HEK293 cells are preferred for IFNW1 production due to their ability to perform human-like post-translational modifications. Key production parameters:
Yield: Optimized for high-density cultures, with stable production over 100 passages .
Purification: Proprietary chromatographic techniques ensure >95% purity .
Lyophilized IFNW1 remains stable at -80°C for 12 months; reconstituted protein is stable at 4°C for 2–7 days .
Carrier proteins (e.g., 0.1% HSA) prevent aggregation during long-term storage .
IFNW1 binds the IFN-α/β receptor (IFNAR1/IFNAR2) to activate JAK-STAT signaling, inducing interferon-stimulated genes (ISGs) like OAS1 and MX1 . Key findings:
ED<sub>50</sub>: <0.01 ng/mL (TF-1 cell cytotoxicity assay) vs. 2–8 pg/mL (vesicular stomatitis virus assay) .
Mechanism: Suppresses viral replication by upregulating ISGs, which inhibit viral RNA/protein synthesis .
IFNAR1-KO HEK293 Cells: CRISPR/Cas9 knockout of IFNAR1 in HEK293S cells blocked ISG activation (e.g., OAS1 mRNA reduced by 90%) and increased adenovirus titers 50-fold .
Reporter Assays: HEK-Blue IFN-α/β cells (STAT1/STAT2/IRF9 pathway) confirm IFNW1 specificity for type I IFN receptors, with no cross-reactivity to IFN-γ or IFN-λ .
Antiviral Therapeutics: IFNW1 reduces hepatitis C viral load in clinical trials .
Vaccine Production: IFNAR1-KO HEK293 cells enhance adenovirus yields by circumventing IFN-mediated antiviral responses .
Parameter | WT HEK293 | IFNAR1-KO HEK293 |
---|---|---|
Adenovirus Titer (Ad-IFNαγ) | 1.0 × 10<sup>7</sup> PFU/mL | 5.0 × 10<sup>8</sup> PFU/mL |
ISG Activation (OAS1) | 100% (baseline) | ≤10% |
IFN omega-1, IFN alpha-II-1, IFNW1.
HEK293 Cells.
LGCDLPQNHG LLSRNTLVLL HQMRRISPFL CLKDRRDFRF PQEMVKGSQL QKAHVMSVLH EMLQQIFSLF HTERSSAAWN MTLLDQLHTG LHQQLQHLET CLLQVVGEGE SAGAISSPAL TLRRYFQGIR VYLKEKKYSD CAWEVVRMEI MKSLFLSTNM QERLRSKDRD LGSSHHHHHH.
Recombinant IFNW1 Human is a single, glycosylated polypeptide chain spanning amino acids 22-195, containing a total of 180 amino acids with a molecular mass of 20.9 kDa. The protein is typically produced with a 6 amino acid histidine-tag at the C-terminus to facilitate purification through affinity chromatography. The full amino acid sequence is: LGCDLPQNHG LLSRNTLVLL HQMRRISPFL CLKDRRDFRF PQEMVKGSQL QKAHVMSVLH EMLQQIFSLF HTERSSAAWN MTLLDQLHTG LHQQLQHLET CLLQVVGEGE SAGAISSPAL TLRRYFQGIR VYLKEKKYSD CAWEVVRMEI MKSLFLSTNM QERLRSKDRD LGSSHHHHHH.
HEK293 cells provide several advantages for IFNW1 expression compared to bacterial or other mammalian systems. As a human-derived cell line, HEK293 cells accurately perform post-translational modifications, particularly glycosylation patterns that are essential for IFNW1's biological activity. Commercial preparations of IFNW1 from HEK293 cells typically achieve >95% purity with endotoxin levels below 1 EU/μg, making them suitable for sensitive biological assays. The use of HEK293 cells ensures proper protein folding and glycosylation, resulting in recombinant IFNW1 that closely resembles the native human protein.
Recombinant IFNW1 solution requires specific storage conditions to maintain stability and biological activity. For short-term usage (2-4 weeks), the protein can be stored at 4°C. For extended storage periods, it is recommended to store IFNW1 at -20°C with the addition of a carrier protein (such as BSA) to prevent protein adsorption to tube surfaces and stabilize the solution. Multiple freeze-thaw cycles should be avoided as they can lead to protein denaturation and loss of activity. When working with IFNW1, researchers should aliquot the stock solution immediately after reconstitution to minimize freeze-thaw cycles.
