Recombinant human IL-36γ (formerly known as IL-1F9) is a member of the IL-1 family of cytokines. It is synthesized as a 19 kDa, 169 amino acid protein without a signal sequence, prosegment, or N-linked glycosylation sites . The protein shares 30% amino acid identity with IL-1ra, and various degrees of identity with other IL-1 family members, including 23% with IL-1β, 33% with IL-36ra, 57% with IL-36α, 35% with IL-37, 45% with IL-36β, and 32% with IL-1F10 . Functionally, IL-36γ activates NF-κB and MAPK pathways through binding to its receptor complex consisting of IL-1Rrp2 and IL-1RAcP .
IL-36γ is expressed by multiple cell types including Langerhans cells, keratinocytes, monocytes, bronchial epithelium, and gastric cells (Chief cells and Parietal cells) . The protein is secreted via a nonclassical pathway that likely requires extracellular ATP . Primary biological activities include:
Downregulation of betacellulin and upregulation of MMP-9 and MMP-10
Induction of multiple chemokines (CXCL1, 2, 3, 8 and CCL2, 3, 20)
Promotion of type-1 immune responses in bacterial infections
Enhancement of CD8+ T cell, NK cell, and γδ T cell activation
For optimal results when working with recombinant human IL-36γ:
Store lyophilized protein at -20°C to -70°C for up to 12 months from date of receipt
Once reconstituted, store at 2-8°C under sterile conditions for up to 1 month
For longer storage after reconstitution, store at -20°C to -70°C for up to 3 months under sterile conditions
Avoid repeated freeze-thaw cycles by aliquoting the reconstituted protein
Reconstitute in sterile PBS or specified buffer according to manufacturer's instructions
Centrifuge the vial prior to opening to ensure complete recovery of the protein
The optimal concentration range of IL-36γ varies depending on the experimental system and readout:
For induction of IL-8 in human epidermoid carcinoma cell line A431, the EC50 is approximately 1.5-9 ng/mL
In studies measuring IL-36γ stimulation of CD8+ T cells, concentrations ranging from 10-100 ng/mL have shown dose-dependent effects
When evaluating inflammatory responses in bronchial epithelial cells, concentrations of 10-100 ng/mL are typically used
For 3D skin equivalent models, 300 ng/mL of IL-36γ has been effective in inducing inflammatory responses
It is recommended to perform dose-response experiments to determine the optimal concentration for your specific cell system and experimental endpoint.
Several methods can be used to confirm the biological activity of recombinant IL-36γ:
IL-8 induction assay: Measure IL-8 secretion from IL-36γ-responsive cells such as HaCaT keratinocytes or A431 cells. The activity can be confirmed by inhibition with IL-36Ra
NF-κB activation assay: Use reporter cell lines expressing NF-κB response elements to measure activation following IL-36γ treatment
TR-FRET binding assay: Utilize Time-Resolved Fluorescence Resonance Energy Transfer to verify binding of IL-36γ to the IL-36R/IL-1RAcP heterodimer
MAPK phosphorylation: Measure phosphorylation of ERK1/2, JNK1/2, p38, and c-Jun following IL-36γ stimulation using Western blotting or phospho-specific flow cytometry
Multiplex cytokine assays: Measure downstream cytokine and chemokine production (IL-6, IL-8, CXCL1) in responsive cell types
To ensure robust experimental design when working with IL-36γ:
Negative controls: Untreated cells or cells treated with heat-inactivated IL-36γ
Positive controls: Cells treated with well-characterized inflammatory cytokines like IL-1β or TNF-α
Specificity controls: Treatment with IL-36Ra (receptor antagonist) to block IL-36γ activity
Receptor validation: Knockdown or knockout of IL-36R in target cells to confirm receptor-specific effects
Dose-response: Include multiple concentrations of IL-36γ to establish dose dependency
Time course: Multiple time points to determine optimal kinetics of the response
Carrier controls: If using carrier proteins like BSA, include carrier-only controls
Full-length IL-36γ (Met1-Asp169) exhibits significantly lower biological activity compared to N-terminally truncated forms:
Truncation of IL-36γ through proteolytic processing dramatically enhances its biological activity
The most active form of human IL-36γ begins at Ser18, resulting in the protein Ser18-Asp169
Various proteases from neutrophils, antigen-presenting cells, and epithelial cells can activate IL-36γ through processing
Activity assays show that truncated forms induce significantly higher IL-8 production compared to full-length protein
In experimental settings, researchers should consider using the truncated, more active form (Ser18-Asp169) for maximum biological effect
For comparative studies, the following table summarizes activity differences:
IL-36γ Form | Amino Acid Range | Relative Activity | Recommended Use |
---|---|---|---|
Full-length | Met1-Asp169 | Low | Structure-function studies |
Truncated | Ser18-Asp169 | High (~100-1000x higher) | Functional assays, in vivo studies |
Several experimental models have been validated for studying IL-36γ in different disease contexts:
In vitro models:
Human keratinocyte cultures (HaCaT cells) for skin inflammation studies
3D skin equivalents combining keratinocytes and fibroblasts for more physiologically relevant skin models
Primary immune cell cultures (T cells, NK cells, macrophages) for immunomodulatory studies
In vivo models:
IL-36γ and IL-36R knockout mice for loss-of-function studies
Bacterial pneumonia models (Streptococcus pneumoniae, Klebsiella pneumoniae) to study protective immune responses
Cancer models using IL-36γ-overexpressing tumor cells (B16 melanoma, 4T1 breast cancer) to evaluate anti-tumor effects
Microparticle-delivered IL-36γ for reconstitution studies in knockout mice
When selecting a model, consider the expression pattern of IL-36R in your model organism, as expression levels vary between tissues and species .
To comprehensively evaluate IL-36γ-induced signaling:
NF-κB pathway activation:
TransAM NF-κB p65 assay to measure nuclear translocation
IκBα degradation assessment by Western blot
NF-κB reporter assays using luciferase-based systems
MAPK pathway activation:
Transcriptional profiling:
RNA-seq or microarray analysis to identify IL-36γ-regulated genes
Real-time PCR for targeted gene expression analysis (IL-8, CXCL1, IL-6)
Chromatin immunoprecipitation (ChIP) to identify transcription factor binding sites
Temporal considerations:
Early signaling events (5-30 minutes): MAPK phosphorylation
Intermediate events (30-120 minutes): NF-κB nuclear translocation
Late events (2-24 hours): Gene expression and protein secretion
Pathway inhibitors for validation:
Researchers frequently encounter these challenges when working with IL-36γ:
Low biological activity: Full-length IL-36γ requires processing for full activity. Solution: Use truncated forms (Ser18-Asp169) or pre-activate with specific proteases
Species-specificity issues: Human IL-36γ shows only 53% amino acid identity with mouse IL-36γ . Solution: Use species-matched recombinant proteins for in vivo or ex vivo studies
Receptor expression variability: IL-36R expression varies across cell types. Solution: Verify IL-36R expression in your experimental system before studying IL-36γ effects
Protein aggregation: IL-36γ may form aggregates during storage or thawing. Solution: Filter reconstituted protein through a 0.2μm filter and avoid repeated freeze-thaw cycles
Carrier protein interference: BSA in some preparations may interfere with certain assays. Solution: Use carrier-free formulations when possible
Signal detection sensitivity: Low-level responses may be difficult to detect. Solution: Use sensitive detection methods like ELISA or multiplex assays, and include positive controls
To ensure reproducibility when working with different IL-36γ preparations:
Standardize activity measurements:
Establish an in-house bioassay (e.g., IL-8 induction in HaCaT cells)
Calculate specific activity (units/mg) for each batch
Normalize dosing based on activity rather than protein concentration
Check protein specifications:
Internal reference standards:
Maintain an internal reference preparation
Perform side-by-side comparisons with new batches
Generate relative potency calculations
Documentation practices:
Record lot numbers, sources, and formulations
Document storage conditions and reconstitution methods
Note any carrier proteins or additives
Cross-validation:
Test multiple biological readouts with each batch
Perform dose-response curves to identify potential shifts in potency
Consider multiple time points to account for kinetic differences
To distinguish between endogenous and exogenously added recombinant IL-36γ:
Tagged recombinant proteins:
Use His-tagged, Flag-tagged, or Fc-fusion IL-36γ
Detect with tag-specific antibodies via Western blot or immunofluorescence
Separate from endogenous protein by size difference on Western blots
Species-specific detection:
Use human IL-36γ in mouse systems with human-specific antibodies
Apply species-specific qPCR primers to differentiate transcript origin
Truncation-specific antibodies:
Generate antibodies that specifically recognize the N-terminus of truncated forms
Use these for selective detection of processed IL-36γ
Mass spectrometry-based approaches:
Use isotope-labeled recombinant protein (15N or 13C labeled)
Perform mass spectrometry to differentiate labeled vs. unlabeled peptides
Quantify relative abundance of endogenous vs. exogenous forms
Timing considerations:
Recent research reveals complex interactions between IL-36γ and other inflammatory mediators:
IL-36γ and IL-1 family cross-regulation:
Interface with type-1 immunity:
Role in tissue remodeling and fibrosis:
Wnt signaling pathway interactions:
These complex interactions suggest that targeting IL-36γ may have context-dependent effects depending on the disease state and inflammatory milieu.
The development of IL-36γ-targeting therapeutics presents various opportunities and considerations:
Cutting-edge approaches for investigating IL-36γ molecular mechanisms include:
TR-FRET displacement assays:
3D skin equivalent models:
Protease activity profiling:
Structural biology approaches:
Crystallography studies of IL-36γ in complex with IL-36R
Molecular dynamics simulations to understand conformational changes
Structure-based drug design for developing targeted IL-36γ modulators
Single-cell analysis:
Single-cell RNA sequencing to identify IL-36γ-responsive cell populations
CyTOF (mass cytometry) for high-dimensional analysis of signaling responses
Spatial transcriptomics to map IL-36γ expression and activity in tissue contexts
These advanced methods are expanding our understanding of IL-36γ biology and facilitating development of targeted therapeutics.