Recombinant Human Interferon beta protein (IFNB1) (Active)

Shipped with Ice Packs
In Stock

Description

Mechanism of Action

IFNB1 binds to the heterodimeric IFNAR1-IFNAR2 receptor, activating JAK-STAT signaling:

  • Receptor Binding: Engages IFNAR1 (low affinity) and IFNAR2 (high affinity), inducing receptor dimerization .

  • Downstream Signaling:

    • Phosphorylates JAK1/TYK2 kinases, leading to STAT1/2 activation and nuclear translocation .

    • Induces >300 interferon-stimulated genes (ISGs), including antiviral mediators like 2'-5' oligoadenylate synthetase and protein kinase R .

    • Enhances MHC I expression, promoting CD8⁺ T-cell activation .

IFNB1 also exhibits JAK-STAT-independent pathways via IFNAR1 alone, regulating autophagy and dopamine turnover in neurons .

Biological Activities

  • Antiviral: Inhibits viral replication in HeLa cells (ED₅₀: 5–30 pg/mL) .

  • Antiproliferative: Suppresses tumor growth by inducing apoptosis (10× more potent than IFN-alpha) .

  • Immunomodulatory:

    • Reduces Th17 cell activity in multiple sclerosis (MS) .

    • Modulates B-cell development and myelopoiesis .

Clinical Use

  • Multiple Sclerosis: IFN-beta-1a (e.g., Rebif®) and IFN-beta-1b (e.g., Betaseron®) reduce relapse rates by 30% .

  • Oncology: Investigated for antitumor effects via STAT1-mediated apoptosis .

Research Tools

  • Functional Assays: Used in antiviral (e.g., encephalomyocarditis virus) and antiproliferative (TF-1 cell) assays .

  • Protein Interaction Studies: Binds IFNAR1 (UniProt: P17181) with Kd ~1 nM .

Production Challenges and Innovations

  • Expression Systems:

    • E. coli: High yield (~0.32 g/L) but requires refolding for soluble periplasmic secretion .

    • CHO/HEK293: Native glycosylation but lower productivity .

  • Stability Enhancements:

    • Hyperglycosylated mutants (e.g., R27T) reduce aggregation and immunogenicity .

    • Lyophilization in PBS/CHAPS buffers extends shelf life to 6 months at -80°C .

Key Research Findings

  • Glycosylation Impact: Deglycosylation reduces IFN-beta-1a activity by 90% due to precipitation .

  • Aggregation Risks: Soluble aggregates in IFN-beta-1b lower antiviral efficacy to 0.7 × 10⁷ IU/mg .

  • Neuronal Protection: Promotes α-synuclein clearance, suggesting therapeutic potential in Parkinson’s disease .

Future Directions

  • Engineered Variants: Hyperglycosylated or stabilized mutants to minimize neutralizing antibodies in MS therapy .

  • Broad-Spectrum Antivirals: Exploration against emerging viruses via ISG-mediated pathways .

Product Specs

Buffer
Lyophilized from a 0.2 µm filtered PBS, pH 7.4, containing 2% HSA and 3% mannitol.
Form
Lyophilized powder
Lead Time
Typically, we can ship the products within 5-10 business days after receiving your orders. Delivery timelines may vary depending on the purchasing method or location. For specific delivery information, please consult your local distributors.
Notes
Repeated freezing and thawing is not recommended. Store working aliquots at 4°C for up to one week.
Reconstitution
We recommend briefly centrifuging this vial prior to opening to ensure the contents settle at the bottom. Reconstitute the protein in deionized sterile water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquot for long-term storage at -20°C/-80°C. Our default final concentration of glycerol is 50% and can be used as a reference.
Shelf Life
Shelf life is influenced by various factors including storage conditions, buffer ingredients, temperature, and the inherent stability of the protein itself. Generally, the shelf life of liquid form is 6 months at -20°C/-80°C. The shelf life of lyophilized form is 12 months at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquoting is necessary for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag-Free
Synonyms
IFN-beta
Datasheet & Coa
Please contact us to get it.
Expression Region
22-187aa
Mol. Weight
20.0 kDa
Protein Length
Full Length of Mature Protein
Purity
>97% as determined by SDS-PAGE.
Research Area
Immunology
Source
E.coli
Species
Homo sapiens (Human)
Target Names
Uniprot No.

