Mouse IL-4 exerts effects through two receptor complexes:
Type I Receptor (hematopoietic cells):
Type II Receptor (non-hematopoietic cells):
Key biological roles include:
T cell polarization: Th2 differentiation via STAT6 activation
Mast cell/basophil survival: PI3K/AKT-mediated apoptosis inhibition
Tissue repair: M2 macrophage expansion (↑ Il10, Vegfa, Tgfb expression)
Kit | Sensitivity | Dynamic Range | Sample Types | Reference |
---|---|---|---|---|
Proteintech KE10010 | 1.0 pg/mL | 7.8–500 pg/mL | Serum, plasma, supernatants |
Model: C57BL/6 mice with myocardial infarction (MI)
Treatment: IL-4/anti-IL-4 complex (5 μg IL-4 + 25 μg antibody)
Results:
↑ M2 macrophages in infarct zone (80% vs 50% in controls)
↑ Microvascular density (32.4 ± 3.1 vs 18.9 ± 2.7 vessels/mm²)
↓ Infarct size by 41% at 28 days post-MI
IL-4 extends basophil survival (72-hour viability: 68% vs 12% controls)
Stabilizes FcεRI expression (MFI 8,432 vs 5,109 in cytokine-free conditions)
Acts via PI3K/AKT without transcriptional changes (post-translational BCL-2 modulation)
Mouse IL-4 shows no cross-reactivity with human cells due to divergent receptor-binding domains . Key residues for species specificity include:
This specificity is exploited in neutralizing antibodies like 11B11 (Bio X Cell BP0045), which blocks IL-4 bioactivity at IC50 < 1 μg/mL .
In the immune system, IL-4 primarily drives the differentiation of naive CD4+ T cells into Th2 cells and influences B cell antibody class switching. In the central nervous system, IL-4 affects cognitive processes through multiple mechanisms, including promotion of neurogenesis, modulation of astrocyte function, and regulation of neuroinflammation .
IL-4 signaling occurs through the IL-4 receptor (IL-4R), which consists of the IL-4Rα chain paired with either the common γ chain (forming type I receptor) or the IL-13Rα1 chain (forming type II receptor). Binding of IL-4 to its receptor activates the Janus kinase (JAK)/Signal Transducer and Activator of Transcription 6 (Stat6) pathway .
In transgenic mouse models, selective expression of IL-4Rα on specific cell types (such as smooth muscle cells) has been used to study the cell-specific effects of IL-4. For example, mice expressing IL-4Rα only on smooth muscle cells (IL-4Rα−/−/SMP8–IL-4Rα+/−) have been created to investigate the direct effects of IL-4 on airway hyperresponsiveness .
IL-4 knockout mice (IL-4−/−) exhibit several distinct phenotypes across multiple systems:
Cognitive deficits: IL-4−/− mice show severe learning defects in spatial learning tasks such as the Morris Water Maze (MWM) .
Neurogenesis impairment: These mice demonstrate reduced adult neurogenesis in specific zones of the adult brain .
Altered astrocyte function: Following learning tasks, IL-4−/− mice fail to induce astrocytic production of brain-derived neurotrophic factor (BDNF), which plays a key role in neuronal survival and dendrite arborization .
Pro-inflammatory meningeal environment: The meningeal myeloid cells (CD11b+) in IL-4−/− mice produce more TNF than their wild-type counterparts .
Resistance to airway hyperresponsiveness: IL-4−/− mice do not develop airway hyperresponsiveness in response to allergen challenges .
These phenotypes can be reversed through bone marrow transplantation from wild-type mice or adoptive transfer of IL-4-competent T cells, demonstrating the dynamic nature of IL-4's effects .
Age-related cognitive decline in mice appears to be partly related to changes in IL-4 signaling. The aging process in mice is associated with decreased hippocampal IL-4 levels concurrent with increases in pro-inflammatory cytokines such as IL-1β and IL-6 . This shift in the inflammatory balance appears to have functional consequences:
Direct intracerebroventricular (i.c.v.) administration of IL-4 can rescue long-term potentiation (LTP) defects observed in aged mice .
IL-4 can counteract the negative effects of IL-1β on LTP when co-administered i.c.v. .
