as a Biomarker |
|---|---|---|
| catenin beta 1 (CTNNB1) | Catenin beta 1 (CTNNB1) encodes beta-catenin, a multifunctional protein involved in the regulation of cell-cell adhesion and gene transcription. In the context of cell adhesion, beta-catenin binds to the cytoplasmic domain of cadherins, linking them to the actin cytoskeleton and thereby contributing to the maintenance of epithelial tissue architecture. Beta-catenin is also a central component of the canonical Wnt signaling pathway. In the absence of Wnt signals, beta-catenin is targeted for degradation by a destruction complex. Upon Wnt activation, beta-catenin accumulates in the cytoplasm and translocates to the nucleus, where it interacts with TCF/LEF transcription factors to regulate the expression of genes involved in cell proliferation, differentiation, and survival. | CTNNB1 is used as a biomarker in several clinical and research contexts. Immunohistochemical detection of beta-catenin localization is applied in the diagnosis and characterization of certain tumors, such as hepatocellular carcinoma, desmoid-type fibromatosis, and colorectal cancer. Aberrant nuclear accumulation of beta-catenin, often due to CTNNB1 mutations, is associated with activation of the Wnt signaling pathway and can assist in distinguishing specific tumor types and subtypes. Additionally, CTNNB1 mutation status is evaluated in molecular pathology for its association with tumorigenesis and potential therapeutic implications. |
| fibroblast growth factor 5 (FGF5) | Fibroblast growth factor 5 (FGF5) is a member of the fibroblast growth factor family, which is involved in a broad range of biological processes including cell growth, differentiation, and tissue repair. FGF5 functions primarily as a signaling molecule that binds to FGF receptors, activating downstream pathways such as the MAPK/ERK cascade. In mammals, FGF5 is best known for its role in regulating the hair growth cycle, particularly the transition from the anagen (growth) phase to the catagen (regression) phase. Loss-of-function mutations in FGF5 result in prolonged hair growth due to delayed entry into the catagen phase. Beyond hair follicle biology, FGF5 has been implicated in the regulation of cardiovascular development and function, as well as in modulating angiogenesis and cell proliferation in certain tissues. | FGF5 has been investigated as a biomarker in several contexts. Elevated expression of FGF5 has been observed in certain malignancies, including prostate and breast cancers, where it has been studied for potential association with tumor progression and prognosis. Additionally, FGF5 gene polymorphisms have been explored in relation to hypertension, with some studies examining its utility in genetic risk stratification. In dermatology, FGF5 is used as a molecular marker to study hair growth disorders and to evaluate the effects of therapeutic agents targeting the hair cycle. Its expression levels and genetic variants are thus measured in research settings to provide insights into disease mechanisms and potential therapeutic responses. |
| glycogen synthase kinase 3 beta (GSK3B) | Glycogen synthase kinase 3 beta (GSK3B) is a serine/threonine kinase that plays a central role in multiple cellular processes. It is involved in the regulation of glycogen metabolism by phosphorylating and inhibiting glycogen synthase, the enzyme responsible for converting glucose to glycogen. Beyond metabolic regulation, GSK3B is a key component of several signaling pathways, including the Wnt/β-catenin, insulin, and PI3K/Akt pathways. It influences cell proliferation, differentiation, apoptosis, and gene expression by phosphorylating various substrates such as transcription factors, structural proteins, and metabolic enzymes. GSK3B activity is tightly regulated by upstream kinases and phosphatases, and its dysregulation has been associated with the pathophysiology of diverse disorders. | GSK3B has been studied as a biomarker in several disease contexts. Altered expression levels or activity of GSK3B have been reported in neurodegenerative diseases such as Alzheimer's disease, where increased kinase activity is associated with abnormal tau phosphorylation. In oncology, aberrant GSK3B signaling has been observed in certain cancers, and its expression or phosphorylation status has been investigated in relation to tumor progression and response to therapy. Additionally, GSK3B has been explored in the context of mood disorders, particularly bipolar disorder, due to its involvement in neuronal signaling pathways. These applications are based on correlations between GSK3B status and disease phenotypes in clinical and experimental studies. |