To effectively detect IFNW1-induced signaling, researchers should employ a multi-faceted approach:
Western Blotting: Monitor phosphorylation of STAT1, STAT2, and JAK1/TYK2 at 15-60 minutes post-stimulation.
qRT-PCR: Measure expression of interferon-stimulated genes (ISGs) including OAS1, Mx1, PKR, and IFITM1 after 4-24 hours of IFNW1 treatment.
Reporter Assays: Use cells transfected with ISRE-luciferase constructs to quantify transcriptional activation.
Immunofluorescence: Visualize nuclear translocation of STAT proteins following IFNW1 stimulation.
For optimal results, establish a dose-response curve (typically 0.1-1000 ng/mL) and time-course experiments. The activation of ISGs can be confirmed through comparison with wild-type cells versus IFNAR1 knockout controls, as IFNAR1-KO cells show significant inhibition of ISG induction when treated with interferons.
Generation of IFNAR1 knockout HEK293 cells provides an essential negative control for studying IFNW1 signaling specificity. The methodology involves:
CRISPR/Cas9 Design: Design sgRNAs targeting exon 2 of human IFNAR1 gene.
Transfection: Transfect HEK293 cells with CRISPR/Cas9 components and sgRNA plasmids.
Clone Selection: Isolate individual clones and expand them.
Validation Methods:
Genomic DNA sequencing to confirm editing
T7 endonuclease 1 (T7E1) assay to detect mutations (expected band sizes of 229 bp and 321 bp)
Western blotting to confirm absence of IFNAR1 protein (glycosylated IFNAR1 typically appears at 90-130 kDa)
Functional validation by treating cells with IFN-α and measuring ISG expression
Successful IFNAR1-KO cells will show constant (non-induced) expression of ISGs including OAS1, Mx1, PKR, and IFITM1 when treated with interferons, confirming receptor deletion.
For comprehensive assessment of IFNW1 purity and integrity, researchers should implement multiple complementary techniques:
SDS-PAGE: Run reduced and non-reduced samples to evaluate purity (>95% expected) and detect any protein aggregation or degradation products.
HPLC: Employ size-exclusion chromatography to quantitatively assess monomer percentage and detect higher-order structures.
Mass Spectrometry: Confirm protein mass (expected 20.9 kDa) and analyze post-translational modifications, particularly glycosylation patterns.
Circular Dichroism: Evaluate secondary structure integrity.
Endotoxin Testing: Use LAL assay to confirm endotoxin levels below 1 EU/μg, essential for cell-based assays.
Functional Assays: Test biological activity through reporter cell lines expressing ISRE-luciferase constructs.
Combined, these analyses provide a comprehensive profile of the recombinant protein's quality and functional status.
When facing inconsistent IFNW1 activity across experimental systems, consider the following systematic troubleshooting approach:
Receptor Expression Analysis: Quantify IFNAR1/IFNAR2 expression levels in target cells via flow cytometry or qRT-PCR, as receptor density significantly impacts sensitivity.
Negative Regulators Assessment: Measure baseline expression of USP18, SOCS1, and SOCS3, which can inhibit IFN signaling.
Cross-Validation: Compare results using multiple readout systems (e.g., ISRE-reporter assays, phospho-STAT Western blots, and qRT-PCR of ISGs).
Reference Standards: Include a well-characterized type I interferon (IFN-α2 or IFN-β) as positive control in parallel experiments.
Protein Quality Control: Verify IFNW1 integrity using SDS-PAGE and activity using a validated bioassay before each experimental series.
If disparities persist, consider cell type-specific factors, as different cell lineages exhibit varied responses to type I interferons due to differences in signal transduction components.
When designing experiments to study IFNW1 signaling, the following controls are essential:
Negative Controls:
Untreated cells (vehicle only)
IFNAR1-knockout cells (to confirm receptor specificity)
Heat-inactivated IFNW1 (to confirm specificity to the protein's native conformation)
Positive Controls:
Well-characterized type I interferons (IFN-α or IFN-β)
Known ISG inducers (e.g., poly(I:C))
Dose-Response Controls:
Titration series of IFNW1 (typically 0.1-1000 ng/mL)
Time-course measurements (15 min to 24h for different endpoints)
Biological Validation:
JAK inhibitors (e.g., Ruxolitinib) to confirm signaling pathway
siRNA knockdown of key signaling components
These controls help distinguish specific IFNW1 effects from background and non-specific responses, particularly important when using HEK293 cells which may have baseline interferon pathway activation.
