Recombinant Human Interleukin-36 gamma protein (IL36G) (Active)

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Lyophilized from a 0.2 µm filtered PBS, pH 7.4.
Form
Lyophilized powder
Lead Time
5-10 business days
Notes
Repeated freezing and thawing is not recommended. Store working aliquots at 4°C for up to one week.
Reconstitution
We recommend that this vial be briefly centrifuged prior to opening to bring the contents to the bottom. Please reconstitute protein in deionized sterile water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% of glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our default final concentration of glycerol is 50%. Customers can use it as a reference.
Shelf Life
The shelf life is influenced by numerous factors, including storage conditions, buffer ingredients, storage temperature, and the inherent stability of the protein itself. Generally, the shelf life of the liquid form is 6 months at -20°C/-80°C. The shelf life of the lyophilized form is 12 months at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquoting is necessary for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag-Free
Synonyms
IL 1 epsilon; IL 1 related protein 2; IL 1(EPSILON); IL 1F9; IL 1H1; IL 1RP2; IL-1 epsilon; IL-1-related protein 2; IL-1F9; IL-1H1; IL-1RP2; IL1E; Il1f9; IL1F9_HUMAN; IL1H1; IL1RP2; IL36 gamma; IL36G; Interleukin 1 epsilon; Interleukin 1 family member 9; Interleukin 1 homolog 1; Interleukin 1 related protein 2; Interleukin 36 gamma; Interleukin-1 epsilon; Interleukin-1 family member 9; Interleukin-1 homolog 1
Datasheet & Coa
Please contact us to get it.
Expression Region
1-169aa
Mol. Weight
18.7 kDa
Protein Length
Full Length
Purity
>95% as determined by SDS-PAGE.
Research Area
Immunology
Source
E.coli
Species
Homo sapiens (Human)
Target Names
IL36G
Uniprot No.

