IFN b Mouse, His

IFN Beta Mouse Recombinant, His Tag
Shipped with Ice Packs
In Stock

Description

Molecular and Biochemical Characteristics

IFN-β Mouse, His is a 19.6 kDa protein expressed in mammalian systems (e.g., HEK 293 cells) to ensure proper post-translational modifications. The His tag facilitates affinity chromatography purification and immunodetection. Key specifications include:

ParameterDetails
Expression SystemHEK 293 cells
Purity≥80% (SDS-PAGE)
BioactivityEC₅₀ ~2.5 U/mL (L929 cell cytopathic inhibition assay)
FormulationCarrier-free, frozen in 20 mM HEPES, pH 6.0, with 0.5M NaCl and 6% glycerol
Storage-70°C or below; avoid freeze-thaw cycles

Antiviral and Antitumor Activity

IFN-β Mouse, His activates the canonical Jak-STAT pathway, inducing antiviral proteins (e.g., RNase L) and regulating apoptosis in infected or malignant cells . It demonstrates higher potency than IFN-α in suppressing tumor growth and viral replication .

Neuroprotection and Autoimmunity

Studies in IFN-β knockout (KO) mice reveal exacerbated experimental autoimmune encephalomyelitis (EAE), with chronic inflammation and microglial activation . IFN-β deficiency correlates with elevated TNF-α and IL-4 in the CNS, underscoring its role in dampening neuroinflammation .

Translational Regulation

The I-TOMCAT mouse model highlights IFN-β’s rapid induction post-viral infection and its suppression by RNase L in macrophages, illustrating a feedback loop to prevent excessive immune activation .

Comparative Analysis of Production Systems

FeatureE. coli Expression Mammalian Expression
GlycosylationNon-glycosylatedGlycosylated (authentic sequence)
Stability EnhancementsEngineered disulfide bond Native folding and modifications
ApplicationsIn vitro assays, structural studiesIn vivo studies, immunological assays
Endotoxin Levels<1 EU/μg <1 EU/μg

Disease Models

  • EAE and Multiple Sclerosis: IFN-β Mouse, His reduces relapse rates and CNS inflammation in EAE models, mimicking its therapeutic use in MS .

  • Viral Infections: Reporter mice (Δβ-luc) show tissue-specific IFN-β induction during influenza or La Crosse virus infections, aiding in tracking early antiviral responses .

Quantification Tools

  • ELISA Kits: Detect IFN-β in serum or tissue culture media with sensitivity down to 15.6 pg/mL .

Key Research Findings

  • Constitutive Expression: Thymic epithelial cells exhibit baseline IFN-β production, suggesting roles in immune homeostasis .

  • Therapeutic Potential: IFN-β suppresses Th17 responses and amplifies IL-10, offering pathways for treating autoimmune disorders .

  • Translational Blockade: RNase L inhibits IFN-β protein synthesis in macrophages, balancing antiviral defense and immunopathology .

References

  1. PBL Assay Science, Catalog 12400

  2. Abcam, Product ab316395

  3. Journal of Immunology (2024), I-TOMCAT model

  4. Journal of Immunology (2003), EAE in IFN-β KO mice

  5. Journal of Immunology (2009), Δβ-luc reporter

Product Specs

Introduction
IFN-beta 1b exhibits antiviral, antibacterial, and anticancer properties. Influenza A viruses not only hinder IFN-beta gene induction but also suppress Type-I IFN signaling by inducing the SOCS-3 protein. Intracellular bacteria and cytosolic poly (dA-dT) activate IFN-beta responses in various human cells without the need for human ZBP1.
Description
Recombinant Mouse IFN beta, produced in E. coli, is a single, non-glycosylated polypeptide chain comprising 182 amino acids (22-182 a.a.) with a molecular weight of 22 kDa. A 21 amino acid sequence is fused to the N-terminus of Mouse IFN beta, and the protein is purified using proprietary chromatographic methods.
Physical Appearance
A clear, sterile solution without any color.
Formulation
The IFN-Beta solution (0.25mg/ml) is formulated in a buffer containing 20mM HEPES (pH 6.0), 0.5M NaCl, and 10% glycerol.
Stability
For short-term storage (2-4 weeks), keep at 4°C. For extended periods, store frozen at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freezing and thawing.
Purity
Purity exceeds 95% as determined by SDS-PAGE analysis.
Synonyms

Leukocyte IFN, B cell IFN, Type I IFN, IFNB1, IFB, IFF, IFNB, IFN-b 1b, MGC96956.

Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MINYKQLQLQ ERTNIRKCQE LLEQLNGKIN LTYRADFKIP MEMTEKMQKS YTAFAIQEML QNVFLVFRNN FSSTGWNETI VVRLLDELHQ QTVFLKTVLE EKQEERLTWE MSSTALHLKS YYWRVQRYLK LMKYNSYAWM VVRAEIFRNF LIIRRLTRNF QN.

Q&A

What is mouse IFN-β and how does it function in the immune system?

Mouse IFN-β is a Type I interferon cytokine that plays a key role in the innate immune response to infection, developing tumors, and other inflammatory stimuli. It functions by binding to the heterodimeric IFNα/β receptor (IFNAR) composed of high-affinity (IFNAR2) and low-affinity (IFNAR1) subunits. This interaction activates the canonical JAK-STAT signaling pathway, resulting in transcriptional activation or repression of interferon-regulated genes. These genes encode various effectors including antiviral proteins, regulators of cell proliferation and differentiation, and immunoregulatory proteins that collectively mediate the interferon response . IFN-β has been well-established as a mainstay therapy for relapse-remitting multiple sclerosis (MS), where it functions both as an immunomodulator and potentially as a neuroprotective agent .

What are the structural characteristics of recombinant mouse IFN-β proteins?

Recombinant mouse IFN-β is available in various forms, with the following structural characteristics:

PropertyE. coli-expressedHEK293-expressed (His-tagged)
Molecular Weight19.6 kDaSimilar to native (~20 kDa)
Formulation20 mM HEPES, pH 6.0, 0.5M NaCl, 6% GlycerolVaries by manufacturer
Purity>95% by SDS-PAGE≥80%
ModificationsEngineered disulfide bond for increased activityHis-tag for purification/detection
Expression SystemBacterial (E. coli)Mammalian (HEK293)

The E. coli-expressed variant has been modified as described by Day et al. (1992) with an engineered disulfide bond that greatly increases specific activity of the recombinant protein . The His-tagged version expressed in mammalian cells offers potentially different post-translational modifications that may better represent the native protein structure .

How should mouse IFN-β be stored to maintain optimal bioactivity?

For retention of full bioactivity, mouse IFN-β should be stored at -70°C or below. It is critical to avoid repeated freeze/thaw cycles as these can significantly degrade the protein and reduce its biological activity. When working with the protein, it is recommended to thaw aliquots quickly and keep the protein on ice while preparing experiments . For long-term storage planning, it is advisable to prepare single-use aliquots immediately upon receiving the protein to minimize potential degradation from repeated thawing and freezing. Researchers should also monitor the protein's activity periodically if stored for extended periods to ensure experimental reproducibility.

How is the bioactivity of mouse IFN-β accurately measured in research settings?

The standard method for measuring mouse IFN-β bioactivity is through a cytopathic effect inhibition assay using mouse L929 cells challenged with encephalomyocarditis virus (EMCV). In this assay, the EC50 for IFN-β is approximately 2.5 U/ml . The methodology involves:

  • Culture L929 cells in appropriate growth medium to 80% confluence

  • Prepare serial dilutions of the IFN-β sample and reference standard

  • Add diluted samples to the L929 cells and incubate for 24 hours

  • Challenge with EMCV at appropriate multiplicity of infection

  • Incubate for an additional 24-48 hours

  • Assess cell viability using MTT or similar viability assay

  • Calculate the protective effects of IFN-β by comparing to the reference standard

Researchers should include appropriate controls including untreated cells, virus-only controls, and reference standard curves to ensure accurate determination of bioactivity. This functional assay is preferred over simple binding assays as it confirms the biological activity of the protein.

What experimental considerations are important when designing in vivo studies with mouse IFN-β?

When designing in vivo studies with mouse IFN-β, researchers should consider several critical factors:

  • Formulation Selection: For in vivo injection studies, carrier-free formulations are strongly recommended to minimize possible immunogenic reactions by the host animal . E. coli-expressed recombinant proteins may contain trace endotoxins that can confound experimental results.

  • Dosage Optimization: Effective doses observed in research range from 100 pg/ml to 10 ng/ml, with 1 ng/ml and 10 ng/ml showing significant biological effects in neural progenitor cell research . Pilot dose-response studies should be conducted for each specific application.