Target Background

Function
Exhibits antiviral, antibacterial, and anticancer properties.
Gene References Into Functions
  1. Data suggest a mechanism by which monocyte chemotactic protein-inducing protein 1 (MCPIP1) negatively regulates type I IFN interferon-beta antiviral defense. PMID: 29920243
  2. Studies have shown that HCMV has developed mutational robustness against IFN-beta by limiting the presence of APOBEC3G hot spots in essential open reading frames of its genome. PMID: 30045985
  3. RIG-I-like receptors play a role in the induction of interferon-beta1 in antiviral gene expression. PMID: 29098213
  4. Transcriptomic analysis of early untreated dermatomyositis muscles revealed that the main cluster of down-regulated genes was mitochondria-related. Histochemical, electron microscopy, and in situ oxygraphy analysis showed mitochondrial abnormalities, including increased reactive oxygen species (ROS) production and decreased respiration, which correlated with low exercise capacities and a type I IFN signature. PMID: 28623559
  5. Gas6 bound to the fiber proteins of adenovirus and suppressed IFN beta production. PMID: 29288958
  6. The overexpression of NPIPB3 restored the interferon-beta responses in severe acute respiratory syndrome coronavirus open reading frame 6 (SARS-CoV ORF6) expressing cells, indicating that the interaction of SARS CoV ORF6 and NPIPB3 reduced Type I interferon antagonism by SARS-CoV ORF6. PMID: 26320399
  7. The results demonstrate that cystatin B interferes with the STAT-1 signaling and IFN-beta-antiviral responses perpetuating HIV in macrophage reservoirs. PMID: 27137788
  8. This review briefly discusses the dysregulation of main T cell subpopulations in CNS autoimmunity and summarizes the T cell targeted effects of endogenous and exogenous IFN-beta in health and EAE/MS, with emphasis on the direct actions of IFN-beta on each T cell subset involved in the disease. PMID: 27033173
  9. c-Cbl negatively regulates IFN-beta signaling and cellular antiviral response by promoting IRF3 ubiquitination and degradation. PMID: 27503123
  10. YPEL5 silencing enhanced the induction of IFNB1 by pattern recognition receptors and phosphorylation of TBK1/IKBKE kinases, whereas co-immunoprecipitation experiments revealed that YPEL5 interacted physically with IKBKE. PMID: 27705791
  11. The effect of topical TREX1 knockdown and local interferon production on HIV transmission in human cervicovaginal explants and humanized mice, is reported. PMID: 27184854
  12. The current knowledge on IFNbeta from its structure, dynamic conformation, signaling pathway, and mechanism of action to its therapeutic effects is summarized. PMID: 27572132
  13. This study shows that the IFN-beta/STAT1 pathway is dysregulated in inflammatory bowel disease. PMID: 27220814
  14. Results suggest that, in addition to its well-known signaling activity, IFN-beta may be directly antimicrobial and be part of a growing family of cytokines and chemokines, called kinocidins, that also have antimicrobial properties. PMID: 28411186
  15. This study demonstrates a novel pathway for elevated IFNbeta signaling in SLE that is not dependent on stimulation by immune complexes but rather is cell intrinsic and critically mediated by IFNbeta and MAVS. PMID: 28471483
  16. G45R mutation of NS1 slightly decreased NS1 binding to dsRNA but did not interfere with its suppression of RIG-I-mediated type I IFN production. PMID: 27405392
  17. Upon influenza virus infection, DPF2 dysregulated IFN-beta induction and expression of cytokines/chemokines and antiviral proteins. This study provides evidence that influenza virus utilizes DPF2 to escape host innate immunity. PMID: 28404846
  18. Overexpression of PKV VP3 blocked IFN-beta-induced activation of the STAT1/STAT2/IRF9 promoter in a dose-dependent manner. PMID: 28441586
  19. Data suggest that EV71 infection in enterocytes does not inhibit phosphorylation of STAT1/2 induced by IFN-beta, but p-STAT1/2 transport into the nucleus is significantly blocked; EV71 infection in enterocytes down-regulates expression of KPNA1 and induces degradation of cellular KPNA1 via caspase-3. [EV17 = Enterovirus 71] PMID: 28455446
  20. This study reveals that decreased induction of both PKR and p300 proteins contribute to impaired induction of IFN-beta in Chronic obstructive pulmonary disease primary bronchial epithelial cells upon influenza infection. PMID: 26807508
  21. Together, these results suggest that RSV infection of MSCs alters their immune regulatory function by upregulating IFN-b and IDO, affecting immune cell proliferation, which may account for the lack of protective RSV immunity and for chronicity of RSV-associated lung diseases such as asthma and COPD. PMID: 27695127
  22. ArfGAP domain-containing protein 2 (ADAP2) is identified as a key novel scaffolding protein that integrates different modules of the RIG-I pathway, located at distinct subcellular locations, and mediates cellular antiviral type I interferon production. PMID: 27956705
  23. TBK1 complexes required for the phosphorylation of IRF3 and the production of interferon-beta have been identified. PMID: 28159912
  24. RNAs isolated from HCV-replicating cells triggered robust IFN-beta and IFN-lambda production through MDA5. PMID: 27655134