Aged microglia become less responsive to IL-4 stimulation, showing decreased sensitivity to its anti-inflammatory effects .
This decreased sensitivity to IL-4 allows microglia to activate more readily in aging brains, contributing to LTP impairment .
These findings suggest that therapeutic strategies aimed at enhancing IL-4 signaling or mimicking its effects might help counter age-related cognitive decline by rebalancing the inflammatory environment in the aging brain.
When assessing airway hyperresponsiveness (AHR) in IL-4 research, several methodological considerations are critical:
Measurement techniques: Different techniques may yield varying results. Research shows that:
Barometric plethysmography (non-invasive)
Airway pressure-time index (APTI, invasive)
FlexiVent (invasive)
Each technique has different sensitivity in detecting AHR. For example, in IL-4Rα−/−/SMP8–IL-4Rα+/− mice, dust mite allergen induced increased responsiveness to methacholine as measured by barometric plethysmography but not by APTI, while flexiVent measurements showed only slight, non-significant increases .
Timing of measurements: The time course of IL-4-induced effects is important. IL-4C (long-acting form of IL-4) can induce some increase in airway responsiveness in as little as 24 hours, but greater effects at higher doses of methacholine are seen after 3 days .
Method of IL-4 administration: Different routes and formulations yield different results:
These methodological differences highlight the importance of using multiple measurement techniques and carefully considering administration routes when studying IL-4-induced AHR.
Cell-specific expression of the IL-4 receptor dramatically affects experimental outcomes, revealing the complex multi-cellular effects of IL-4 signaling:
Smooth muscle-specific IL-4Rα expression:
Mice expressing IL-4Rα only on smooth muscle cells (IL-4Rα−/−/SMP8–IL-4Rα+/−) develop airway hyperresponsiveness in response to IL-4 or IL-13 stimulation
This occurs without the development of airway eosinophilia or goblet cell hyperplasia seen in mice with normal IL-4Rα expression
These findings indicate that direct IL-4 signaling in smooth muscle is sufficient for AHR development
Selective deletion of IL-4Rα from smooth muscle:
Mice with IL-4Rα selectively deleted from smooth muscle still develop AHR, goblet cell hyperplasia, and airway eosinophilia in response to allergen challenges
This suggests that while smooth muscle IL-4Rα is sufficient for AHR development, it is not necessary when other cell types can respond to IL-4/IL-13
Epithelial cell-specific effects:
These findings highlight the importance of using cell-type-specific genetic approaches to dissect the complex mechanisms of IL-4 signaling in vivo.
Creating and validating IL-4 related knockout mice requires careful consideration of several factors:
For IL-4 knockout mice:
Targeting strategy: Complete deletion of the IL-4 gene or insertion of a disruptive sequence is commonly used.
Validation approaches:
For IL-4Rα conditional knockout mice:
Cre-loxP system: The most common approach uses tissue-specific Cre recombinase expression to delete floxed IL-4Rα alleles.
Promoter selection: Critical for cell-type specificity (e.g., SMP8 promoter for smooth muscle-specific expression) .
Validation requirements:
Use littermate controls whenever possible
Include both wild-type (IL-4Rα+/+) and heterozygous (IL-4Rα+/−) controls
Consider the background strain (most studies use BALB/c background)
Cognitive assessment in IL-4 knockout mice requires a comprehensive approach using multiple complementary tests:
Morris Water Maze (MWM):
Contextual Fear Conditioning:
Assesses hippocampal-dependent associative memory
Freezing behavior quantification provides measure of memory formation and recall
Novel Object Recognition:
Tests recognition memory with less stress than water-based tests
Utilizes mice's natural preference for novel objects
Electrophysiological measurements:
Combined behavioral and immunological assessment:
These approaches should be combined with careful control of variables such as age, sex, housing conditions, and testing environment to ensure reproducible results.