  • Administration Route: The route of administration (intraperitoneal, intravenous, subcutaneous, or intracerebroventricular) can significantly affect the biodistribution, half-life, and ultimate activity of IFN-β.

  • Temporal Considerations: The timing of IFN-β administration relative to disease onset or experimental intervention is critical. Recent research indicates that IFN-β's effects on apoptosis and immune modulation may be time-dependent.

  • Control Groups: Appropriate vehicle controls should match the IFN-β formulation buffer exactly to account for potential effects of components like HEPES or glycerol.

  • Readout Selection: Multiple readouts should be employed to comprehensively assess IFN-β effects, including both molecular markers (STAT phosphorylation, downstream gene expression) and functional outcomes.

How can researchers optimize detection of His-tagged mouse IFN-β in experimental samples?

Optimizing detection of His-tagged mouse IFN-β requires selecting appropriate methodologies based on the experimental context:

  • Western Blotting:

    • Use anti-His antibodies for direct detection of the tagged protein

    • Alternatively, use specific anti-mouse IFN-β antibodies for confirmation

    • Include positive controls with known concentrations of recombinant protein

    • Optimize blocking conditions to prevent non-specific binding

    • Consider using HRP-conjugated anti-His antibodies to eliminate secondary antibody steps

  • ELISA:

    • Sandwich ELISA using anti-His capture antibody and anti-IFN-β detection antibody

    • Direct ELISA using plate-bound anti-IFN-β antibody and anti-His detection

    • Standard curves should be prepared using the same recombinant protein

    • Sample dilution series help identify optimal detection range

  • Immunoprecipitation:

    • Nickel-NTA or cobalt-based resins can pull down His-tagged proteins

    • Magnetic beads conjugated with anti-His antibodies offer an alternative approach

    • Stringent washing conditions are required to reduce background

  • Mass Spectrometry:

    • For complex mixtures, enrichment using metal affinity chromatography before MS analysis

    • Tryptic digestion and peptide mapping can verify both the His-tag and IFN-β sequence

How does mouse IFN-β activate the JAK-STAT pathway and what downstream effects occur?

Mouse IFN-β activates the JAK-STAT pathway through a coordinated sequence of molecular events:

  • Receptor Binding: IFN-β binds to the IFNAR1-IFNAR2 heterodimeric receptor complex . This binding event initiates conformational changes in the receptor.

  • JAK Activation: The receptor-associated Janus kinases (JAKs), primarily JAK1 and TYK2, are activated through trans-phosphorylation.

  • STAT Recruitment and Phosphorylation: The activated JAKs phosphorylate tyrosine residues on the receptor's cytoplasmic domain, creating docking sites for STAT proteins. STAT1 and STAT2 are recruited and phosphorylated.

  • STAT Dimerization and Nuclear Translocation: Phosphorylated STAT1 and STAT2 form heterodimers that associate with IRF9 to form the ISGF3 complex, which translocates to the nucleus.

  • Transcriptional Regulation: The ISGF3 complex binds to interferon-stimulated response elements (ISREs) in the promoters of interferon-stimulated genes (ISGs) .

Key downstream effects include:

  • Upregulation of antiviral proteins

  • Modulation of apoptotic pathways (evidenced by decreased apoptosis in neural progenitor cells)

  • Increased expression of GFRA2, NOD1, Caspases 1 and 12, and TNFSF10

  • Activation of immunoregulatory pathways that modulate inflammatory responses

This signaling cascade represents a key mechanism by which IFN-β exerts its biological effects across multiple cell types and experimental systems.

What is the effect of mouse IFN-β on neural progenitor cell survival and differentiation?

Research demonstrates that mouse IFN-β has significant effects on neural progenitor cell (NPC) survival but not on differentiation:

Effects on Survival:

  • Mouse NPCs express high levels of IFNAR, making them responsive to IFN-β treatment

  • IFN-β treatment (1-10 ng/ml) significantly decreases apoptosis in NPCs upon growth factor withdrawal

  • At 1 ng/ml concentration, IFN-β reduced apoptosis by approximately 23% compared to control conditions

  • The anti-apoptotic effect appears to be mediated through the JAK/STAT pathway activation

Effects on Differentiation:

  • IFN-β treatment showed no effect on NPC differentiation into neuronal lineages, as evidenced by unchanged expression of β-tubulinIII and doublecortin markers

  • Similarly, no changes were observed in oligodendrocyte markers NG2 and PDGFRα following IFN-β treatment

  • Astrocyte markers GLAST and GFAP also remained unchanged after IFN-β treatment

These findings suggest that IFN-β plays a neuroprotective role by promoting NPC survival without altering cell fate determination or differentiation pathways. This selective effect on survival may contribute to the therapeutic benefits observed in multiple sclerosis treatment, where IFN-β may help maintain the endogenous NPC pool without disrupting normal developmental processes.

