The protein is available in two lyophilized formulations:
Reconstitution is performed at 100 µg/mL in PBS, with BSA added for carrier-containing formulations .
ED₅₀: 3–18 ng/mL for IL-6 secretion in NIH/3T3 fibroblasts .
Mechanism: Activates NF-κB and MAPK pathways via IL-1Rrp2/IL-1RAcP receptors, inducing proinflammatory cytokines (e.g., IL-6, CXCL1, IL-1β) .
Cell Targets:
Psoriasis: Promotes epidermal hyperplasia and IL-17/IL-23 axis activation .
Lung Inflammation: Amplifies neutrophilic influx and GM-CSF-dependent cytokine storms in COPD and viral exacerbation models .
Atherosclerosis: Enhances macrophage foam cell formation in ApoE knockout mice .
Inflammatory Disease Modeling:
Immune Cell Activation Studies:
Drug Screening:
UniGene: Mm.249379
IL-36 gamma (also known as IL-1F9) is a 17-18 kDa cytokine belonging to the IL-1 family. The recombinant mouse IL-36 gamma protein typically contains amino acids 13-164 and is expressed in E. coli expression systems. It functions as an agonist of NF-kappa B activation through the orphan IL-1-receptor-related protein 2 (IL-1Rrp2). IL-36 gamma is part of the IL-36 signaling system present in epithelial barriers and participates in local inflammatory responses similar to the IL-1 system, with which it shares the coreceptor IL-1RAP. This protein is primarily involved in skin inflammatory responses, neutrophilic airway inflammation, and innate immune responses to fungal pathogens .
IL-36 gamma is predominantly expressed by multiple cell types involved in first-line defense against pathogens. The highest levels of IL-36 gamma are produced by Langerhans cells, keratinocytes, and stomach chief cells and parietal cells. These cells contribute significantly to the first-line defense against pathogens in the skin, lungs and digestive tract. Additionally, expression can be induced in monocytes, bronchial epithelia, and other epithelial cells under specific stimulatory conditions . Within research contexts, understanding the cell-specific expression patterns is crucial for designing physiologically relevant experiments.
Several stimuli can induce IL-36 gamma expression:
Lipopolysaccharide (LPS) treatment of monocytes
IL-alpha/beta treatment of keratinocytes and bronchial epithelia
IL-17 stimulation of keratinocytes and bronchial epithelia
TNF-alpha treatment of keratinocytes and bronchial epithelia
Pro-inflammatory cytokines including IL-1β, IL-18, and IFN-γ in dendritic cells and macrophages
Additionally, IL-36 cytokines can enhance their own expression, creating a positive feedback loop that amplifies inflammatory responses. This auto-induction has been observed in multiple cell types including dendritic cells, keratinocytes, and epithelial cells .
N-terminal processing is critical for the full biological activity of IL-36 gamma. Unlike other IL-1 family members that possess caspase cleavage sites, IL-36 cytokines require post-translational processing at the N-terminal region to be fully active. Specifically, processing of IL-36γ proximal to S18 dramatically enhances its biological activity by 1,000-10,000 fold. This explains why early research showed variable results regarding the concentration of IL-36γ required to observe biological effects in vitro (ranging from 50 ng/ml to 500 ng/ml in different studies). Full-length recombinant IL-36 proteins typically appear less active than their endogenous counterparts due to this requirement for N-terminal trimming .
IL-36 gamma primarily activates two major signaling pathways:
NF-κB Signaling Pathway:
Activation occurs through binding to IL-1Rrp2 (IL-36R) which recruits IL-1RAcP
This forms a functional receptor complex that initiates signal transduction
Leads to nuclear translocation of NF-κB and subsequent gene transcription
MAPK Signaling Pathway:
Includes p38, JNK, and ERK1/2 activation
Leads to activation of transcription factors like c-Jun
These pathways can be monitored experimentally through:
Western blotting for phosphorylated forms of IκB, p38, JNK, and ERK1/2
Nuclear translocation assays for NF-κB p65 subunit
Reporter gene assays using NF-κB responsive elements
RT-qPCR for downstream target genes
Pharmacological inhibitors can be used to confirm pathway involvement (e.g., BAY 11-7082 for NF-κB, SB203580 for p38 MAPK) .
Distinguishing between processed and unprocessed IL-36 gamma requires careful experimental design:
Activity Comparison:
Use dose-response curves to compare potency (ED50 values)
Processed IL-36 gamma typically shows ED50 values in the low ng/mL range (3-18 ng/mL)
Unprocessed forms require 1000-10000 fold higher concentrations
Biochemical Verification:
SDS-PAGE analysis under reducing conditions (processed IL-36 gamma appears at ~17 kDa)
N-terminal sequencing to confirm processing site (proximal to S18)
Mass spectrometry to determine exact molecular weight
Functional Assays:
NF-κB activation assays in responsive cell lines (e.g., NIH-3T3)
IL-6 secretion assays (a common readout for IL-36 activity)
Cell-specific responses (e.g., DC maturation markers CD83 and CD86)
For controlled experiments, researchers should consider using commercially available truncated recombinant proteins (aa 13-164) which mimic the processed form, or enzymatically process full-length proteins in vitro prior to use .