| insulin like growth factor 1 receptor (IGF1R) | The insulin-like growth factor 1 receptor (IGF1R) is a transmembrane tyrosine kinase receptor that is activated primarily by its ligand, insulin-like growth factor 1 (IGF-1). Upon ligand binding, IGF1R undergoes autophosphorylation and initiates intracellular signaling cascades, notably the PI3K-AKT and MAPK pathways. These pathways regulate a variety of cellular processes, including proliferation, differentiation, survival, and metabolism. IGF1R plays a significant role in normal growth and development, as well as in tissue repair. Dysregulation of IGF1R signaling has been implicated in several pathological conditions, including cancer, due to its involvement in promoting cell survival and resistance to apoptosis. | IGF1R has been studied as a biomarker in various clinical contexts. In oncology, IGF1R expression levels have been investigated in relation to tumor progression, prognosis, and response to targeted therapies, particularly in cancers such as breast, lung, and sarcomas. Elevated IGF1R expression has been associated with certain tumor subtypes and resistance to some anticancer treatments. Additionally, IGF1R has been explored as a potential biomarker for predicting response to IGF1R-targeted therapies. Beyond oncology, IGF1R has been evaluated in metabolic and growth disorders, such as growth hormone insensitivity syndromes, where receptor function or expression may be altered. |
| interleukin 17A (IL17A) | Interleukin 17A (IL17A) is a pro-inflammatory cytokine produced primarily by a subset of T helper cells known as Th17 cells. IL17A plays a critical role in host defense against extracellular bacteria and fungi by inducing the expression of antimicrobial peptides, chemokines, and other cytokines in various cell types, including epithelial cells, endothelial cells, and fibroblasts. This leads to the recruitment and activation of neutrophils and other immune cells at sites of infection or tissue injury. IL17A is also involved in the regulation of tissue inflammation and has been implicated in the pathogenesis of several autoimmune and inflammatory disorders due to its ability to promote chronic inflammatory responses. | IL17A has been utilized as a biomarker in multiple clinical and research settings to assess inflammatory activity, particularly in autoimmune and inflammatory diseases such as psoriasis, rheumatoid arthritis, ankylosing spondylitis, and inflammatory bowel disease. Its levels in blood, tissue, or other biological fluids have been measured to evaluate disease activity, monitor therapeutic response, and characterize immune profiles in affected individuals. Elevated IL17A concentrations have been associated with disease severity and progression in certain conditions. |
| patched 1 (PTCH1) | Patched 1 (PTCH1) is a transmembrane glycoprotein that functions as the primary receptor for members of the Hedgehog (Hh) family of signaling proteins. PTCH1 is a key component of the Hedgehog signaling pathway, which regulates cell growth, differentiation, and tissue patterning during embryonic development. In the absence of Hedgehog ligands, PTCH1 inhibits the activity of the Smoothened (SMO) protein, thereby repressing downstream signaling. When Hedgehog ligands bind to PTCH1, this inhibition is relieved, allowing SMO activation and subsequent transcription of Hedgehog target genes. PTCH1 also plays a role in maintaining tissue homeostasis in adults and acts as a tumor suppressor by regulating cellular proliferation. | PTCH1 has been utilized as a biomarker in various clinical and research contexts, particularly in oncology. Mutations or loss of function in PTCH1 are associated with several cancers, including basal cell carcinoma, medulloblastoma, and nevoid basal cell carcinoma syndrome (Gorlin syndrome). Detection of PTCH1 mutations or altered expression can aid in the diagnosis, risk assessment, and molecular characterization of these conditions. PTCH1 status is also examined to help identify tumors with aberrant Hedgehog pathway activation, which may have therapeutic implications. |