How does IFN-β contribute to neuroprotection in mouse models of neuroinflammatory disease?

Mouse IFN-β contributes to neuroprotection in neuroinflammatory disease models through multiple mechanisms:

  • Direct Anti-apoptotic Effects: IFN-β directly reduces apoptosis in neural progenitor cells (NPCs) through activation of survival pathways, as demonstrated by decreased TUNEL-positive cells following IFN-β treatment . This protective effect is most pronounced at concentrations of 1-10 ng/ml.

  • STAT Pathway Activation: IFN-β treatment upregulates STAT1 and STAT2 signaling in NPCs, which leads to transcription of anti-apoptotic genes and activation of the NF-κB pathway that promotes cell survival .

  • Modulation of Neurotrophic Factors: IFN-β treatment upregulates GFRA2 (Glial Cell Line-Derived Neurotrophic Factor Family Receptor Alpha 2), which may enhance responsiveness to neurotrophic factors that support neuronal survival .

  • Immunomodulatory Effects: Beyond direct cellular effects, IFN-β modulates the immune response by:

    • Downregulating MHC class II expression on antigen-presenting cells

    • Decreasing T-cell activation and CNS infiltration

    • Reducing inflammatory cytokine production

  • Caspase Regulation: IFN-β treatment affects caspase expression, particularly Caspases 1 and 12, which may participate in membrane repair and cell survival mechanisms .

These multifaceted neuroprotective mechanisms likely contribute to IFN-β's therapeutic efficacy in multiple sclerosis, where it not only modulates peripheral immune responses but also directly enhances the survival capacity of CNS cells.

How can mouse IFN-β be used to study JAK-STAT pathway homeostasis in immune cells?

Mouse IFN-β provides an excellent tool for studying JAK-STAT pathway homeostasis in immune cells, as highlighted by recent research showing this pathway's importance in maintaining T cell and macrophage homeostasis . Researchers can implement the following experimental approaches:

  • Temporal Activation Studies:

    • Treat cells with IFN-β at different time points (5min, 15min, 30min, 1hr, 4hr, 24hr)

    • Measure phosphorylation kinetics of JAK1, TYK2, STAT1, and STAT2 by western blot or phospho-flow cytometry

    • Evaluate nuclear translocation of STAT complexes using cellular fractionation or imaging

  • Dose-Response Relationships:

    • Apply concentration gradients (100pg/ml to 100ng/ml) to identify threshold effects

    • Correlate STAT activation levels with functional outcomes (gene expression, cell survival)

    • Generate mathematical models of pathway sensitivity and adaptation

  • Feedback Regulation Analysis:

    • Monitor expression of negative regulators (SOCS, PIAS proteins) following IFN-β stimulation

    • Use knockdown/knockout approaches for these regulators to assess their contribution to pathway homeostasis

    • Measure pathway refractory periods after initial stimulation

  • Cell-Type Specificity:

    • Compare JAK-STAT responses between different immune cell populations (T cells vs. macrophages)

    • Assess how cell differentiation state affects pathway responsiveness

    • Identify cell-type specific downstream targets using RNA-seq

  • Pathway Crosstalk Examination:

    • Simultaneously activate multiple pathways (e.g., IFN-β + IL-6 or TNF-α)

    • Determine how concurrent signaling modifies JAK-STAT activation patterns

    • Map signaling node interactions through phosphoproteomic approaches

These approaches can provide insights into how the JAK-STAT pathway maintains cellular homeostasis and how its dysregulation may contribute to immune pathologies.

What are the considerations when using mouse IFN-β to investigate tumor microenvironment modulation?

