The protein is produced in either E. coli or HEK 293 cells, with variations in carrier protein inclusion :
Recombinant Mouse IL-4 exhibits potent bioactivity across immune cell types:
Mechanism: Binds IL-4Rα/γc (Type I) or IL-4Rα/IL-13Rα1 (Type II) receptors, activating STAT6 signaling .
Functional Roles:
Infection with IL-4-expressing ectromelia virus (ECTV-IL4) in resistant mice caused lethal mousepox by suppressing CTL/NK cytolytic activity and IFN-γ production .
Mortality reached 80% in C57BL/6 mice, mimicking outcomes in susceptible strains .
Overexpression in murine asthma models increased pulmonary lymphocyte infiltration and IgE levels, highlighting its role in allergic pathways .
Used to validate IL-4Rα antagonists in autoimmune disease models, showing reduced Th2-mediated pathology .
Recombinant mouse IL-4 is a single, non-glycosylated polypeptide chain consisting of 121 amino acid residues. The typical amino acid sequence spans His23-Ser140, with an N-terminal Met addition when expressed in bacterial systems. The molecular mass ranges from 13.2 to 13.7 kDa as determined by SEC-MALS (Size Exclusion Chromatography-Multi-Angle Light Scattering) analysis, confirming that the protein exists primarily as a monomer in solution .
The biological activity of recombinant mouse IL-4 is primarily measured through a proliferation assay using HT-2 mouse T cell lines. The effective dose for 50% maximal response (ED50) typically ranges between 0.2-1.5 ng/mL, corresponding to a specific activity of ≥5×10^6 U/mg . This standardized assay is calibrated against reference standards such as those provided by the WHO/National Institute for Biological Standards and Control (NIBSC code 91/656) .
For researchers conducting activity assays, it's important to note that biological response curves are typically sigmoidal, and accurate determination requires testing multiple concentrations spanning at least one log above and below the expected ED50. Results should be analyzed using appropriate curve-fitting software to determine precise activity metrics .
Recombinant mouse IL-4 is typically supplied as a lyophilized product that should be stored at -20°C until reconstitution. Upon receipt, the product should be immediately stored at the recommended temperature to maintain stability and biological activity .
For reconstitution:
Lyophilized mouse IL-4 should be reconstituted using deionized sterile-filtered water to a final concentration of 0.1–1.0 mg/mL in a minimal volume of 100 μL
For preparations containing BSA as a carrier protein, reconstitute at 100 μg/mL in sterile PBS containing at least 0.1% human or bovine serum albumin
For carrier-free preparations, reconstitute at 100 μg/mL in sterile PBS
Further dilutions should be prepared with buffer containing 0.1% bovine serum albumin (BSA) or human serum albumin (HSA) in phosphate-buffered saline
After reconstitution, it is recommended to aliquot the protein to avoid repeated freeze-thaw cycles, which can diminish biological activity. Reconstituted aliquots should be stored at –20°C or below for optimal stability .
While conventional IL-4 is encoded by four exons, an alternatively spliced isoform known as IL-4δ2 is encoded by exons 1, 3, and 4, omitting exon 2. Experimental studies comparing human IL-4 isoforms in mouse models have revealed significant functional differences. Both isoforms cause similar pulmonary infiltration of T and B lymphocytes when delivered to mouse lungs using replication-deficient adenovirus-mediated gene delivery, but notably, they do not induce eosinophil infiltration .
The most striking differences between these isoforms appear in their modulation of the cytokine milieu. IL-4δ2 induces higher levels of pro-inflammatory cytokines (TNF-α, IL-1, MCP-1) and Th1 cytokines (IL-12, IFN-γ) compared to conventional IL-4. This indicates that IL-4δ2 may have a more pronounced pro-inflammatory effect than conventional IL-4, suggesting potentially distinct signaling mechanisms or receptor engagement patterns .
These findings have important implications for experimental design, as researchers should carefully consider which IL-4 isoform is most appropriate for their specific research questions, especially in studies examining inflammatory responses or Th1/Th2 balance.
IL-4 receptor signaling involves complex interactions between IL-4 and its receptors, with several critical molecular determinants. The human IL-4 receptor contains a "gene regulation domain" between amino acids 558-657 that is essential for signaling. Within this domain, three conserved tyrosine residues (Y2, Y3, and Y4) play crucial roles in signal transduction .
For researchers studying IL-4 signaling:
Consider potential differences in signaling mechanisms between mouse and human IL-4 systems
Evaluate receptor expression levels when interpreting signaling experiments, as high receptor density may compensate for partial signaling defects
Remember that extended domain structure plays an essential role in the recognition and function of juxtatyrosine sequences in IL-4R signaling
Cross-species activity of IL-4 presents both challenges and opportunities for researchers. Structural analysis indicates that key residues in human IL-4 that define its binding to human IL-4Rα (Glu-9 and Arg-88), as well as minor determinants (Arg-53 and Arg-85), are all present in mouse IL-4. Direct binding experiments using BIAcore systems have confirmed that glycosylated mouse IL-4 can bind to human IL-4 receptor α (IL-4Rα) .