  25. These results suggest that inhibition of RIG-I-mediated type I interferon responses by Enterovirus 71 may contribute to the pathogenesis of viral infection. PMID: 27633794
  26. The interactions of IRF1, IFN-beta and IRF5 are involved in the M1 polarization of macrophages and have antitumor functions. PMID: 27176664
  27. The results demonstrate that PASylation has a positive impact on stability, solubility, and functional activity of IFN-beta1b and potentially might improve pharmacokinetic properties of the molecule as a therapeutic agent. PMID: 27833991
  28. Data suggest that interferon beta (IFN-beta) might be involved in modulating the expressions of interferon regulatory factor 1 (IRF1) and interferon regulatory factor-5 protein (IRF5) as well as maintaining the M1 polarization status and its function. PMID: 27363262
  29. Results suggest that serum levels of IFN-beta do not correlate well with the severity of dengue illness, but there is a clear association between changes in IFN-beta levels and the days of evolution during the acute phase of the disease. PMID: 26252251
  30. Overexpression of KLF4 inhibited virus-induced activation of ISRE and IFN-b promoter in cells, while knockdown of KLF4 potentiated viral infection-triggered induction of IFNB1 and downstream genes and attenuated viral replication. PMID: 25531393
  31. These findings suggest that RNF166 positively regulates RNA virus-triggered IFN-beta production by enhancing the ubiquitination of TRAF3 and TRAF6. PMID: 26456228
  32. The suppression of type I IFN production by HTLV-1 Tax through interaction with and inhibition of TBK1 kinase that phosphorylates IRF3. PMID: 26819312
  33. IFN-b could be an effective agent not only through its cell growth inhibitory effect on GSCs but also as a means of targeting the interconversion between GSCs and non-GSCs. PMID: 26397698
  34. These results demonstrate that poliovirus infection actively suppresses the host type I interferon response by blocking activation of IRF-3 and suggests that this is not mediated by cleavage of MDA-5 or IPS-1. PMID: 26437794
  35. The Hantaan virus A9 N protein can influence the host innate immune response by regulating the activation of IFNbeta. PMID: 26196448
  36. Together, the data demonstrate that interferon-beta inhibits foamy virus early in infection and that MxB is not a restriction factor of foamy virus. PMID: 26609934
  37. The C-terminal region of Toscana Virus NSs protein is critical for interferon-beta antagonism and protein stability. PMID: 26474372
  38. These findings support a model in which a measles virus defective interfering RNA is sensed by PACT and RIG-I to initiate an innate antiviral response via activation of interferon-beta production. PMID: 26608320
  39. Data suggest microRNA-302c regulates interferon-beta production in innate/mucosal immunity partly via down-regulation of interferon regulatory factors IRF3/7; studies were conducted in lung epithelial cells infected with influenza A virus H3N2. PMID: 26602079
  40. Data show that the MAPKKK6 ASK2, a modulator of MAPKKK5 ASK1 signaling, was essential for ASK1-dependent apoptosis, but not for inducing interferon-beta (IFNB) expression. PMID: 26243192
  41. Data suggest ADAP2 (an interferon-beta stimulated gene) disrupts pathogenic internalization of some RNA viruses/virions (dengue, vesicular stomatitis) by altering ADP ribosylation factor 6-mediated processes including up-regulation of pinocytosis. PMID: 26372645
  42. Data suggest that naturally occurring iDVGs (immunostimulatory defective viral genomes) trigger robust host antiviral/innate immunity responses including/requiring up-regulation of IFNB1 and IFNL1 (interferon lambda 1) in respiratory mucosa. PMID: 26336095
  43. Data identify an IFNbeta1-dependent, cell-autonomous mechanism that contributes to the therapeutic resistance of melanoma via the PKCepsilon-ATF2 regulatory axis. PMID: 25728676
  44. Early IFN-beta induction regulated virus replication in glioblastoma cells, whereas delayed IFN-beta induction resulted in efficient virus replication in neuroblastoma cells. PMID: 25920530
  45. Virion-associated R6 the type I IFN response by preventing efficient binding of IRF3/CBP complexes to the IFN-beta promoter in the context of infection. PMID: 25972548
  46. New IFN-beta response genes were identified in B cells, with possible implications to B cell-specific functions. PMID: 25025430
  47. Reduced myelin basic protein-induced CD4+ T-cell autoreactivity in interferon-beta-treated multiple sclerosis patients may be mediated by monocyte-derived interleukin-10. PMID: 25738751
  48. Influenza virus-infected cells respond with increased induction of interferon beta upon Staphylococcus aureus super-infection, however, subsequent interferon-stimulated gene expression are rather impaired due to a block of STAT1-STAT2 dimerization. PMID: 25293394
  49. Paracrine signaling increases the number of cells that express Ifnb1 over time, calibrating the immune response to viral infection. PMID: 25670204
  50. Data suggest that BRD4 (bromodomain containing 4) is essential for Toll-like receptor-stimulated interferon-beta (IFNB) gene transcription by permitting transcription factors to interact with the IFNB promoter in plasmacytoid dendritic cells. PMID: 25891802