Bone marrow transplantation (BMT) is a powerful approach to determine whether IL-4 effects are mediated by bone marrow-derived immune cells. Based on published protocols that have successfully demonstrated restoration of cognitive function in IL-4−/− mice, the following methodology is recommended:
Recipient preparation:
Age: 6-8 weeks old IL-4−/− mice
Irradiation: Lethal dose (typically 900-1000 cGy) split into two equal doses 3-4 hours apart
Housing: Sterile conditions with antibiotic water (Neomycin 100 mg/L) for 2 weeks post-irradiation
Donor cell preparation:
Source: Wild-type mice (IL-4+/+) matched for background strain
Harvest: Femurs and tibias
Cell isolation: Flush bones with sterile PBS + 2% FBS
Cell preparation: Red blood cell lysis, filtering through 70μm mesh
Cell count: 5-10 × 10^6 viable cells per recipient
Transplantation procedure:
Route: Intravenous injection via tail vein
Monitoring: Daily for 2 weeks for signs of graft rejection or infection
Engraftment verification:
Timeline: Allow 8-12 weeks for complete reconstitution
Verification methods:
Flow cytometry of peripheral blood using congenic markers (CD45.1/CD45.2)
PCR genotyping of blood cells
Functional testing: Ex vivo stimulation of splenocytes to verify IL-4 production
Controls:
This protocol has been shown to successfully reverse the learning defects exhibited by IL-4−/− mice, confirming that bone marrow-derived cells (primarily T cells) are the critical source of IL-4 for cognitive function .
Research on IL-4 in mouse models has important implications for understanding and potentially treating human neurological disorders:
Alzheimer's Disease (AD):
Multiple Sclerosis (MS):
Age-related cognitive decline:
Neuroinflammatory conditions:
Translation to humans requires consideration of species differences in immune-brain interactions and validation in human tissues. Post-mortem studies of human brain tissue and imaging studies of neuroinflammation will be crucial to bridge the gap between mouse models and human applications.
Understanding species-specific differences in IL-4 signaling is critical when translating findings from mouse models to human applications:
Receptor structure and distribution:
While both species express IL-4Rα with similar structure, there are differences in tissue distribution and expression levels
Cell-type specific expression patterns may vary between species
Downstream signaling pathways:
Although both human and mouse IL-4 signaling activates the JAK/STAT6 pathway, there are species-specific differences in pathway regulation
Human cells may have different thresholds for activation or different kinetics of signal transduction
Cellular responses:
Human macrophages treated with IL-4 [hM(IL4)] promote epithelial wound repair through different mechanisms than mouse macrophages
Human IL-4-treated macrophages exhibit a CCL18+CD14low/− phenotype with specific gene expression patterns (RNA-seq revealed IL-4 affected expression of 996 genes in human macrophages)
Cross-reactivity limitations:
Human IL-4 can bind to mouse IL-4 receptors, but mouse IL-4 cannot activate human IL-4 receptors
This asymmetric cross-reactivity is important when designing preclinical studies
Environmental influences:
Laboratory mice are raised in controlled environments, whereas humans are exposed to diverse environmental factors that affect immune function
These differences may impact how IL-4 signaling operates in real-world versus laboratory conditions
These differences necessitate cautious interpretation when extrapolating mouse findings to human applications and highlight the importance of validating key findings in human cells or tissues when possible.
Several approaches for targeting IL-4 signaling show promise for neurological conditions, based on mouse model findings:
Direct IL-4 administration:
Gene therapy approaches:
Cell-based therapies:
Transplantation of IL-4-competent bone marrow or adoptive transfer of IL-4-producing T cells reverses cognitive deficits in IL-4−/− mice
M2-skewed macrophages (generated through IL-4 exposure) improve Morris Water Maze performance when administered i.c.v. or i.v.
Potential for ex vivo generation of IL-4-producing cells for therapeutic use in humans
Pharmacological induction of IL-4:
Compounds that increase endogenous IL-4 production could circumvent delivery challenges
Small molecules targeting upstream regulators of IL-4 expression represent an unexplored therapeutic avenue
Combination approaches:
Co-administration of IL-4 with other neurotrophic factors like BDNF might enhance efficacy
Combined anti-inflammatory and pro-regenerative approaches may provide synergistic benefits
Development pathway considerations:
Target validation in human tissues and biomarker development
Delivery optimization to overcome blood-brain barrier limitations
Dose-finding studies to determine therapeutic window
Safety monitoring for potential systemic immune effects of IL-4 modulation
The most promising approach may depend on the specific neurological condition being targeted, with different strategies optimal for acute versus chronic conditions.