Recent research has highlighted IFN-β's role in controlling tumor-associated macrophage recruitment and modulating antitumor immunity . When designing experiments to investigate these effects, researchers should consider:

  • Tumor Model Selection:

    • Choose models with defined macrophage involvement (e.g., breast cancer, melanoma)

    • Consider both orthotopic and subcutaneous models for comparison

    • Genetically engineered mouse models may provide more physiological contexts than xenografts

  • Delivery Method Optimization:

    • Local vs. systemic administration affects IFN-β distribution in the tumor microenvironment

    • Consider using engineered slow-release formulations for sustained exposure

    • Cell-specific targeting approaches (nanoparticles, antibody conjugates) may enhance specificity

  • Timing and Duration:

    • Apply IFN-β at different tumor stages (initiation, established, advanced)

    • Evaluate acute versus chronic exposure effects

    • Implement pulse treatment regimens to assess adaptive responses

  • Comprehensive Immune Profiling:

    • Monitor changes in tumor-associated macrophage phenotypes (M1 vs. M2)

    • Assess infiltrating T cell subsets and activation status

    • Evaluate dendritic cell maturation and antigen presentation capacity

    • Analyze NK cell recruitment and cytotoxic activity

  • Mechanistic Dissection:

    • Use IFNAR knockout mice or conditional knockouts to identify cell-specific requirements

    • Implement JAK/STAT inhibitors to block specific pathway components

    • Consider combinatorial approaches with checkpoint inhibitors or chemotherapy

  • Multi-dimensional Readouts:

    • Integrate spatial transcriptomics, multi-parameter flow cytometry, and imaging

    • Monitor both tumor growth kinetics and metastatic potential

    • Evaluate long-term memory responses after treatment cessation

These considerations will help researchers develop robust experimental designs to investigate how IFN-β shapes the tumor microenvironment and potentially enhances anti-tumor immunity.

How can differential responses to E. coli-expressed versus mammalian-expressed mouse IFN-β be experimentally addressed?

Differential responses to E. coli-expressed versus mammalian-expressed mouse IFN-β can arise from structural and post-translational differences. A systematic experimental approach to address these differences includes:

  • Structural Comparison Analysis:

    • Perform circular dichroism spectroscopy to compare secondary structure profiles

    • Use differential scanning calorimetry to assess thermal stability differences

    • Implement native mass spectrometry to analyze conformational states

  • Post-translational Modification Characterization:

    • Use glycan-specific staining and mass spectrometry to identify glycosylation patterns in mammalian-expressed IFN-β

    • Assess phosphorylation and other modifications using phosphoproteomic approaches

    • Map disulfide bond patterns through non-reducing SDS-PAGE and mass spectrometry

  • Receptor Binding Kinetics:

    • Compare binding affinities to IFNAR1 and IFNAR2 using surface plasmon resonance

    • Analyze association and dissociation rates to identify kinetic differences

    • Perform competitive binding assays with labeled IFN-β variants

  • Signaling Pathway Activation:

    • Compare dose-response curves for STAT1/2 phosphorylation

    • Assess temporal differences in signaling duration and magnitude

    • Evaluate activation of alternative signaling pathways beyond canonical JAK-STAT

  • Functional Outcomes Comparison:

    • Compare anti-viral potency in standard protection assays

    • Assess anti-proliferative effects across multiple cell types

    • Evaluate immunomodulatory activities in primary immune cells

    • Compare anti-apoptotic effects in neural progenitor cells using the established growth factor withdrawal model

  • In Vivo Pharmacokinetics and Distribution:

    • Compare half-lives and tissue distribution using labeled proteins

    • Assess immunogenicity profiles through antibody development monitoring

    • Evaluate efficacy in disease models such as experimental autoimmune encephalomyelitis

This systematic approach will help identify critical differences that may impact experimental outcomes and therapeutic applications of different mouse IFN-β preparations.

What are common challenges when working with mouse IFN-β and how can they be addressed?

Researchers frequently encounter several challenges when working with mouse IFN-β. Here are systematic approaches to address these issues:

  • Protein Stability and Activity Loss:

    • Problem: IFN-β activity decreases with storage or handling.

    • Solution: Store at -70°C or below , prepare single-use aliquots, avoid freeze/thaw cycles, and add carrier proteins (BSA) to dilute solutions to prevent adsorption to surfaces.

  • Batch-to-Batch Variability:

    • Problem: Different lots show inconsistent bioactivity.

    • Solution: Perform rigorous bioactivity testing (cytopathic effect inhibition assay ) for each new lot, normalize doses by bioactivity rather than protein concentration, and maintain reference standards.