Several methodological approaches can be employed:
In Vitro Models:
Primary cell cultures (keratinocytes, bronchial epithelial cells, dendritic cells)
Co-culture systems to study cell-cell interactions
Organotypic 3D cultures to mimic tissue architecture
Stimulation with disease-relevant triggers (e.g., TLR ligands, allergens)
In Vivo Models:
Transgenic mouse models with IL-36 pathway alterations
Disease-specific models:
Imiquimod-induced psoriasis
Oxazolone-induced colitis
Asthma models
Viral infection models
Mechanistic Studies:
Neutralizing antibodies against IL-36 gamma or its receptor
siRNA knockdown in relevant cell types
CRISPR/Cas9-mediated gene editing
Receptor antagonist (IL-36Ra) studies to block signaling
Translational Approaches:
IL-36 gamma operates within complex inflammatory networks:
Synergistic Interactions:
IL-36 gamma synergizes with IL-1β, TNF-α, and IL-17A to induce robust inflammatory responses
Acts with IL-12 to drive Th1 differentiation and IFN-γ production
Potentiates responses to pathogen-associated molecular patterns (PAMPs)
Amplification Loops:
IL-36 gamma induces production of IL-1β, TNF-α, and IL-6
These cytokines further induce IL-36 gamma expression
Creates a positive feedback loop that can amplify inflammation
Cross-regulation:
IL-36Ra antagonizes IL-36 gamma effects by competing for receptor binding
IL-10 and TGF-β may suppress IL-36 gamma-induced responses
Type I interferons can modulate IL-36 pathway activation
Cell-specific Effects:
In dendritic cells: induces IL-12, IL-1β, IL-6, TNF-α, and IL-23
In T cells: promotes Th1 and Th9 responses while inhibiting Treg cells
In epithelial cells: induces antimicrobial peptides and chemokines
This complex network can be studied using cytokine blocking antibodies, receptor antagonists, and analysis of downstream signaling pathway interactions .
For optimal results with recombinant mouse IL-36 gamma:
Storage Conditions:
Store lyophilized protein at -20°C to -80°C
After reconstitution, store at -80°C in single-use aliquots
Avoid repeated freeze-thaw cycles (no more than 3)
Short-term storage (1-2 weeks) at 4°C may be acceptable for reconstituted protein
Reconstitution Guidelines:
Reconstitute in sterile water or PBS
Gentle swirling is recommended rather than vortexing
Allow protein to stand for 10-15 minutes at room temperature
Filter sterilization (0.22 μm) may be necessary for cell culture applications
Stability Considerations:
Several reliable assays can measure IL-36 gamma activity:
Cell-based Bioassays:
NIH-3T3 mouse embryonic fibroblast IL-6 secretion assay (ED50: 3-18 ng/mL)
NF-κB reporter cell lines (e.g., HEK293 cells transfected with IL-36R and NF-κB reporter)
Dendritic cell maturation assay (measuring CD83, CD86 upregulation)
Biochemical Assays:
SDS-PAGE with silver staining for protein integrity (17 kDa band)
ELISA for protein quantification
Western blot for detection of IL-36 gamma in complex samples
Molecular Assays:
RT-qPCR for downstream gene expression (IL-6, IL-8, CCL20)
Signaling pathway activation (phospho-IκB, phospho-p38)
Chromatin immunoprecipitation for NF-κB binding to target promoters
Functional Readouts:
Optimization strategies for different model systems include:
In Vitro Cell Culture Systems:
Cell type selection: Choose physiologically relevant cells (keratinocytes, bronchial epithelial cells, dendritic cells)
Concentration range: Titrate IL-36 gamma between 0.1-100 ng/mL for processed forms
Time course: Monitor responses at multiple time points (4, 8, 24, 48 hours)
Synergy testing: Combine with other stimuli (TLR ligands, cytokines) at sub-optimal doses
Controls: Include IL-36Ra as negative control and known IL-36 inducers as positive controls
Ex Vivo Tissue Explants:
Tissue preparation: Maintain consistent size and viability
Culture conditions: Use specialized media with minimal serum
Delivery method: Consider slow-release delivery systems for sustained exposure
Analysis: Combine histology, gene expression, and secreted factors
In Vivo Models:
Delivery route: Local vs. systemic administration affects outcomes
Dosing schedule: Single bolus vs. repeated administration
Genetic background: Consider strain-specific differences in inflammatory responses
Age and sex: Control for these variables as they affect inflammatory responses
Readouts: Combine systemic (serum cytokines) and local (tissue) measurements
Translational Research:
Critical quality control parameters include:
Purity Assessment:
SDS-PAGE analysis (>95% purity recommended)
Silver staining for visualization of contaminants
High-resolution techniques like capillary electrophoresis for detailed analysis