Show More

Hide All

Database Links

HGNC: 5434

OMIM: 147640

KEGG: hsa:3456

STRING: 9606.ENSP00000369581

UniGene: Hs.93177

Protein Families
Alpha/beta interferon family
Subcellular Location
Secreted.

Q&A

What is the molecular structure and basic properties of recombinant human interferon beta protein?

Recombinant human interferon beta-1 (IFNB1) is a secreted glycoprotein that belongs to the Alpha/beta interferon protein family. The canonical protein has 187 amino acid residues with a molecular mass of approximately 22.3 kDa. It undergoes post-translational modifications, notably glycosylation, which can affect its biological activity .

IFNB1 is encoded by the IFNB1 gene located on chromosome 9 in humans. The protein plays a crucial role in adaptive immune responses and B cell differentiation. Alternative names include IFF, IFN-beta, IFNB, interferon beta, fibroblast interferon, and interferon-beta .

How does recombinant IFNB1 differ from native human interferon beta?

Recombinant IFNB1 produced in prokaryotic systems like E. coli often lacks the post-translational modifications (particularly glycosylation) found in native human interferon beta. When expressed in E. coli, the recombinant protein typically has a molecular weight of approximately 18 kDa (as seen in SDS-PAGE analysis), compared to the fully glycosylated native protein at 22.3 kDa .

What are the optimal conditions for expressing recombinant IFNB1 in E. coli systems?

