At Protheragen, we offer specialized biomarker analysis services tailored for Idiopathic Pulmonary Fibrosis (IPF) research and therapeutic development. Our comprehensive biomarker panel is designed to support drug discovery and preclinical development by providing deep insights into the molecular and cellular mechanisms underlying IPF pathophysiology. Please note that all Protheragen services are exclusively focused on advancing drug discovery through preclinical stages and do not include clinical diagnostic services.
Effective therapeutic intervention for Idiopathic Pulmonary Fibrosis begins with the discovery and identification of relevant biomarkers. Protheragen’s biomarker discovery services leverage advanced screening technologies to identify molecular signatures associated with disease progression, inflammation, and tissue remodeling. Our approach encompasses high-throughput screening, rigorous validation, and integration of multi-dimensional data to ensure that candidate biomarkers are robust and informative for drug development. The screening and validation process includes initial candidate identification, orthogonal confirmation, and assessment of biological relevance to IPF.
Multi Omics: Our cutting-edge multi-omics platform integrates genomics, transcriptomics, proteomics, and metabolomics to provide a comprehensive understanding of biological systems in Idiopathic Pulmonary Fibrosis. By analyzing DNA, RNA, protein, and metabolite profiles, we enable the identification of novel biomarker candidates and elucidate complex disease pathways such as extracellular matrix remodeling, immune activation, and fibrotic signaling. This holistic approach enhances our ability to dissect the molecular networks involved in IPF, facilitating the discovery of actionable targets for preclinical drug development.
Candidate Validation: Protheragen’s validation strategies combine experimental and computational methods to assess the association of candidate biomarkers with IPF pathophysiology. We conduct preliminary screening using disease-relevant models and biological samples, followed by in-depth evaluation of biomarker specificity, sensitivity, and mechanistic relevance. Promising candidates are prioritized based on criteria such as reproducibility, biological plausibility, and potential utility in monitoring disease activity or therapeutic response.
Diverse Technological Platforms: We offer custom assay development across a range of technological platforms, including immunoassays, mass spectrometry, flow cytometry, molecular diagnostics, and advanced histopathology. Our platforms are adaptable to specific biomarker characteristics and project requirements, ensuring optimal performance and data quality for each analyte of interest.
Immunoassays: We utilize ELISA, chemiluminescent, and multiplex immunoassay formats for quantitative and multiplexed protein detection.
Mass Spectrometry: LC-MS/MS is employed for high-sensitivity, high-specificity quantification and characterization of proteins and peptides.
Flow Cytometry: Multiparametric flow cytometry enables the analysis of cell-surface and intracellular biomarkers within heterogeneous cell populations.
Molecular Diagnostics: Nucleic acid-based assays, including PCR and qPCR, are used for gene expression and mutation analysis.
Histopathology And Imaging: Immunohistochemistry, in situ hybridization, and advanced digital imaging are applied for spatial and morphological biomarker evaluation in tissue samples.
Rigorous Method Validation: All analytical methods undergo rigorous validation in accordance with established guidelines to assess accuracy, precision, specificity, sensitivity, linearity, and reproducibility. Quality control measures are integrated throughout the workflow, including the use of controls, standard curves, and repeat analyses, to ensure data integrity and reliability.
Our quantitative analysis capabilities encompass both absolute and relative quantification of biomarkers in diverse biological matrices. We employ standardized protocols and calibration strategies to enable reproducible and comparable results across experiments, supporting robust decision-making in preclinical research.
Sample Analysis: Protheragen processes a wide range of sample types, including serum, plasma, tissue extracts, and cell culture supernatants. Our analysis protocols are optimized for each matrix to maximize analyte recovery and minimize matrix effects. Stringent quality measures, such as sample tracking, contamination control, and pre-analytical validation, are implemented to safeguard sample integrity and analytical accuracy.