Biological Activity:
Specific activity measurement (units/mg)
Dose-response curves in standard bioassays (e.g., NIH-3T3 IL-6 induction)
Comparison to reference standards or previous lots
Endotoxin Testing:
LAL (Limulus Amebocyte Lysate) assay
Acceptable levels: ≤0.005 EU/μg protein
Critical for preventing false inflammatory responses
Protein Characterization:
Mass spectrometry for molecular weight confirmation
N-terminal sequencing to verify processing site
Circular dichroism for secondary structure analysis
Size exclusion chromatography for aggregation assessment
Stability Testing:
Accelerated and real-time stability studies
Activity retention over time under recommended storage conditions
Freeze-thaw stability assessment
Lot-to-Lot Consistency:
IL-36 gamma offers valuable insights into several disease mechanisms:
Psoriasis Research:
IL-36 gamma is markedly elevated in psoriatic skin
Can be used to model keratinocyte activation and proliferation
Useful for studying cross-talk between keratinocytes and immune cells
Applications in testing anti-inflammatory compounds targeting the IL-36 pathway
Inflammatory Bowel Disease (IBD):
IL-36 gamma promotes CD4+ T cell-dependent colitis
Can be used to study epithelial-immune cell interactions in intestinal inflammation
Useful for investigating the role of IL-36/IL-36R signaling in producing pro-inflammatory cytokines
Applications in developing biomarkers for intestinal inflammation
Pulmonary Inflammation:
IL-36 gamma is elevated in asthma and during viral infections
Contributes to neutrophil influx in the lungs
Can be used to model inflammatory disorders of the lung
Applications in understanding airway epithelial responses to pathogens
Cardiovascular Research:
Researchers face several technical challenges when working with IL-36 gamma:
Protein Processing and Activation:
Challenge: Full-length recombinant proteins often show reduced activity
Solution: Use pre-processed recombinant proteins (aa 13-164) or develop controlled enzymatic processing protocols
Alternative: Generate expression constructs lacking N-terminal sequences
Receptor Complexity:
Challenge: IL-36R forms complexes with IL-1RAcP and exhibits cell-type specific expression
Solution: Perform receptor expression profiling before experiments
Alternative: Use reporter cell lines with controlled receptor expression
Downstream Signaling Redundancy:
Challenge: Overlapping signaling pathways with other IL-1 family members
Solution: Use combination approaches with selective inhibitors and genetic knockdowns
Alternative: Develop more specific readouts for IL-36-specific signaling
Translating In Vitro Findings:
Challenge: Cell culture conditions don't replicate the complex tissue environment
Solution: Adopt 3D culture systems and co-culture approaches
Alternative: Validate findings in ex vivo tissue explants before animal studies
Species Differences:
Several promising research directions are emerging:
IL-36 in Trained Immunity:
Investigation of IL-36 gamma's role in programming innate immune memory
Study of epigenetic changes induced by IL-36 signaling
Potential applications in vaccine adjuvant development
Tissue-Specific Functions:
Exploration of IL-36 functions beyond skin and mucosa
Investigation of IL-36 in adipose tissue inflammation and metabolism
Study of neuron-glia interactions mediated by IL-36
Precision Medicine Applications:
Development of IL-36 pathway biomarkers for patient stratification
Identification of genetic variants affecting IL-36 responsiveness
Design of targeted therapies for IL-36-driven inflammatory conditions
Microbiome Interactions:
Study of how commensal and pathogenic microbes regulate IL-36 expression
Investigation of IL-36's role in maintaining barrier immunity
Exploration of microbiome manipulation as a strategy to modulate IL-36 pathways
Novel Therapeutic Approaches:
IL-36 gamma interacts with diverse immune cell populations through direct and indirect mechanisms:
Dendritic Cells (DCs):
T Cells:
Direct effects: Enhances IFN-γ, IL-4, IL-17 production in CD4+ T cells
Differentiation: Promotes Th1 and Th9 responses, inhibits Treg functions
Methods: Intracellular cytokine staining, CFSE proliferation assays, ChIP for epigenetic changes
Applications: Understanding T cell polarization in inflammatory conditions
Macrophages:
Neutrophils:
Indirect recruitment: Via IL-36-induced chemokines
Function: Contributes to neutrophil influx in lungs during inflammation
Methods: Chemotaxis assays, intravital microscopy, neutrophil extracellular trap (NET) formation
Applications: Understanding neutrophilic inflammation in asthma and other conditions