Optimized conditions for high-yield expression of recombinant IFNB1 in E. coli BL21 (DE3) have been determined through response surface methodology based on central composite design (CCD). The following parameters represent optimal conditions:

ParameterOptimal Value
IPTG concentration0.7 mM
Induction start time (OD600 nm)0.58 (early exponential phase)
Post-induction time5 hours
Post-induction temperature37°C

These optimized conditions can yield approximately 40.2% of total cell protein as recombinant IFNB1 . It's important to note that induction at the early exponential phase rather than mid or late exponential phase results in significantly higher protein yields.

Why is periplasmic expression preferred for recombinant IFNB1 production in E. coli?

Periplasmic expression of recombinant IFNB1 in E. coli offers several advantages over cytoplasmic expression:

  • Human IFNB1 is toxic to bacterial hosts when accumulated in the cytoplasm, whereas periplasmic localization mitigates this toxicity .

  • The oxidizing environment of the periplasm facilitates proper disulfide bond formation, which is crucial for the correct folding and activity of IFNB1.

  • Periplasmic expression with leader sequences (such as pelB) allows for simpler extraction and purification procedures with fewer contaminating host proteins.

  • Studies have demonstrated that periplasmic expression systems using the pET-25b(+) vector with a pelB fusion tag can achieve yields of approximately 0.32 g/L of culture medium, which is superior to previous reports of cytoplasmic expression .

What strategies can overcome the toxicity of IFNB1 to bacterial expression hosts?

Several strategies have been developed to address the inherent toxicity of IFNB1 to bacterial expression hosts:

  • Periplasmic expression: Directing the recombinant IFNB1 to the periplasmic space using signal sequences like pelB significantly reduces toxicity to the host cell by sequestering the protein away from critical cellular machinery .

  • Controlled induction systems: Using tightly regulated promoters like T7 with IPTG induction allows precise timing of expression initiation. Optimizing induction conditions (starting at OD600 nm of 0.58) helps balance cell growth with protein production .

  • Codon optimization: Adapting the human IFNB1 gene sequence to E. coli codon usage preferences improves translation efficiency and reduces the metabolic burden on the host .

  • Host strain selection: E. coli BL21 (DE3) strains with reduced protease activity and enhanced ability to express toxic proteins can improve yields.

  • Fusion tags: N-terminal fusion partners can sometimes reduce toxicity while improving solubility and facilitating purification.

What are the most effective methods for detecting and quantifying recombinant IFNB1 protein?

Multiple analytical techniques have been validated for detecting and quantifying recombinant IFNB1:

  • Western Blot: The most widely used application for interferon beta 1 antibodies, providing specific detection based on molecular weight (~18 kDa for non-glycosylated recombinant form). Anti-β-IFN monoclonal antibodies (typically diluted 1:500 in TBS-T) provide high sensitivity and specificity .

  • Dot Blot: A simpler alternative to Western blot that doesn't require protein separation. Direct application of 1.5 μL extracted recombinant IFNB1 to nitrocellulose membranes, followed by antibody detection, provides a rapid qualitative assessment .

  • ELISA: Provides quantitative measurement with high sensitivity, particularly useful for detecting IFNB1 in complex mixtures or biological samples .

  • SDS-PAGE with densitometry analysis: Can estimate the percentage of IFNB1 relative to total cell protein (typically 35-40% under optimized conditions) .

  • Flow Cytometry: Useful for analyzing cellular responses to IFNB1 or detecting cell-surface bound protein .

How can researchers confirm the biological activity of recombinant IFNB1?

Confirming the biological activity of recombinant IFNB1 is essential for research applications. Multiple assay approaches include:

  • Antiviral activity assays: Measuring the ability of IFNB1 to protect cells against viral infection (e.g., vesicular stomatitis virus) is the gold standard for interferon activity. The activity is typically expressed in International Units (IU).

  • Reporter gene assays: Using cells transfected with an interferon-stimulated response element (ISRE) driving expression of a reporter gene (like luciferase) to measure IFNB1 signaling capacity.

  • STAT1 phosphorylation: Detecting phosphorylation of STAT1 by Western blot after treating cells with the recombinant IFNB1.