Multiple studies have demonstrated that human IL-4 can be active in mice or rats in vivo, functioning similarly to mouse IL-4. This cross-species activity enables at least limited testing of anti-human IL-4 therapies in mouse models, creating opportunities for translational research .
Potentially higher doses may be required for cross-species applications
Validation of receptor binding and downstream signaling is essential
Comparisons with species-matched controls should be included whenever possible to assess relative activity
E. coli Expression Systems:
The BL21(DE3)-CodonPlus E. coli strain with pET-21b (+) vector has been successfully used to express mouse IL-4, though the protein is often produced as an insoluble molecule requiring refolding
After induction with IPTG, the expressed protein should be analyzed by SDS-PAGE to confirm correct size (typically 13-17.5 kDa depending on the construct)
If the protein is expressed in inclusion bodies, solubilization with guanidine hydrochloride and dithiothreitol followed by refolding and purification by chromatography may be necessary
Purification Considerations:
Western blot analysis using anti-IL-4 specific antibodies should be performed to confirm the identity of the purified protein
The purity should exceed 97% as determined by SDS-PAGE analysis
Endotoxin levels should be measured and maintained below 1.0 EU/μg to avoid confounding experimental results
For researchers requiring glycosylated IL-4, mammalian expression systems may be preferable, though they typically yield lower amounts of protein compared to bacterial systems.
Validating the bioactivity of recombinant IL-4 is critical for experimental reproducibility. A multi-faceted approach is recommended:
Proliferation Assays:
HT-2 mouse T cell proliferation assay remains the gold standard, with expected ED50 values between 0.2-1.5 ng/mL
Cell viability should be measured using an appropriate method (MTT, XTT, or alamarBlue assays)
A dose-response curve using at least 8 concentration points spanning 0.01-10 ng/mL should be established
Receptor Binding Confirmation:
Surface plasmon resonance (e.g., BIAcore) to measure direct binding kinetics to IL-4Rα
Flow cytometry to assess binding to receptor-expressing cells
Downstream Signaling Validation:
Western blot analysis of STAT6 phosphorylation
Reporter gene assays for IL-4-responsive elements
Activity Comparison Table:
Assay Type | Expected Result for Active IL-4 | Typical Detection Range | Controls |
---|---|---|---|
HT-2 Proliferation | ED50: 0.2-1.5 ng/mL | 0.01-10 ng/mL | Positive: IL-2 (2-10 ng/mL) Negative: Heat-inactivated IL-4 |
STAT6 Phosphorylation | Detectable within 15-30 min | 1-20 ng/mL | Positive: Commercial IL-4 Negative: Medium only |
CD23 Expression | 2-5 fold increase | 5-20 ng/mL | Positive: Commercial IL-4 Negative: Isotype control |
Receptor Binding | KD: 0.1-1 nM | 0.1-100 nM | Positive: Commercial IL-4 Negative: Unrelated protein |
When designing experiments involving recombinant mouse IL-4 treatment, researchers should consider several factors to ensure robust and reproducible results:
Dosage Determination:
Conduct preliminary dose-response studies covering at least 3 logs (e.g., 0.1-100 ng/mL)
For in vivo studies, consider both local concentration at target tissues and systemic distribution
Account for potential differences in potency between different preparations of IL-4
Timing and Duration:
Acute vs. chronic exposure should be determined based on research objectives
For signaling studies, early time points (5-60 minutes) are critical for capturing phosphorylation events
For gene expression changes, later time points (2-24 hours) may be more appropriate
For phenotypic changes (e.g., T cell differentiation), extended exposure (3-7 days) is typically required
Delivery Methods:
In vitro: Direct addition to culture medium, with consideration of protein stability over time
In vivo: Consider adenovirus-mediated gene delivery for sustained expression, as demonstrated for IL-4 isoforms in lung studies
For localized delivery, options include osmotic pumps or matrix-embedded slow-release formulations
Control Conditions:
Vehicle control (buffer with matching carrier protein concentration)
Heat-inactivated IL-4 as a negative control
Species-matched positive controls for cross-species experiments
Consider including other cytokines (e.g., IL-13) to distinguish IL-4-specific effects from shared receptor signaling
Researchers occasionally encounter variable or contradictory results when using recombinant IL-4 in experiments. Several approaches can help resolve these issues:
Protein Quality Assessment:
Verify activity using standardized bioassays (HT-2 proliferation)
Check for protein degradation by SDS-PAGE and Western blot
Measure endotoxin levels, as contamination can confound immunological experiments
Receptor Expression Analysis:
Quantify IL-4Rα expression levels on target cells, as receptor density impacts signaling sensitivity
Consider analyzing common IL-4R polymorphisms that might affect responsiveness
Remember that high receptor expression can compensate for partial signaling defects in IL-4R mutants
Experimental Conditions Audit:
Document exact protocol parameters including cell density, media composition, and timing
Assess the impact of serum components, which may contain IL-4 binding proteins
Control for inadvertent activation of cells during processing, which can alter cytokine responsiveness
Reconciliation Strategies:
Directly compare IL-4 preparations using side-by-side assays
Use multiple readouts (e.g., STAT6 phosphorylation, gene expression, and functional responses)