Target Background

Function
Interleukin-36 gamma (IL-36γ) is a cytokine that binds to and signals through the IL1RL2/IL-36R receptor, subsequently activating NF-κB and MAPK signaling pathways in target cells. It constitutes a part of the IL-36 signaling system, believed to be present in epithelial barriers and involved in local inflammatory responses. This system shares similarities with the IL-1 system, featuring the coreceptor IL1RAP. IL-36γ appears to play a role in skin inflammatory responses by influencing keratinocytes, dendritic cells, and indirectly T-cells, promoting tissue infiltration, cell maturation, and proliferation. In cultured keratinocytes, it induces the expression of macrophage, T-cell, and neutrophil chemokines, including CCL3, CCL4, CCL5, CCL2, CCL17, CCL22, CL20, CCL5, CCL2, CCL17, CCL22, CXCL8, CCL20, and CXCL1. It also stimulates its own expression and that of prototypic cutaneous proinflammatory parameters such as TNF-α, S100A7/psoriasin, and inducible NOS. It may contribute to proinflammatory responses during specific neutrophilic airway inflammation. It activates mitogen-activated protein kinases and NF-κB in primary lung fibroblasts and stimulates the expression of IL-8, CXCL3, and Th17 chemokine CCL20 in lung fibroblasts. IL-36γ may be involved in the innate immune response to fungal pathogens, such as Aspergillus fumigatus.
Gene References Into Functions
  1. Serum IL-36γ levels were elevated in active systemic lupus erythematosus patients and correlated with disease activity and arthritis. PMID: 29571080
  2. Cathepsin S was identified as the primary IL-36γ-activating protease expressed in epithelial cells. PMID: 28289191
  3. Enhanced expression of IL-36γ was observed in plasma and bronchoalveolar lavage fluid of patients with acute respiratory distress syndrome due to bacterial pneumonia. PMID: 28176791
  4. IL-36-mediated IL-6 and CXCL8 production in human lung fibroblasts and bronchial epithelial cells may be implicated in pulmonary inflammation, especially caused by bacterial or viral infections. PMID: 28869889
  5. With a focus on the skin as a target for microbial and viral invasion, a review summarizes the current understanding of IL-36 (IL-36α, IL-36β, and IL-36γ) functions. One proposed physiological function of IL-36s is to counter microbial immune evasion. [Review] PMID: 28811383
  6. IL-36γ inhibits differentiation and induces inflammation of keratinocytes via the Wnt signaling pathway in psoriasis. PMID: 28924372
  7. IL-36γ-stimulated endothelial cells secrete the proinflammatory chemokines IL-8, CCL2, and CCL20. PMID: 27673278
  8. Skin injury increases IL-36γ through the activation of the TLR3-SLUG-VDR axis, and IL-36γ induces REG3A to promote wound healing. PMID: 28774595
  9. Autocrine and Paracrine Regulation of Keratinocyte Proliferation through a Novel Nrf2-IL-36γ Pathway. PMID: 27183581
  10. Researchers demonstrate that Mycobacterium tuberculosis infection of macrophages induces IL-36γ production in a two-stage-regulated manner. PMID: 27389350
  11. IL-36γ, a member of the IL-1 superfamily, is involved in host defense and contributes to proinflammatory responses and the development of inflammatory diseases. PMID: 27853811
  12. IL-36γ is significantly more strongly expressed in the epidermis of patients with psoriasis-based erythroderma than in other inflammatory skin diseases. PMID: 26524325
  13. A study shows that plasma concentrations of IL-36α and IL-36γ are overexpressed in active systemic lupus erythematosus patients and that IL-36α exhibits a substantial pro-inflammatory effect through regulation of IL-6 and CXCL8 production. PMID: 26516833
  14. IRF6 is likely to promote inflammation to P. gingivalis through its regulation of IL-36γ. PMID: 26819203
  15. Findings indicate that Interleukin (IL)-1β-induced interleukin 36γ (IL-36γ) expression is mediated by the activation of transcriptional factors, NF-κB p65 and AP-1 (c-jun). PMID: 26562662
  16. IL36G was identified as a strong regulator of skin pathology in both lesional and non-lesional skin samples. PMID: 25897967
  17. Decreased Langerhans cell responses to IL36G: altered innate immunity in patients with recurrent respiratory papillomatosis. PMID: 24950037
  18. IL-36γ expression inversely correlated with the progression of human melanoma and lung cancer. PMID: 26321222
  19. IL-36γ is a valuable biomarker in psoriasis patients, both for diagnostic purposes and measurement of disease activity during the clinical course. PMID: 25525775
  20. CAMP induces IL-36γ expression leading to the initiation of skin inflammation and occasional exacerbations of psoriasis. PMID: 25305315
  21. IL-36 promotes myeloid cell infiltration, activation, and inflammatory activity in the skin. PMID: 24829417
  22. This is the first report of extracellular release of endogenous IL-36γ through pyroptosis, suggesting a function of IL-36γ as an alarmin. PMID: 22318382
  23. Data presented herein shed further light on the involvement of T-bet in innate immunity and suggest that IL-36γ, besides IFNγ, may contribute to the functions of this transcription factor in immunopathology. PMID: 23095752
  24. Interleukin-36 (IL-36) ligands require processing for full agonist (IL-36α, IL-36β, and IL-36γ) or antagonist (IL-36Ra) activity. PMID: 21965679
  25. Regulation and function of the IL-1 family cytokine IL-1F9 in human bronchial epithelial cells. PMID: 20870894
  26. Expression of IL-1F9 is increased in human plaque psoriasis skin and is overexpressed in a transgenic mouse psoriasis model. PMID: 21242515
  27. IL-1F6 and IL-1F8, in addition to IL-1F9, activate the pathway leading to NF-κB in an IL-1Rrp2-dependent manner in Jurkat cells. PMID: 14734551
  28. This is the first report of IL-1 genotype association with the inflammation of skeletal muscle following acute resistance exercise that may potentially affect the adaptations to chronic resistance exercise. PMID: 15331687
  29. This report demonstrates expression of IL1F9 by bronchial epithelial cells induced by pro-inflammatory stimuli, suggesting a function of this molecule in airway inflammation. PMID: 15701729

Show More

Hide All

Database Links

HGNC: 15741

OMIM: 605542

KEGG: hsa:56300

STRING: 9606.ENSP00000259205

UniGene: Hs.211238

Protein Families
IL-1 family
Subcellular Location
Cytoplasm. Secreted.
Tissue Specificity
Highly expressed in tissues containing epithelial cells: skin, lung, stomach and esophagus. Expressed in bronchial epithelial. In skin is expressed only in keratinocytes but not in fibroblasts, endothelial cells or melanocytes. Up-regulated in lesional ps

Q&A

What is the structural and functional characterization of recombinant human IL-36γ?

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 .

Which cell types express IL-36γ and what are its primary biological activities?

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:

  • Activation of NF-κB and MAPK signaling pathways

  • Downregulation of betacellulin and upregulation of MMP-9 and MMP-10

  • Activation of macrophages and fibroblasts

  • 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

How should researchers store and reconstitute recombinant IL-36γ protein?