  • MHC upregulation assays: Measuring the ability of IFNB1 to increase MHC class I expression on target cells using flow cytometry.

  • Anti-proliferative assays: Quantifying the growth inhibition of specific cell lines (e.g., Daudi B cells) that are sensitive to interferon treatment.

How does the expression system affect post-translational modifications of recombinant IFNB1?

The choice of expression system significantly impacts the post-translational modifications (PTMs) of recombinant IFNB1, which in turn affects its biological properties:

  • E. coli expression systems: Produce non-glycosylated IFNB1 with a molecular weight of approximately 18 kDa. While the core biological activity is preserved, the protein typically has a shorter half-life in circulation and potentially altered receptor binding kinetics .

  • Mammalian expression systems (CHO, HEK293): Provide glycosylation patterns more similar to native human IFNB1, resulting in a protein of approximately 22.3 kDa. These systems produce IFNB1 with longer circulation half-life but at significantly higher production costs.

  • Yeast expression systems (P. pastoris, S. cerevisiae): Generate IFNB1 with glycosylation, but the pattern differs from human glycosylation (typically hyper-mannosylation), which can affect immunogenicity and bioactivity.

The impact of these differences should be carefully considered when designing experiments, as they may influence experimental outcomes, particularly in immunological studies or when translating findings to in vivo contexts.

What are the challenges in maintaining consistency between different batches of recombinant IFNB1?

Maintaining batch-to-batch consistency of recombinant IFNB1 preparations presents several challenges:

  • Expression level variability: Even with optimized protocols, slight variations in growth conditions, induction timing, or media composition can lead to different expression levels. Standardizing the OD600 nm at induction to precisely 0.58 and maintaining post-induction time at exactly 5 hours helps minimize this variability .

  • Protein folding and solubility: IFNB1 can form inclusion bodies or aggregate during expression, particularly at high expression levels. The percentage of soluble vs. insoluble protein may vary between batches, affecting yield and activity.

  • Purification consistency: Multi-step purification processes can introduce variability in final purity and recovery. Standardized protocols with quality control checkpoints help maintain consistency.

  • Endotoxin contamination: As a product expressed in Gram-negative bacteria, endotoxin levels must be monitored and controlled, particularly for cell-based assays where endotoxin can confound results.

  • Stability during storage: IFNB1 stability can vary based on buffer conditions, storage temperature, and freeze-thaw cycles, necessitating careful handling protocols to maintain activity across batches.

What strategies can resolve poor solubility or inclusion body formation during recombinant IFNB1 expression?

When encountering solubility issues or inclusion body formation with recombinant IFNB1 expression, researchers can implement several targeted strategies:

  • Optimize induction conditions: Lowering the induction temperature to 25-30°C, reducing IPTG concentration, or inducing at a higher cell density can increase the proportion of soluble protein.

  • Periplasmic targeting: Directing the protein to the periplasmic space using appropriate signal sequences (like pelB) has been shown to significantly improve IFNB1 solubility and reduce inclusion body formation .

  • Co-expression with chaperones: Co-expressing molecular chaperones (GroEL/GroES, DnaK/DnaJ/GrpE) can assist proper protein folding and reduce aggregation.

  • Fusion tags: N-terminal solubility enhancers like thioredoxin (Trx), glutathione S-transferase (GST), or maltose binding protein (MBP) can improve solubility, though they require subsequent tag removal.

  • Refolding strategies: If inclusion bodies are unavoidable, optimized denaturation and refolding protocols using gradual dialysis or dilution with redox pairs can recover active protein.

Each approach may require optimization for the specific expression construct and conditions, often necessitating empirical testing to determine the most effective solution.

How can researchers troubleshoot low biological activity of purified recombinant IFNB1?

When purified recombinant IFNB1 exhibits lower-than-expected biological activity, several methodical troubleshooting approaches should be considered:

  • Protein misfolding: Examine disulfide bond formation, as incorrect disulfide pairing can dramatically reduce activity. Consider using mild oxidation conditions during purification to promote proper disulfide formation.