Consider the presence of inhibitory factors or receptor antagonists in your experimental system
Several common pitfalls can compromise IL-4 functional studies. Awareness and preventive measures include:
Storage and Handling Issues:
Avoid repeated freeze-thaw cycles that diminish biological activity
Prepare single-use aliquots after reconstitution
Use a manual defrost freezer for storage to maintain consistent temperature
Reconstitution Errors:
Follow manufacturer's recommendations for reconstitution buffer and concentration
Include carrier protein (0.1% BSA or HSA) for dilutions to prevent loss through adsorption to plastics
Allow complete solubilization before use (gentle swirling rather than vortexing)
Experimental Design Flaws:
Inadequate concentration range in dose-response studies
Insufficient time points to capture transient signaling events
Lack of appropriate positive and negative controls
Failure to account for endogenous IL-4 production in mixed cell populations
Data Interpretation Challenges:
Over-interpretation of in vitro findings without in vivo validation
Failure to consider IL-4 isoform-specific effects, as conventional IL-4 and IL-4δ2 may have distinct functions
Not accounting for differences between human and mouse IL-4 systems when translating findings across species
Overlooking the importance of juxtatyrosine residues and extended domain structures in IL-4R signaling
Distinguishing direct IL-4 effects from indirect consequences in complex systems presents a significant challenge. Methodological approaches to address this include:
Receptor Blocking Studies:
Use neutralizing antibodies against IL-4 or IL-4Rα
Apply soluble IL-4 receptors to competitively inhibit IL-4 binding
Compare results with IL-4Rα knockout or knockdown models
Cell-Specific Response Analysis:
Isolate individual cell populations before and after IL-4 treatment
Use cell type-specific markers in flow cytometry or immunohistochemistry
Employ single-cell RNA sequencing to identify direct responders
Consider conditional IL-4Rα knockout models targeting specific cell populations
Timing Analysis:
Establish a detailed time course to distinguish primary (rapid) from secondary (delayed) responses
Compare kinetics of different outcome measures to establish likely causal relationships
Use transcription or translation inhibitors to block secondary effects
Pathway Delineation:
Target specific components of the IL-4 signaling cascade (e.g., JAK1, STAT6) to confirm mechanistic involvement
Compare responses to IL-13, which shares the Type II IL-4 receptor but may elicit distinct outcomes
Consider the functional differences between IL-4 isoforms, as IL-4δ2 induces different cytokine profiles compared to conventional IL-4
When comparing experimental results obtained using different recombinant IL-4 preparations, researchers should consider several variables that can impact outcomes:
Protein Characteristics:
Sequence variations: Confirm identical amino acid sequences (His23-Ser140 with N-terminal Met is standard for E. coli-derived mouse IL-4)
Post-translational modifications: E. coli-derived IL-4 lacks glycosylation, which may affect activity compared to mammalian-expressed protein
Purity: Standardize to >97% purity as determined by SDS-PAGE analysis
Endotoxin levels: Ensure comparable and low endotoxin content (<1.0 EU/μg)
Activity Standardization:
Normalize treatments based on specific activity rather than mass concentration
Consider calibrating against reference standards such as WHO/NIBSC preparations (code 91/656)
Perform side-by-side bioactivity assays when directly comparing different preparations
Document ED50 values (expected range: 0.2-1.5 ng/mL for HT-2 proliferation)
Experimental Design Harmonization:
Standardize carrier protein concentrations across preparations
Use consistent reconstitution and storage protocols
Apply identical experimental conditions (cell density, media composition, incubation times)
Include internal controls to normalize between experiments
Recent and ongoing research continues to expand our understanding of IL-4 biology in ways that may impact experimental approaches:
Isoform-Specific Functions:
Research on splice variants such as IL-4δ2 reveals distinct functional profiles compared to conventional IL-4. IL-4δ2 appears to induce stronger pro-inflammatory and Th1 responses, suggesting potential specialized roles in immune regulation. Future studies may uncover additional splice variants with unique functions .
Cross-Species Applications:
The discovery that human IL-4 can be functionally active in mice opens new possibilities for translational research. This cross-species activity enables limited pre-clinical testing of human IL-4-targeting therapies in animal models, potentially accelerating therapeutic development .
Receptor Signaling Complexity:
The importance of extended domain structure and juxtatyrosine sequences in IL-4R signaling indicates more complex receptor-cytokine interactions than previously appreciated. Future research may reveal additional structural determinants of signaling specificity and strength .
Emerging Technologies:
New methodologies including CRISPR-based gene editing, single-cell analysis, and advanced structural biology techniques promise to further deepen our understanding of IL-4 biology. These approaches may reveal novel regulatory mechanisms, interaction partners, and therapeutic targets related to IL-4 signaling.
As research progresses, experimental design should adapt to incorporate these emerging insights, ensuring that IL-4 studies remain at the cutting edge of immunological research.