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

What is the optimal concentration range for IL-36γ in cell-based assays?

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.

How can researchers verify the biological activity of recombinant IL-36γ?

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

What controls should be included when using recombinant IL-36γ in experiments?

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

How does processed/truncated IL-36γ differ from full-length protein in biological activity?

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 FormAmino Acid RangeRelative ActivityRecommended Use
Full-lengthMet1-Asp169LowStructure-function studies
TruncatedSer18-Asp169High (~100-1000x higher)Functional assays, in vivo studies

What experimental models are most suitable for studying IL-36γ functions in disease contexts?

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

  • Bronchial epithelial cell cultures for respiratory research

  • 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 .

How can researchers effectively measure IL-36γ-induced signaling pathways?

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:

    • Western blotting for phosphorylated ERK1/2, JNK1/2, p38, and c-Jun

    • Flow cytometry with phospho-specific antibodies for single-cell analysis

    • Kinase activity assays for downstream substrates

  • 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:

    • IKK inhibitors (e.g., BMS-345541) for NF-κB pathway

    • MAPK inhibitors (U0126 for ERK, SP600125 for JNK, SB203580 for p38)

    • MyD88 inhibitors to block upstream signaling

What are the common technical challenges when working with recombinant IL-36γ?

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

How can researchers effectively compare results obtained with different sources or batches of recombinant IL-36γ?

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:

    • Verify amino acid sequence (full-length vs. truncated forms)

    • Confirm purity (≥95% by SDS-PAGE is standard)

    • Validate by Western blot with specific antibodies

  • 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

What techniques are recommended for detecting endogenous versus exogenous IL-36γ in experimental systems?

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:

    • Measure baseline levels before adding recombinant protein

    • Track kinetics of IL-36γ degradation in your experimental system

    • Consider that exogenous IL-36γ may induce endogenous IL-36γ expression

How does IL-36γ interact with other inflammatory pathways in complex disease models?

Recent research reveals complex interactions between IL-36γ and other inflammatory mediators:

  • IL-36γ and IL-1 family cross-regulation:

    • IL-36γ and IL-1β show reciprocal regulation in various tissues

    • IL-36Ra and IL-1Ra have opposing effects on infectious keratitis outcomes

    • IL-36γ can induce IL-1β expression in some contexts but suppress it in others

  • Interface with type-1 immunity:

    • IL-36γ synergizes with IL-12 to enhance CD8+ T cell and NK cell activity

    • It promotes IFN-γ production from multiple lymphocyte populations

    • IL-36γ acts as a crucial proximal component in protective type-1-mediated lung immunity

  • Role in tissue remodeling and fibrosis:

    • IL-36 signaling promotes secretion of profibrotic mediators

    • IL-36γ may serve as a bridge between inflammation and fibrosis in multiple organs

    • This pathway influences tissue repair mechanisms in inflammatory conditions

  • Wnt signaling pathway interactions:

    • IL-36γ stimulates autophagy and induces WNT5A expression

    • It activates the COX-2/AKT/mTOR pathway via noncanonical Wnt signaling

    • Wnt signaling inhibitors can blunt IL-36γ-driven inflammatory responses

These complex interactions suggest that targeting IL-36γ may have context-dependent effects depending on the disease state and inflammatory milieu.

What are the therapeutic implications of using recombinant IL-36γ or IL-36γ antagonists?

The development of IL-36γ-targeting therapeutics presents various opportunities and considerations:

What advanced analytical methods are being developed to study IL-36γ binding and activation mechanisms?

Cutting-edge approaches for investigating IL-36γ molecular mechanisms include:

  • TR-FRET displacement assays:

    • Novel Time-Resolved Fluorescence Resonance Energy Transfer methods measure IL-36γ binding to IL-36R/IL-1RAcP heterodimer

    • Allow high-throughput screening of potential antagonists

    • Enable quantitative measurement of binding kinetics and affinities

  • 3D skin equivalent models:

    • Complex in vitro systems combining keratinocytes and fibroblasts

    • Enable assessment of IL-36γ effects in physiologically relevant contexts

    • Allow evaluation of both cytokine release and tissue morphology changes

  • Protease activity profiling:

    • Identification of specific proteases that cleave and activate IL-36γ

    • Determination of precise cleavage sites and resulting activity modulation

    • Understanding the role of neutrophil-, APC-, and epithelial cell-derived enzymes in IL-36γ processing

  • 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.

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.