High Throughput Capabilities: Our high-throughput analytical platforms, including multiplex immunoassays and automated sample processing systems, enable the simultaneous analysis of multiple biomarkers from limited sample volumes. This approach increases efficiency, conserves precious samples, and accelerates the generation of comprehensive biomarker profiles for IPF research.
| Gene Target | Biological Function | Application as a Biomarker |
|---|---|---|
| C-C motif chemokine ligand 2 (CCL2) | C-C motif chemokine ligand 2 (CCL2), also known as monocyte chemoattractant protein-1 (MCP-1), is a member of the CC chemokine family. It is primarily involved in the recruitment of monocytes, memory T cells, and dendritic cells to sites of tissue injury and inflammation. CCL2 is produced by a variety of cell types, including endothelial cells, fibroblasts, monocytes, and smooth muscle cells, in response to pro-inflammatory stimuli. By binding to its receptor CCR2, CCL2 mediates chemotaxis and plays a central role in initiating and sustaining inflammatory responses. It is also implicated in the regulation of immune cell trafficking and has been associated with the pathogenesis of several inflammatory and autoimmune diseases. | CCL2 has been studied as a biomarker for various conditions characterized by inflammation and immune activation. Elevated levels of CCL2 in biological fluids, such as blood or cerebrospinal fluid, have been observed in diseases including cardiovascular disease, rheumatoid arthritis, multiple sclerosis, and certain cancers. Its measurement is used in research settings to assess the presence and extent of inflammatory processes, monitor disease activity, and investigate the response to therapeutic interventions. |
| chitinase 3 like 1 (CHI3L1) | Chitinase 3 like 1 (CHI3L1), also known as YKL-40, is a secreted glycoprotein that belongs to the glycoside hydrolase family 18. Unlike true chitinases, CHI3L1 lacks chitinase enzymatic activity due to mutations in its catalytic domain. It is produced by a variety of cell types, including macrophages, neutrophils, chondrocytes, synovial cells, and certain cancer cells. CHI3L1 is involved in several biological processes such as tissue remodeling, inflammation, cell proliferation, and angiogenesis. It has been implicated in the regulation of immune responses and extracellular matrix organization, particularly in the context of inflammation and tissue injury. | CHI3L1 has been studied as a biomarker in multiple clinical contexts, primarily due to its elevated expression in various inflammatory and neoplastic conditions. Increased levels of CHI3L1 have been observed in the serum, plasma, or synovial fluid of patients with diseases such as rheumatoid arthritis, osteoarthritis, asthma, chronic obstructive pulmonary disease, liver fibrosis, and several types of cancer. It has been investigated for its potential utility in monitoring disease activity, progression, and prognosis, as well as in differentiating between disease states. |
| interleukin 1 receptor antagonist (IL1RN) | Interleukin 1 receptor antagonist (IL1RN) is a naturally occurring anti-inflammatory protein that belongs to the interleukin 1 cytokine family. IL1RN binds competitively to the interleukin 1 type I receptor (IL-1RI) without eliciting a signal, thereby inhibiting the binding and activity of pro-inflammatory cytokines interleukin-1 alpha (IL-1α) and interleukin-1 beta (IL-1β). This antagonistic action regulates the immune response by limiting the effects of IL-1-mediated inflammation, fever, and tissue destruction. IL1RN is produced by a variety of cell types, including monocytes, macrophages, neutrophils, and epithelial cells, and plays a critical role in controlling inflammatory processes in both normal and disease states. | IL1RN has been investigated as a biomarker in several inflammatory and autoimmune conditions. Its concentration in biological fluids, such as serum or synovial fluid, has been measured in the context of diseases including rheumatoid arthritis, sepsis, and inflammatory bowel disease. Changes in IL1RN levels have been associated with the presence and severity of inflammation, and its measurement has been used to monitor disease activity, assess prognosis, and evaluate the response to anti-inflammatory therapies in research and clinical studies. |