| steroid 5 alpha-reductase 2 (SRD5A2) | Steroid 5 alpha-reductase 2 (SRD5A2) is an enzyme that catalyzes the conversion of testosterone to dihydrotestosterone (DHT), a more potent androgen. This reaction is a key step in androgen metabolism, particularly in tissues such as the prostate, skin, and reproductive organs. SRD5A2 is encoded by the SRD5A2 gene and is primarily expressed in androgen-sensitive tissues. DHT produced by SRD5A2 plays a crucial role in the development of male external genitalia during embryogenesis and contributes to the growth and function of the prostate and other androgen-dependent tissues in adulthood. | SRD5A2 has been studied as a biomarker in several clinical contexts. Mutations in the SRD5A2 gene are associated with 5 alpha-reductase deficiency, a condition characterized by impaired virilization in genetic males. Assessment of SRD5A2 expression or activity has been utilized in the evaluation of prostate disorders, including benign prostatic hyperplasia and prostate cancer. Additionally, SRD5A2 genotyping may inform pharmacogenetic responses to 5 alpha-reductase inhibitors used in the treatment of these conditions. |
| transforming growth factor beta 2 (TGFB2) | Transforming growth factor beta 2 (TGFB2) is a member of the TGF-beta superfamily of cytokines, which are multifunctional peptides regulating cell proliferation, differentiation, migration, and apoptosis. TGFB2 is involved in the regulation of embryonic development, immune system modulation, extracellular matrix production, and tissue repair. It exerts its effects primarily through binding to TGF-beta receptors, activating SMAD-dependent and SMAD-independent signaling pathways. TGFB2 plays a critical role in maintaining tissue homeostasis and has been implicated in processes such as fibrosis, angiogenesis, and regulation of inflammatory responses. | TGFB2 has been studied as a biomarker in various pathological conditions, including fibrotic diseases, cardiovascular disorders, and certain cancers. Altered expression levels of TGFB2 have been associated with disease progression and severity in these contexts. For example, increased TGFB2 expression has been observed in some tumors and fibrotic tissues, correlating with disease state or prognosis. Measurement of TGFB2 levels in biological samples (such as plasma, serum, or tissue biopsies) has been utilized in research settings to assess its potential as an indicator of disease activity or therapeutic response. |
| tumor necrosis factor (TNF) | Tumor necrosis factor (TNF), also known as TNF-alpha, is a pro-inflammatory cytokine primarily produced by activated macrophages, T lymphocytes, and natural killer (NK) cells. TNF plays a central role in the regulation of immune responses, inflammation, and apoptosis. It mediates its effects by binding to two distinct cell surface receptors, TNFR1 and TNFR2, triggering intracellular signaling cascades that result in the activation of nuclear factor-kappa B (NF-κB) and mitogen-activated protein kinases (MAPKs). These pathways regulate the expression of genes involved in inflammation, cell proliferation, differentiation, and survival. TNF is involved in host defense against infections and tumor cells, but excessive or dysregulated TNF production is associated with chronic inflammatory and autoimmune diseases. | TNF levels in blood, serum, or tissue samples have been utilized as a biomarker to assess the degree of inflammation and immune activation in various clinical contexts. Elevated TNF concentrations have been observed in conditions such as rheumatoid arthritis, inflammatory bowel disease, sepsis, and certain malignancies. Measurement of TNF can provide supportive information about disease activity, prognosis, and response to anti-TNF therapies in inflammatory and autoimmune disorders. |
Explore Research Opportunities with Protheragen. Our biomarker research services offer advanced capabilities for the discovery, validation, and analysis of molecular targets relevant to Alopecia. We emphasize the exploratory and research-focused nature of our work, providing comprehensive support from target identification through preclinical development. Please note that all biomarkers discussed are research targets only; we do not claim any biomarkers as validated or mandatory for Alopecia research. Our services are exclusively focused on preclinical research stages, maintaining scientific objectivity and rigor throughout every project.
We invite you to connect with Protheragen to discuss collaborative opportunities in exploratory biomarker research for Alopecia. Our team is dedicated to scientific collaboration and knowledge exchange, supporting innovative approaches in preclinical research. Let's advance the understanding of Alopecia together through objective, data-driven biomarker analysis.
Make Order
Experimental Scheme
Implementation
Conclusion