  • Endotoxin Contamination:

    • Problem: E. coli-expressed proteins may contain endotoxins.

    • Solution: Verify endotoxin levels (<1 EU/μg ), use carrier-free formulations for in vivo studies, and include appropriate controls to distinguish IFN-β effects from endotoxin effects.

  • Poor Detection Sensitivity:

    • Problem: Difficulty detecting low concentrations of IFN-β.

    • Solution: Optimize antibody pairs for ELISA, use biotin-streptavidin amplification, implement more sensitive readout systems, and consider concentrating samples using immunoprecipitation.

  • Complex Downstream Effects Interpretation:

    • Problem: Distinguishing direct effects from secondary responses.

    • Solution: Include time-course analyses, use JAK-STAT inhibitors as controls, implement IFNAR knockdown/knockout controls, and use multi-omics approaches to map response networks.

  • In Vivo Delivery Challenges:

    • Problem: Short half-life and poor tissue penetration.

    • Solution: Optimize delivery vehicles, consider PEGylation to extend half-life, implement local delivery methods for CNS applications, and use imaging techniques to track distribution.

  • Experimental Reproducibility:

    • Problem: Variable results between experiments.

    • Solution: Standardize experimental protocols, control cell density and passage number, verify receptor expression levels, and implement robust statistical approaches for data analysis.

How can researchers optimize mouse IFN-β treatment protocols for studies involving neural progenitor cells?

Based on published research findings , optimizing mouse IFN-β treatment protocols for neural progenitor cell (NPC) studies requires careful consideration of multiple parameters:

  • Concentration Optimization:

    • Research indicates that 1-10 ng/ml IFN-β provides optimal anti-apoptotic effects

    • 1 ng/ml concentration reduced apoptosis by approximately 23%

    • Higher concentrations (100 ng/ml) showed reduced efficacy, suggesting a bell-shaped dose-response curve

    • Recommendation: Perform pilot dose-response studies spanning 100 pg/ml to 100 ng/ml for each specific NPC population

  • Timing Considerations:

    • Administer IFN-β simultaneously with growth factor withdrawal for maximum protective effect

    • Consider pre-treatment (6-24 hours) to allow for transcriptional changes before challenge

    • For long-term studies, implement periodic re-treatment as receptor desensitization may occur

  • Culture Condition Standardization:

    • Maintain consistent cell density (typical recommendations: 50,000-100,000 cells/cm²)

    • Standardize NPC passage number (preferably passages 2-5)

    • Verify IFNAR expression using RT-qPCR and immunocytochemistry before experiments

    • Control for variables like oxygen tension, pH, and media composition

  • Endpoint Selection:

    • TUNEL assay has been validated for apoptosis assessment following IFN-β treatment

    • Complement with other methodologies (Annexin V staining, caspase activity assays)

    • Include molecular readouts (STAT phosphorylation, target gene expression)

    • Consider functional outcomes for long-term studies (neurosphere formation capacity)

  • Growth Factor Withdrawal Protocol:

    • Complete withdrawal of both EGF and FGF is recommended for studying anti-apoptotic effects

    • Gradual reduction may better model physiological stress

    • Include positive controls (known anti-apoptotic factors like BDNF)

  • Verification of Mechanism:

    • Include JAK inhibitors (e.g., ruxolitinib) to confirm pathway dependence

    • Measure STAT1/2 phosphorylation to verify receptor activation

    • Analyze expression of identified target genes (GFRA2, NOD1, Caspases 1/12, TNFSF10)

Implementation of these optimized protocols will enhance reproducibility and maximize the reliability of data generated in NPC studies using mouse IFN-β.

What controls and validation steps are essential when studying signaling pathways activated by mouse IFN-β?

When studying signaling pathways activated by mouse IFN-β, several essential controls and validation steps ensure experimental rigor and reproducibility:

  • Receptor Expression Verification:

    • Confirm IFNAR expression in target cells before experiments using:

      • RT-qPCR for mRNA levels

      • Flow cytometry or immunocytochemistry for surface expression

      • Western blotting for total receptor protein

    • Include receptor-negative cells as controls

  • Pathway Component Controls:

    • Positive controls:

      • Include known IFN-β responsive cell lines (L929 cells)

      • Use alternative STAT pathway activators (IL-6, IFN-γ) as comparative controls

    • Negative controls:

      • Include JAK inhibitors (ruxolitinib, tofacitinib)