  • Aggregation states: Check for protein aggregation using size exclusion chromatography or dynamic light scattering. Even small percentages of aggregates can significantly reduce specific activity.

  • Contaminants and inhibitors: Ensure high purity (>95%) and test for the presence of inhibitory contaminants that may co-purify with IFNB1.

  • Assay interference: Validate that components in the protein buffer (particularly detergents, reducing agents, or high salt) aren't interfering with activity assays.

  • Receptor binding capacity: Assess receptor binding directly using surface plasmon resonance or similar techniques to determine if the binding interface is properly structured.

  • Storage conditions: Test different storage conditions (buffer composition, pH, temperature) as IFNB1 activity can be sensitive to storage parameters.

What considerations are important when designing dose-response experiments with recombinant IFNB1?

Designing robust dose-response experiments with recombinant IFNB1 requires careful consideration of several factors:

  • Activity standardization: Calibrate recombinant IFNB1 preparations against international standards to express dosing in International Units (IU) rather than mass units, enabling comparison across different preparations.

  • Concentration range: Use a wide concentration range spanning at least 4-5 orders of magnitude (typically 0.01-1,000 IU/mL) with appropriate spacing between doses to capture both the threshold of response and saturation effects.

  • Cell type selection: Different cell types exhibit varying sensitivities to IFNB1. Established interferon-responsive cell lines (e.g., A549, HeLa, U937) provide more consistent responses than primary cells, which show donor-to-donor variability.

  • Temporal dynamics: IFNB1 responses evolve over time with early (1-4 hours), intermediate (8-24 hours), and late (24-72 hours) gene expression patterns. Design time-course experiments to capture the appropriate window for the biological effect of interest.

  • Biological replicates: Include sufficient biological replicates (minimum n=3) to account for inherent variability in IFNB1 responses.

  • Appropriate controls: Include both negative controls (buffer only) and positive controls (commercial IFNB1 with known activity) in each experiment.

How should researchers design experiments to investigate IFNB1 signaling pathway specificity?

When investigating IFNB1 signaling pathway specificity, researchers should consider these methodological approaches:

  • Receptor validation: Confirm the involvement of the IFNAR1/IFNAR2 receptor complex using neutralizing antibodies or receptor knockout/knockdown models. This distinguishes IFNB1-specific effects from potential contaminant-induced responses.

  • Pathway inhibitors: Use specific inhibitors for JAK-STAT (e.g., JAK inhibitors), MAPK (e.g., U0126), or PI3K (e.g., wortmannin) pathways to dissect which downstream cascades mediate observed effects.

  • Phosphoprotein analysis: Implement phospho-specific Western blots or phosphoproteomics to track activation of signaling intermediates (STAT1, STAT2, STAT3, ERK, PI3K) with temporal resolution.

  • Transcriptional profiling: Compare gene expression changes induced by IFNB1, interferon alpha, and interferon gamma to identify type I interferon-specific vs. interferon-common transcriptional responses.

  • Reporter constructs: Use pathway-specific reporter constructs (ISRE, GAS) to distinguish between different interferon-activated transcriptional programs.

  • Genetic validation: Employ cells with genetic deficiencies in specific pathway components to confirm their necessity in observed IFNB1 responses.

How does recombinant IFNB1 activity differ from other type I interferons in experimental systems?

Recombinant IFNB1 exhibits distinct biological activities compared to other type I interferons, particularly the various interferon alpha subtypes:

  • Receptor binding affinity: IFNB1 generally shows higher binding affinity for the IFNAR1/IFNAR2 receptor complex than most interferon alpha subtypes, resulting in more sustained signaling and different activation thresholds.

  • Antiviral potency: On a per-unit basis, IFNB1 typically demonstrates stronger antiviral activity against certain viruses (particularly in neuronal cells) compared to interferon alpha subtypes.

  • Immunomodulatory effects: IFNB1 tends to have stronger anti-inflammatory properties, with greater ability to suppress IL-1β, TNF-α, and IL-6 production by activated immune cells, while some interferon alpha subtypes show more pro-inflammatory characteristics.

  • Cell type specificity: Different tissues and cell types show variable responsiveness to IFNB1 versus interferon alpha subtypes, reflecting tissue-specific expression patterns of receptor components and downstream signaling modulators.

  • Gene induction profiles: While many interferon-stimulated genes are induced by both IFNB1 and interferon alpha subtypes, transcriptional profiling studies have identified distinct gene signatures, with some genes preferentially induced by IFNB1.

When designing comparative experiments, researchers should standardize different interferons based on specific activity (IU) rather than mass units to make meaningful comparisons.

What methodological approaches allow direct comparison between different interferon preparations?

To enable valid direct comparisons between different interferon preparations, researchers should implement these methodological approaches:

  • Standardization against reference materials: Calibrate all interferon preparations against international reference standards (e.g., from NIBSC or WHO) to express activity in International Units (IU), enabling normalized dosing across different preparations.

  • Bioactivity normalization: Perform preliminary dose-response experiments to determine EC50 values for each preparation in a standard antiviral protection assay, then use equi-active concentrations in comparative studies.

  • Receptor occupation assays: Quantify receptor binding parameters (kon, koff, KD) using surface plasmon resonance to normalize doses based on receptor occupancy rather than mass or nominal activity units.

  • Cross-platform validation: When comparing interferons across different experimental systems, include internal standards that are tested in all platforms to detect and correct for system-specific biases.

  • Time-course experiments: Different interferons may exhibit distinct temporal dynamics; therefore, measurements at multiple time points are essential for valid comparisons.

  • Statistical design: Implement factorial experimental designs that account for preparation, dose, time, and their interactions, followed by appropriate statistical analysis (typically ANOVA with post-hoc tests).

What are the emerging research areas for recombinant IFNB1 beyond its established applications?

Several emerging research areas are expanding the scientific understanding and application potential of recombinant IFNB1:

  • Cancer immunotherapy combinations: Investigating IFNB1 as an adjunct to immune checkpoint inhibitors (anti-PD-1/PD-L1) or CAR-T cell therapies to enhance tumor antigen presentation and immune cell recruitment.

  • Neuroinflammatory conditions: Exploring the neuroprotective mechanisms of IFNB1 beyond multiple sclerosis, including Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis models.

  • Targeted delivery systems: Developing cell-type or tissue-specific delivery mechanisms for IFNB1 to concentrate activity at disease sites while minimizing systemic effects.

  • Structural biology and protein engineering: Using advanced structural analysis to design IFNB1 variants with modified receptor binding properties, altered signaling bias, or enhanced stability.

  • Biomarker development: Identifying predictive biomarkers of IFNB1 response to enable personalized dosing and treatment strategies in clinical applications.

  • Host-microbiome interactions: Investigating how IFNB1 modulates the gut microbiome composition and how these changes feedback to influence immune responses and disease progression.

What technological advances are improving recombinant IFNB1 research quality and reproducibility?

Recent technological advances are significantly enhancing the quality and reproducibility of recombinant IFNB1 research:

  • CRISPR-Cas9 genome editing: Enabling precise genetic manipulation of IFNB1 signaling components in cellular models, creating cleaner experimental systems with fewer off-target effects than traditional RNA interference approaches.

  • Single-cell analysis technologies: Revealing cell-specific responses to IFNB1 that were previously masked in bulk population assays, providing insights into cellular heterogeneity in IFNB1 responsiveness.

  • Protein production advances: Improved expression systems with higher yields (40.2% of total cell protein) and more consistent post-translational modifications are increasing batch-to-batch reproducibility .

  • Automated high-throughput screening: Facilitating systematic testing of IFNB1 in combination with other agents across dose ranges and time courses, generating more comprehensive datasets.

  • Standardized reporting frameworks: Development of minimum information guidelines for interferon research, similar to MIAME for microarray experiments, improving methodological transparency and reproducibility.

  • Advanced computational modeling: Systems biology approaches that integrate multiple data types to predict IFNB1 network responses, generating testable hypotheses about complex interferon-regulated processes.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.