| interleukin 4 (IL4) | Interleukin 4 (IL4) is a cytokine produced primarily by activated T helper 2 (Th2) cells, mast cells, and basophils. It plays a central role in regulating immune responses, particularly by promoting the differentiation of naive CD4+ T cells into Th2 cells. IL4 stimulates B cell proliferation, survival, and class switching to immunoglobulin E (IgE) and IgG1 isotypes. It also suppresses the production of pro-inflammatory cytokines and inhibits the differentiation of Th1 cells, thereby contributing to the regulation of humoral and allergic immune responses. IL4 influences the expression of major histocompatibility complex (MHC) class II molecules and the upregulation of adhesion molecules on endothelial cells, further impacting immune cell interactions. | IL4 levels have been measured in various biological fluids as an indicator of Th2-mediated immune activity. Elevated IL4 concentrations have been associated with allergic diseases such as asthma, atopic dermatitis, and allergic rhinitis. It has also been studied in the context of autoimmune diseases, certain infections, and cancer, where its presence may reflect immune modulation or disease activity. IL4 is commonly assessed in research and clinical studies to monitor immune responses and to help characterize immunological profiles in various disease states. |
| interleukin 6 (IL6) | Interleukin 6 (IL6) is a multifunctional cytokine produced by a variety of cell types, including monocytes, macrophages, T cells, B cells, fibroblasts, and endothelial cells. Its primary biological functions involve the regulation of immune responses, inflammation, hematopoiesis, and the acute phase response. IL6 acts as both a pro-inflammatory and anti-inflammatory cytokine, depending on the cellular context. It stimulates the differentiation of B cells into plasma cells, promotes T cell proliferation and differentiation, and induces the hepatic synthesis of acute-phase proteins such as C-reactive protein (CRP) and fibrinogen. Additionally, IL6 plays a role in the regulation of metabolic, regenerative, and neural processes. | IL6 is widely used as a biomarker to assess the presence and severity of inflammation and immune activation. Elevated levels of IL6 in blood or other bodily fluids have been associated with infectious diseases, autoimmune disorders, inflammatory conditions, and certain cancers. In clinical settings, IL6 measurement can aid in the evaluation of disease activity, monitoring of treatment response, and prognosis in conditions such as sepsis, rheumatoid arthritis, and COVID-19, among others. |
| matrix metallopeptidase 2 (MMP2) | Matrix metallopeptidase 2 (MMP2) is a member of the matrix metalloproteinase family of zinc-dependent endopeptidases. MMP2 primarily degrades components of the extracellular matrix (ECM), including type IV collagen, gelatin, and elastin. This proteolytic activity is essential for normal physiological processes such as embryonic development, tissue remodeling, angiogenesis, and wound healing. MMP2 activity is tightly regulated at the levels of transcription, activation of the zymogen form, and inhibition by tissue inhibitors of metalloproteinases (TIMPs). Dysregulation of MMP2 expression or activity can contribute to pathological processes involving excessive ECM degradation. | MMP2 has been studied as a biomarker in various pathological conditions, particularly in cancer, cardiovascular diseases, and fibrotic disorders. In oncology, elevated MMP2 expression or activity in tumor tissue or biological fluids has been associated with tumor invasion, metastasis, and poor prognosis in several cancer types. In cardiovascular disease, increased MMP2 levels have been observed in conditions such as atherosclerosis and heart failure, reflecting its role in ECM remodeling. MMP2 has also been investigated in the context of chronic inflammatory and fibrotic diseases, where its levels may correlate with disease progression or severity. |
| matrix metallopeptidase 7 (MMP7) | Matrix metallopeptidase 7 (MMP7), also known as matrilysin, is a member of the matrix metalloproteinase (MMP) family. MMP7 is a zinc-dependent endopeptidase that primarily degrades components of the extracellular matrix (ECM), including proteoglycans, fibronectin, elastin, and casein. Unlike many other MMPs, MMP7 has a broad substrate specificity and is secreted by epithelial cells rather than stromal cells. It participates in tissue remodeling, wound healing, and the regulation of inflammatory processes. MMP7 also plays a role in the activation of other proteinases and in the cleavage of cell surface molecules, thereby influencing cell signaling, migration, and apoptosis. | MMP7 has been investigated as a biomarker in several disease contexts, particularly in oncology and fibrotic diseases. Elevated levels of MMP7 have been reported in the serum, plasma, or tissue of individuals with various cancers, including colorectal, pancreatic, and lung cancer, as well as in chronic inflammatory and fibrotic conditions such as idiopathic pulmonary fibrosis (IPF) and inflammatory bowel disease. In these contexts, MMP7 measurements have been used in research studies to assess disease presence, activity, or prognosis. |
| telomerase reverse transcriptase (TERT) | Telomerase reverse transcriptase (TERT) is the catalytic subunit of the enzyme telomerase, which is responsible for the addition of telomeric repeats (TTAGGG) to the ends of linear chromosomes. This process maintains telomere length, thereby preserving chromosomal stability and integrity during cell division. In most somatic cells, TERT expression is low or absent, resulting in progressive telomere shortening and eventual cellular senescence. In contrast, TERT is actively expressed in stem cells, germ cells, and the majority of cancer cells, supporting sustained proliferative capacity by counteracting telomere attrition. | TERT has been utilized as a biomarker in various clinical and research contexts. Elevated TERT expression or the presence of TERT promoter mutations has been associated with a range of malignancies, including gliomas, melanomas, and urothelial cancers. Detection of TERT promoter mutations or increased TERT mRNA levels can aid in tumor diagnosis, prognostication, and monitoring of disease progression. Additionally, TERT alterations may serve as indicators of malignant transformation in certain precancerous lesions. |
| transforming growth factor beta 1 (TGFB1) | Transforming growth factor beta 1 (TGFB1) is a multifunctional cytokine that plays a pivotal role in the regulation of cell growth, differentiation, apoptosis, and immune responses. TGFB1 is a member of the TGF-beta superfamily and is synthesized as a latent precursor that is activated extracellularly. It exerts its effects by binding to specific serine/threonine kinase receptors, which activate intracellular SMAD signaling pathways. TGFB1 is involved in the maintenance of tissue homeostasis, modulation of inflammation, regulation of extracellular matrix production, and control of cellular proliferation. It is also a key mediator in processes such as wound healing, fibrosis, and immune system regulation. | TGFB1 has been studied as a biomarker in various pathological conditions, including fibrotic diseases, cancer, and immune-mediated disorders. Elevated levels of TGFB1 in biological fluids or tissues have been associated with the progression of organ fibrosis (such as in liver, lung, and kidney), as well as with tumor progression and metastasis in several cancer types. Measurement of TGFB1 expression or concentration has been used in research settings to assess disease activity, prognosis, and response to therapy in these contexts. |
| tumor necrosis factor (TNF) | Tumor necrosis factor (TNF) is a pro-inflammatory cytokine primarily produced by activated macrophages, as well as by a variety of other immune and non-immune cells. TNF plays a central role in the regulation of immune responses, inflammation, and apoptosis. It acts by binding to its receptors, TNFR1 and TNFR2, leading to the activation of intracellular signaling pathways such as NF-κB and MAPK. These pathways mediate the expression of genes involved in inflammation, cell survival, proliferation, and death. TNF is critical in the defense against infections and in the pathogenesis of various inflammatory and autoimmune diseases. | TNF is measured in biological fluids such as serum, plasma, or synovial fluid to assess inflammatory status. Its concentration has been associated with disease activity and severity in conditions such as rheumatoid arthritis, inflammatory bowel disease, sepsis, and certain cancers. TNF levels are used in research and clinical studies to monitor response to anti-TNF therapies and to investigate the inflammatory state in various disorders. |
Explore Research Opportunities with Protheragen. Our biomarker research services for Idiopathic Pulmonary Fibrosis leverage advanced analytical technologies and a comprehensive panel of research targets to support drug discovery and preclinical development. All biomarkers discussed are for research use only and serve as exploratory targets; we do not claim any biomarker as validated or mandatory for IPF research. Our focus is on providing scientific insights and supporting preclinical research with objectivity and rigor.
We invite you to connect with Protheragen to discuss collaborative opportunities in biomarker research for Idiopathic Pulmonary Fibrosis. Our team is committed to advancing scientific discovery through exploratory research and knowledge exchange. Let’s work together to drive innovation in preclinical biomarker studies.
Make Order
Experimental Scheme
Implementation
Conclusion