      • Implement STAT1/2 knockdown/knockout systems

      • Use IFNAR blocking antibodies to confirm receptor specificity

  • Signal Validation Approaches:

    • Multi-method verification:

      • Western blotting for phosphorylated STATs

      • Immunocytochemistry for STAT nuclear translocation

      • Reporter assays (ISRE-luciferase) for transcriptional activation

      • ChIP assays to confirm STAT binding to target promoters

  • Temporal Resolution:

    • Implement detailed time-course analysis:

      • Early timepoints (5, 15, 30 min) for activation kinetics

      • Intermediate timepoints (1-4 hours) for primary transcriptional responses

      • Late timepoints (24-72 hours) for secondary responses and pathway resolution

    • Monitor both pathway activation and deactivation dynamics

  • Dose-Response Validation:

    • Generate complete dose-response curves (10 pg/ml to 1 μg/ml)

    • Calculate EC50 values for different readouts

    • Identify potential biphasic responses that may indicate pathway switching

  • Downstream Target Verification:

    • Validate key target genes using multiple approaches:

      • RT-qPCR for mRNA expression

      • Western blotting for protein levels

      • Functional assays specific to the target's role

    • Implement siRNA knockdown of key targets to confirm their role in observed phenotypes

  • Cross-Pathway Interaction Controls:

    • Pre-treat cells with inhibitors of parallel pathways (MAPK, PI3K)

    • Assess pathway activation in cells with genetic modifications in complementary pathways

    • Perform co-immunoprecipitation experiments to identify physical interactions between pathway components

How is mouse IFN-β being used to study mechanisms of mRNA vaccine-induced immunity?

Recent research has revealed critical insights into how mouse IFN-β contributes to mRNA vaccine-induced immunity. A 2024 study published in Nature Communications demonstrated that innate immune responses against mRNA vaccines promote cellular immunity through IFN-β at the injection site . Based on this and related research, several key experimental approaches are being implemented:

  • Local Immune Response Characterization:

    • Site-specific analysis of IFN-β production following mRNA vaccination

    • Temporal mapping of IFNAR signaling activation in different cell populations

    • Spatial transcriptomics to identify IFN-β-responsive cells in proximity to vaccine administration

  • Cell-Specific Contribution Analysis:

    • Conditional IFNAR knockout models to determine cell type-specific requirements

    • Bone marrow chimeras to distinguish hematopoietic versus non-hematopoietic contributions

    • Single-cell RNA sequencing to identify transcriptional signatures in responding populations

  • Mechanistic Dissection Approaches:

    • RNA modifications to modulate IFN-β induction by mRNA vaccines

    • Lipid nanoparticle formulation optimization to target specific IFN-β-producing cells

    • Combined blockade of IFN-β and other innate cytokines to map pathway redundancy

  • Translational Relevance Assessment:

    • Correlation of IFN-β levels with antibody titer development

    • Analysis of T cell priming efficiency in presence/absence of IFN-β signaling

    • Evaluation of memory cell formation and long-term protective immunity

This research direction represents a critical frontier in understanding how mRNA vaccines initiate effective adaptive immune responses and how modulation of the IFN-β pathway might enhance vaccine efficacy or reduce adverse effects.

What role does mouse IFN-β play in the termination of STING responses and how can this be experimentally investigated?

Recent research has demonstrated that termination of STING (Stimulator of Interferon Genes) responses is mediated via ESCRT-dependent degradation, with IFN-β playing a significant regulatory role in this process . This emerging area can be experimentally investigated through:

  • Temporal Relationship Characterization:

    • Time-course experiments measuring STING pathway activation followed by IFN-β production

    • Live-cell imaging using fluorescent reporters for both STING activation and IFN-β expression

    • Pulse-chase experiments to track STING protein degradation kinetics following IFN-β treatment

  • ESCRT Machinery Interaction Studies:

    • Co-immunoprecipitation experiments to identify interactions between STING, ESCRT components, and IFN-β-induced factors

    • CRISPR screening for ESCRT pathway components that influence STING degradation

    • Microscopy-based colocalization analysis of STING with ESCRT machinery following IFN-β exposure

  • Mechanistic Dissection Approaches:

    • Compare STING degradation kinetics in wild-type versus IFNAR knockout cells

    • Implement ubiquitination site mapping before and after IFN-β treatment

    • Use reconstitution systems with mutant STING proteins resistant to ESCRT-mediated degradation