When faced with conflicting data regarding IL-4's effects across different mouse models, researchers should consider several factors:
Genetic background differences:
Model-specific variations:
Measurement technique sensitivity:
Contextual IL-4 signaling:
IL-4's effects depend on the local microenvironment and concurrent signaling pathways
Both IFN-γ and IL-4 can elicit neuroprotective phenotypes in astrocytes through different mechanisms
IL-4 responses may differ based on prior exposure to other cytokines (e.g., IL-1β pretreated astrocytes respond differently to IL-4)
Reconciliation strategies:
Direct comparison studies using multiple models under identical conditions
Comprehensive phenotyping using multiple complementary techniques
Single-cell analysis to identify cell-type specific responses that may explain model differences
When publishing seemingly conflicting results, researchers should explicitly discuss potential reasons for discrepancies and design experiments to directly test hypotheses about these differences.
Measuring IL-4 production and signaling in mouse brain tissue presents several technical challenges that researchers should address:
Low abundance detection:
IL-4 is present at very low concentrations in brain tissue
Recommended approaches:
Use high-sensitivity ELISA kits (detection limit <1 pg/ml)
Consider amplification steps for immunohistochemistry
Employ multiplex cytokine assays to maximize data from limited samples
Regional heterogeneity:
IL-4 production and receptor expression vary across brain regions
Solutions:
Precise microdissection techniques for region-specific analysis
Single-cell approaches to characterize cell type-specific expression
In situ hybridization to visualize mRNA distribution while preserving spatial context
Cell type identification:
Multiple cell types can produce or respond to IL-4
Approaches:
Flow cytometry with appropriate markers for microglia, astrocytes, neurons
Immunofluorescence co-staining with cell type-specific markers
FACS sorting followed by qPCR or cytokine measurements
Rapid degradation:
IL-4 has a short half-life in tissues
Recommendations:
Rapid tissue processing (flash freezing within minutes)
Use of protease inhibitors in extraction buffers
Consider measuring downstream signaling (p-STAT6) as a more stable readout
Blood contamination:
Blood cells can be sources of IL-4, confounding brain tissue measurements
Solutions:
Perfusion with PBS prior to tissue collection
Inclusion of CD45 staining to identify blood-derived cells
Comparison with blood IL-4 levels as control
Validation strategies:
Always include positive controls (e.g., spleen tissue from immunized mice)
Use IL-4−/− mice as negative controls
Consider reporter mice (IL-4-GFP) for direct visualization of producing cells
By addressing these challenges methodically, researchers can obtain more reliable data about IL-4 production and signaling in the mouse brain.
Robust experimental design requires comprehensive controls when studying IL-4 in disease models:
Essential genetic controls:
Complete knockout controls:
IL-4−/− mice (global deletion)
IL-4Rα−/− mice (receptor deletion)
These verify that observed effects are truly IL-4-dependent
Cell-specific controls:
For conditional knockout/transgenic studies:
Cre-only controls (without floxed gene)
Floxed gene without Cre
Wild-type littermates
These control for potential effects of the genetic manipulation itself
Heterozygote controls:
Essential experimental controls:
Vehicle controls:
Match all aspects of treatment except the active component
For cytokine administration, include heat-inactivated cytokine controls
Timing controls:
Dose-response controls:
Include multiple doses to establish relationship between IL-4 levels and outcomes
Important for distinguishing physiological versus pharmacological effects
Cross-phenotype controls:
Measurement technique controls:
These comprehensive controls help ensure that findings are robust, reproducible, and specifically related to IL-4 signaling rather than experimental artifacts.
Mouse recombinant IL-4 is a protein that is typically produced using recombinant DNA technology. This involves inserting the gene encoding mouse IL-4 into a suitable expression system, such as Escherichia coli (E. coli) or mammalian cells, to produce the protein in large quantities . The recombinant protein is then purified and lyophilized for use in various applications. The molecular weight of mouse IL-4 is approximately 13.5 kDa, and it consists of 120 amino acid residues .
IL-4 is a member of the short four-helix bundle family of cytokines and is primarily produced by Th2 cells in response to antigen receptor engagement . It has several important functions in the immune system:
Recombinant mouse IL-4 is widely used in research and clinical applications. Some of its common uses include: