IFN-Omega 1 Human Recombinant produced in E.Coli is a single, non-glycosylated, polypeptide chain containing 172 amino acids and having a molecular mass of 19.9kDa.
The IFN-Omega 1 is purified by proprietary chromatographic techniques.
IFN-Omega 1, a type I interferon, is produced by virus-infected leukocytes. It is similar to other Type I IFNs, including IFN-alpha and IFN-beta, in its antiviral and antiproliferative functions, as well as its signaling pathway, which involves the IFNAR-1/IFNAR-2 receptor complex. IFN-Omega 1 shares about 75% of its amino acid sequence with IFN-alpha. The presence of two conserved disulfide bonds in its structure is crucial for its biological activity.
Recombinant Human IFN-Omega 1, produced in E. coli, is a single, non-glycosylated polypeptide chain. It comprises 172 amino acids, resulting in a molecular weight of 19.9kDa. The purification process involves proprietary chromatographic techniques to ensure the protein's purity.
IFN omega-1, IFN alpha-II-1, IFNW1.
IFNW1 (Interferon omega-1) is a member of the type I interferon family, also known as Interferon alpha-II-1. It belongs to the broader alpha/beta interferon family and functions as a secreted protein involved in immune response . Type I interferons are cytokines that play essential roles in inflammation, immunoregulation, tumor cell recognition, and T-cell responses . Unlike other interferons, IFNW1 has distinct structural and functional characteristics while maintaining the core interferon signaling mechanisms.
IFNW1 serves several critical biological functions:
Antiviral defense through induction of interferon-stimulated genes (ISGs)
Modulation of immune cell activity and inflammation
Potential roles in tumor surveillance and anti-cancer immunity
Regulation of gene expression through interferon-responsive elements
As documented in the INTERFEROME database, type I interferons like IFNW1 regulate numerous genes involved in immune response, cell growth control, and pathogen defense mechanisms . The protein is primarily expressed in immune cells and functions in the extracellular space as a signaling molecule .
Several methodologies can be employed for reliable IFNW1 detection and quantification:
The Human Interferon Omega-1 ELISA Kit offers a detection range of 15.6-1000 pg/mL with high specificity for natural and recombinant human IFNW1 . For optimal results, sample processing should minimize protein degradation and maintain the native state of IFNW1.
When designing experiments to investigate IFNW1 signaling:
Cell System Selection: Choose appropriate cell types that express type I interferon receptors. The INTERFEROME database can help identify cells with robust interferon responses .
Dose-Response Assessment: Establish optimal IFNW1 concentrations. Commercial IFNW1 should be tested with appropriate ranges (typically 1-1000 pg/mL) to determine cell-specific responses .
Time-Course Experiments: Monitor signaling events from early (minutes) to late (hours/days) to capture the complete signaling cascade.
Signaling Readouts:
Phosphorylation of STAT1/STAT2 (Western blot)
Nuclear translocation of STAT complexes (immunofluorescence)
Interferon-stimulated gene expression (qPCR, RNA-seq)
Reporter assays with interferon-sensitive response elements
Controls: Include positive controls (well-characterized type I interferons like IFN-α) and negative controls (receptor-blocking antibodies or pathway inhibitors).
Cross-Species Considerations: For comparative studies, consider universal type I interferon reagents that exhibit bioactivity across species .
Researchers face several technical challenges when working with IFNW1:
Low Endogenous Expression: IFNW1 is often expressed at low levels, making detection difficult.
Solution: Use high-sensitivity detection methods and consider stimulation protocols to enhance expression.
Protein Stability: Like other interferons, IFNW1 may have limited stability in experimental conditions.
Species Specificity: Interferons often show strong species-specificity.
Distinguishing from Other Type I IFNs: IFNW1 shares similarities with other type I interferons.
Solution: Use highly specific antibodies and detection reagents validated for IFNW1 specifically.
Validating Biological Activity: Confirming functional activity can be challenging.
IFNW1, like other type I interferons, orchestrates significant changes in gene expression during immune responses:
Activation of JAK-STAT Pathway: Upon receptor binding, IFNW1 triggers phosphorylation of JAK kinases and subsequent STAT activation.
ISG Induction: The INTERFEROME database documents numerous interferon-regulated genes that respond to type I interferons like IFNW1 . These genes include antiviral effectors, immunomodulators, and cell cycle regulators.
Transcriptional Regulation: Activated STAT complexes bind to interferon-stimulated response elements (ISREs) in promoter regions of target genes.
Temporal Expression Patterns: IFNW1 induces both rapid (primary) and delayed (secondary) gene expression responses, creating cascades of regulation.
Cell-Type Specific Patterns: The INTERFEROME database reveals that interferon responses vary by cell type, with distinct patterns of gene induction in different tissues .
Cross-Talk with Other Pathways: IFNW1 signaling interacts with other cellular pathways including NF-κB, MAPK, and PI3K, creating complex regulatory networks.
The comprehensive analysis of IRG (interferon-regulated gene) signatures can be performed using computational tools available through resources like INTERFEROME .
Distinguishing direct from indirect IFNW1 effects requires sophisticated experimental approaches:
Temporal Analysis: Early responses (30-120 minutes) likely represent direct IFNW1 targets, while later responses (4-24 hours) often include secondary effects.
Transcriptional Inhibition: Using actinomycin D to block new transcription can help identify primary response genes that do not require new protein synthesis.
ChIP-seq: Chromatin immunoprecipitation with antibodies against STAT1/STAT2 can identify genomic regions directly bound by IFNW1-activated transcription factors.
Genetic Approaches: Cells lacking specific signaling components (IFNAR1/2, JAKs, STATs) can help establish dependency relationships.
Bioinformatic Analysis: The INTERFEROME database can assist in identifying interferon signatures in gene lists generated by high-throughput experiments .
Motif Analysis: Examining promoter regions for ISRE and GAS elements can predict direct STAT binding sites.
These approaches collectively enable researchers to map the direct regulatory network downstream of IFNW1 signaling versus secondary cascades.
IFNW1-induced gene expression shows significant tissue specificity:
Tissue-Specific Response Patterns: The INTERFEROME database integrates tissue expression patterns of interferon-regulated genes across 79 human and 61 mouse tissues .
Cell Type Determinants: Different cell types express varying levels of receptor components, signaling molecules, and transcriptional cofactors, resulting in tissue-specific IFNW1 responses.
Basal Expression Variations: INTERFEROME data reveal that interferon-regulated genes have distinct basal expression patterns across tissues, influencing their responsiveness to IFNW1 .
Functional Consequences: Tissue-specific IFNW1 responses may explain differential outcomes during infection, inflammation, or therapeutic interferon administration.
Methodology for Analysis: Researchers can submit gene lists to INTERFEROME to generate heat maps depicting tissue expression patterns, allowing identification of tissue-specific interferon signatures .
This tissue-specific regulation underlies the diverse physiological and pathological roles of IFNW1 throughout the body.
IFNW1 contributes to antiviral immunity through multiple mechanisms:
IFNW1 research offers important insights into autoimmune pathology:
Interferon Signature in Autoimmunity: Many autoimmune diseases exhibit an "interferon signature," with elevated expression of interferon-regulated genes.
Mechanistic Contributions: Studies suggest type I interferons like IFNW1 may contribute to autoimmunity through:
Genetic Associations: Polymorphisms in genes encoding interferon pathway components have been linked to autoimmune susceptibility.
Biomarker Potential: IFNW1 expression or its regulated genes may serve as biomarkers for disease activity or treatment response.
Therapeutic Implications: Understanding IFNW1's role could inform development of targeted therapies for interferon-driven autoimmune conditions.
Research Approaches: The INTERFEROME database provides tools to identify interferon signatures in gene lists from autoimmune disease studies .
Investigating IFNW1 in cancer contexts requires specialized approaches:
Expression Analysis: Examining IFNW1 expression in tumor versus normal tissues using qPCR, immunohistochemistry, or public cancer genomics databases.
Functional Assays:
Direct anti-proliferative effects on cancer cell lines
Apoptosis induction in tumor cells
Effects on cancer cell migration and invasion
Modulation of tumor microenvironment
Immune Cell Interaction: Assessing how IFNW1 affects:
Dendritic cell maturation and function
NK cell activation and cytotoxicity
T cell priming and effector function
Myeloid-derived suppressor cell activity
In Vivo Models: Using mouse models with human tumor xenografts or syngeneic tumors to evaluate IFNW1 effects on tumor growth and immune infiltration.
Combination Approaches: Testing IFNW1 with checkpoint inhibitors, chemotherapy, or radiation to identify synergistic anti-tumor effects.