  • Feedback Loop Analysis:

    • Measure how STING-dependent IFN-β production subsequently affects STING stability

    • Assess how IFN-β-mediated STING degradation affects secondary responses to STING agonists

    • Develop mathematical models of this feedback system

  • Physiological Relevance Assessment:

    • Compare viral infection outcomes in systems with normal versus disrupted STING degradation

    • Evaluate autoimmune phenotypes when STING degradation is impaired

    • Assess therapeutic potential of modulating this pathway in inflammatory conditions

This research direction provides important insights into the resolution phase of innate immune responses and how dysregulation of this process might contribute to persistent inflammation or autoimmunity.

How might mouse IFN-β research inform development of next-generation therapeutic approaches for neuroinflammatory disorders?

Current research on mouse IFN-β is driving several innovative therapeutic strategies for neuroinflammatory disorders:

  • Cell Type-Specific Targeting Approaches:

    • Neural progenitor cell-directed IFN-β delivery systems

    • Development of IFNAR agonists with enhanced CNS penetration

    • Cell-selective activation strategies to minimize systemic effects

  • Pathway-Selective Modulation:

    • Design of biased IFNAR agonists that preferentially activate neuroprotective pathways

    • Development of small molecules that enhance specific aspects of IFN-β signaling

    • Combination approaches targeting complementary survival pathways

  • Timing-Optimized Interventions:

    • Pulsed delivery systems that prevent receptor desensitization

    • Stage-specific treatment protocols based on disease progression

    • Biomarker-guided administration to identify optimal treatment windows

  • Novel Delivery Technologies:

    • Blood-brain barrier-penetrating nanoparticles loaded with IFN-β

    • Gene therapy approaches for localized, sustained IFN-β production

    • Cell-based delivery using engineered stem cells that secrete IFN-β

  • Combination Therapeutic Strategies:

    • IFN-β with neurotrophic factors to enhance neuroprotection

    • Targeting complementary aspects of neuroinflammation

    • Sequential therapy approaches (inflammatory phase → repair phase)

  • Personalized Medicine Applications:

    • Patient stratification based on IFNAR pathway genomics

    • Pharmacogenomic approaches to predict responders

    • Monitoring systems for real-time assessment of pathway activation

The direct neuroprotective effects of IFN-β on neural progenitor cells, mediated through anti-apoptotic mechanisms and STAT pathway activation , provide strong rationale for these next-generation approaches that may enhance efficacy while reducing systemic side effects in conditions like multiple sclerosis.

Product Science Overview

Structure and Production

Recombinant Mouse IFN-β (His Tag) is a mouse full-length protein expressed in HEK 293 cells . The recombinant protein is typically produced in a non-glycosylated form and contains a His tag at the C-terminus, which facilitates its purification and detection . The molecular mass of the recombinant protein is approximately 22 kDa .

Biological Activity

IFN-β is part of the type I IFN multigene family, which includes several subtypes such as IFN-α, IFN-ε, IFN-κ, IFN-ω, IFN-τ, IFN-δ, and IFN-ζ . Type I interferons are expressed as a first line of defense against viruses and play a critical role in the antiviral response. They exert their antiviral activity through various mechanisms, including blocking viral entry into cells, controlling viral transcription, cleaving RNA, and preventing translation .

In addition to its antiviral properties, IFN-β modulates both innate and adaptive immune responses. It induces natural killer cell cytotoxicity, enhances the expression of major histocompatibility complex class I molecules on most cells, and upregulates costimulatory molecules on antigen-presenting cells . IFN-β also acts directly on CD8 T cells to promote clonal expansion and memory formation in response to viral infections .

Applications

Recombinant Mouse IFN-β (His Tag) is widely used in various research applications, including:

  • sELISA (sandwich ELISA): It is used as a standard protein in sELISA assays to quantify the presence of IFN-β in biological samples .
  • SDS-PAGE: The protein is suitable for use in SDS-PAGE for analyzing protein expression and purity .
Therapeutic Potential

IFN-β has been used in the treatment of multiple sclerosis (MS) in humans. It has been suggested that IFN-β inhibits the differentiation of human Th17 cells, which play a central role in the immunopathogenesis of MS . Animal studies have shown that mice deficient in the type I IFN receptor (IFNAR) have increased susceptibility to experimental autoimmune encephalomyelitis (EAE), a model for MS .

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.