IFNW1 offers valuable opportunities for comparative studies with other type I interferons due to its distinct receptor binding properties and downstream signaling characteristics. Researchers should consider:
Receptor Binding Affinity Analysis: Compare IFNW1 binding affinity to IFNAR1/IFNAR2 versus IFN-α and IFN-β using surface plasmon resonance (SPR) or biolayer interferometry.
Signaling Kinetics Comparison: Analyze phosphorylation kinetics and magnitude of STAT1/2, measuring both rapid responses (5-60 minutes) and sustained activation (2-24 hours).
Transcriptome Analysis: Perform RNA-Seq following treatment with equipotent doses of different type I IFNs to identify unique IFNW1-induced gene signatures.
Anti-Proliferative Activity: Compare growth inhibition profiles against various cell lines using MTT or similar viability assays.
Antiviral Activity Spectrum: Test protection against different viral challenges, particularly RNA viruses, using plaque reduction assays.
These comparative approaches can reveal unique properties of IFNW1 that distinguish it functionally from other members of the type I interferon family.
Despite advances in IFNW1 research, several critical knowledge gaps persist that merit further investigation:
Receptor Complex Dynamics: The specific stoichiometry and structural changes in the IFNAR1/IFNAR2 complex upon IFNW1 binding remain poorly characterized, requiring advanced techniques like cryo-EM or FRET-based approaches.
Signalosome Composition: The complete composition of IFNW1-specific signaling complexes, particularly non-canonical components beyond the JAK-STAT pathway, requires proteomic investigation.
Cell Type-Specific Responses: Systematic profiling of IFNW1 responses across diverse primary human cell types is lacking, particularly in immune and epithelial cells.
Regulatory Mechanisms: The specific negative regulators controlling IFNW1 signaling duration and magnitude need elucidation.
Physiological Role: The unique physiological functions of IFNW1 compared to other type I interferons remain unclear, requiring in vivo models with selective IFNW1 manipulation.
Addressing these gaps requires interdisciplinary approaches combining structural biology, systems biology, and in vivo models to fully understand IFNW1's unique properties in the interferon family.
To investigate interactions between IFNW1 and viral antagonists, researchers should implement a systematic multi-technique approach:
Co-Immunoprecipitation Studies: Express tagged viral proteins in HEK293 cells and perform pull-down assays with IFNW1 or components of its signaling pathway (JAK1, TYK2, STATs) to identify direct interactions.
Proximity Ligation Assays: Visualize and quantify IFNW1-viral protein interactions in situ within intact cells.
CRISPR Screens: Perform genome-wide CRISPR screens in virus-infected cells treated with IFNW1 to identify host factors required for viral antagonism.
Comparative Inhibition Assays: Compare the ability of viral antagonists to block signaling induced by IFNW1 versus other type I IFNs (IFN-α, IFN-β) through reporter assays and phospho-STAT measurements.
Structural Studies: Use hydrogen-deuterium exchange mass spectrometry or cryo-EM to characterize structural interfaces between viral antagonists and IFNW1 signaling components.
These approaches can reveal whether viruses have evolved specific mechanisms to counteract IFNW1 signaling and how these mechanisms might differ from those targeting other type I interferons.
Interferon-Omega 1 is a single, glycosylated polypeptide chain consisting of 180 amino acids, with a molecular mass of 20.9 kDa . It is produced using recombinant DNA technology in HEK293 cells, which are human embryonic kidney cells . The recombinant form of IFNW1 is fused to a 6 amino acid His-Tag at the C-terminus and is purified using proprietary chromatographic techniques .
IFNW1 interacts with specific cell surface receptors, triggering a cascade of events that lead to the activation of various antiviral genes and immune modulatory pathways . This interaction primarily involves the Janus kinase (JAK)-Signal Transducer and Activator of Transcription (STAT) signaling pathways . The activation of these pathways results in the transcription of interferon-stimulated genes (ISGs) that possess potent antiviral properties .
Research on IFNW1 aims to understand its structural intricacies, signaling pathways, and broader implications in immune regulation and disease . Scientists are exploring its potential applications in therapeutic interventions, particularly in the treatment of viral infections such as hepatitis . The recombinant form of IFNW1 is used in laboratory research to study its effects and potential therapeutic benefits .
The recombinant IFNW1 solution is typically stored at 4°C if it will be used within 2-4 weeks. For longer storage periods, it is recommended to freeze the solution at -20°C with the addition of a carrier protein to avoid multiple freeze-thaw cycles . The solution is formulated with 10% glycerol and phosphate-buffered saline (pH 7.4) to maintain its stability .