Transcriptional Profiling: Using the INTERFEROME database to identify cancer-relevant interferon-regulated gene signatures .
Single-cell technologies are revolutionizing IFNW1 research:
Single-Cell RNA Sequencing (scRNA-seq):
Reveals heterogeneity in IFNW1 responses within seemingly homogeneous cell populations
Identifies rare IFNW1-producing cells during immune responses
Maps dynamic changes in IFNW1-responsive transcriptomes at unprecedented resolution
CyTOF/Mass Cytometry:
Simultaneously measures multiple phospho-proteins in IFNW1 signaling pathways
Analyzes 30+ parameters to correlate IFNW1 signaling with cellular phenotypes
Identifies differential responses across immune cell subsets
Spatial Transcriptomics:
Maps IFNW1 expression and responses within tissue microenvironments
Preserves spatial context of IFNW1-responding cells in relation to other cell types
Live-Cell Imaging:
Tracks real-time dynamics of IFNW1 signaling in individual cells
Visualizes STAT nuclear translocation and transcriptional activity
CRISPR Screening:
Identifies genes required for IFNW1 production or response through high-throughput functional genomics
Discovers novel regulatory factors in IFNW1 biology
These technologies enable researchers to address fundamental questions about cell-to-cell variability in IFNW1 responses, spatial organization of responding cells, and temporal dynamics of signaling.
Research into IFNW1-based therapeutics encompasses several strategies:
Recombinant Protein Development:
Delivery Systems:
PEGylation to increase half-life
Nanoparticle encapsulation for targeted delivery
Cell-specific targeting approaches to reduce systemic side effects
Combination Therapies:
Testing IFNW1 with checkpoint inhibitors for cancer treatment
Combining with antiviral drugs for infectious diseases
Exploring synergies with conventional therapies
Biomarker Development:
Identifying patient populations likely to respond to IFNW1-based therapy
Developing companion diagnostics using interferon-regulated gene signatures
Antagonistic Approaches:
Developing IFNW1 signaling inhibitors for autoimmune conditions
Targeted blocking of specific downstream pathways
These approaches leverage the understanding of IFNW1 biology while addressing the challenges of therapeutic development in this field.
Computational approaches offer powerful tools for advancing IFNW1 research:
Systems Biology Modeling:
Creating mathematical models of IFNW1 signaling networks
Simulating pathway dynamics under various conditions
Predicting outcomes of pathway perturbations
Database Resources:
Machine Learning Applications:
Pattern recognition in large-scale IFNW1 response data
Prediction of IFNW1-regulated genes based on promoter elements
Classification of IFNW1 responses across diseases
Network Analysis:
Mapping protein-protein interaction networks involving IFNW1
Identifying key nodes in IFNW1 signaling cascades
Discovering pathway crosstalk with other immune signaling systems
Structural Biology and Molecular Dynamics:
Modeling IFNW1-receptor interactions
Virtual screening for modulators of IFNW1 signaling
Predicting functional consequences of genetic variants
These computational approaches complement experimental studies, accelerating discovery by generating testable hypotheses and providing frameworks for interpreting complex datasets.
IFN-Omega 1 was discovered relatively recently compared to other interferons. It is naturally occurring in the human body and is produced through genetic engineering for research and therapeutic purposes . The protein consists of 172 or 174 amino acids and has an apparent molecular mass of about 25 kDa . It contains a single carbohydrate group, which is mainly composed of biantennary complex oligosaccharides with variable amounts of N-acetyl neuraminic acid .
The gene encoding IFN-Omega 1 is located on chromosome 9, along with other type I interferon genes . However, only one of these genes is functional, giving rise to the IFN-Omega protein, while the others are non-functional pseudogenes . The IFN-Omega gene is not expressed in unstimulated cells; its expression is induced by viral infections .
IFN-Omega 1 exhibits about 75% sequence homology with IFN-Alpha and contains two conserved disulfide bonds that are crucial for its biological activities . It signals through the IFNAR-1/IFNAR-2 receptor complex, similar to other type I interferons, and exerts antiviral and antiproliferative activities . In quantitative terms, IFN-Omega is a significant component of human leukocyte interferon, contributing to its total antiviral activity .
The therapeutic potential of IFN-Omega 1 is still being explored. Its ability to modulate immune responses and inhibit viral replication makes it a promising candidate for treating various viral infections and possibly certain cancers . However, more research is needed to fully understand its physiological role and therapeutic applications .