Recombinant Human Interleukin-36 alpha protein (IL36A) (Active)

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Lyophilized from a 0.2 µm filtered 2x PBS solution, pH 7.4.
Form
Lyophilized powder
Lead Time
5-10 business days
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to collect the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting the solution at -20°C/-80°C. Our standard protocol uses 50% glycerol. This can be used as a reference.
Shelf Life
Shelf life depends on various factors including storage conditions, buffer composition, temperature, and the inherent stability of the protein. Generally, the liquid form has a 6-month shelf life at -20°C/-80°C, while the lyophilized form has a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is necessary for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag-Free
Synonyms
FIL1; FIL1 epsilon; FIL1(EPSILON); FIL1E; IL 1 epsilon; IL 1F6; IL 1H1; IL-1 epsilon; IL-1F6; IL1E; IL1F6; IL1F6_HUMAN; IL1RP2; IL36 alpha; IL36A; Interleukin 1 epsilon; Interleukin 1 family member 6 (epsilon); Interleukin 1 family member 6; Interleukin 36 alpha; Interleukin-1 epsilon; Interleukin-1 family member 6; MGC129552; MGC129553; MGC151479; MGC151481; OTTMUSP00000012798; RP23-176J12.4
Datasheet & Coa
Please contact us to get it.
Expression Region
1-158aa
Mol. Weight
17.7 kDa
Protein Length
Full Length
Purity
>95% as determined by SDS-PAGE.
Research Area
Immunology
Source
E.coli
Species
Homo sapiens (Human)
Target Names
IL36A
Uniprot No.

Target Background

Function

Interleukin-36 alpha (IL-36α) is a cytokine that binds to and signals through the IL1RL2/IL-36R receptor. This interaction activates NF-κB and MAPK signaling pathways in target cells, triggering a pro-inflammatory response. IL-36α is part of the IL-36 signaling system, believed to be present in epithelial barriers and involved in localized inflammatory responses. Functionally similar to the IL-1 system, it shares the coreceptor IL1RAP. IL-36α appears to be involved in skin inflammation, acting on keratinocytes, dendritic cells, and indirectly on T-cells to induce tissue infiltration, cell maturation, and proliferation. In cultured keratinocytes, it induces the expression of chemokines (CCL3, CCL4, CCL5, CCL2, CCL17, CCL22, CL20, CCL5, CCL2, CCL17, CCL22, CXCL8, CCL20, and CXCL1) and proinflammatory cytokines (TNF-α, IL-8, and IL-6). In cultured monocytes, it upregulates the expression of IL-1α, IL-1β, and IL-6. In myeloid dendritic cells, it promotes cell maturation by upregulating surface expression of CD83, CD86, and HLA-DR. In monocyte-derived dendritic cells, it facilitates dendritic cell maturation and drives T-cell proliferation. IL-36α may also play a role in pro-inflammatory lung responses.

Gene References Into Functions
  • Elevated serum IL-36α levels are associated with active systemic lupus erythematosus, correlating with disease activity and arthritis. (PMID: 29571080)
  • IL-36α plays a significant role in pancreatic inflammation and fibrosis through an autocrine mechanism. (PMID: 28099250)
  • The IL-36R pathway is implicated in colonic inflammation and may represent a therapeutic target. (PMID: 26813344)
  • IL-36α inhibits the growth of epithelial ovarian cancer cells, suggesting therapeutic potential. (PMID: 28621240)
  • Increased Interleukin-36 expression is linked to Inflammatory Bowel Disease. (PMID: 26752465)
  • IL-36α exhibits pro-inflammatory effects on cartilage, increasing markers of inflammation and cartilage catabolism. (PMID: 26560022)
  • Elevated plasma concentrations of IL-36α and IL-36γ are observed in active systemic lupus erythematosus, with IL-36α exhibiting pro-inflammatory effects through IL-6 and CXCL8 regulation. (PMID: 26516833)
  • IL-36α increases the maturation of monocyte-derived dendritic cells. (PMID: 25700962)
  • The IL36A-IL36R axis is modulated in patients with primary Sjogren's syndrome. (PMID: 25902739)
  • High IL-36α expression is found in colorectal cancer, with low expression correlating with poor prognosis. (PMID: 25550854)
  • IL-36α expression is pivotal in determining the prognosis of hepatocellular carcinoma (HCC). (PMID: 24061617)
  • IL-36α is expressed in psoriatic and rheumatoid arthritis synovium. (PMID: 23268368)
  • Increased IL-1F6 expression is observed in human plaque psoriasis and in lesional skin of transgenic mice. (PMID: 21242515)
  • A functional IL-6 polymorphism is weakly associated with bone mineral density and postmenopausal bone loss. (PMID: 12110411)
  • Dysregulated expression in transgenic mice promotes cutaneous inflammation, highlighting potential therapeutic targets for inflammatory skin disorders. (PMID: 17908936)
  • IL-1ε, an IL-1 agonist, activates NF-κB through IL-1 receptor-related protein 2, suggesting an independent signaling system in human epithelial barriers. (PMID: 11466363)
Database Links

HGNC: 15562

OMIM: 605509

KEGG: hsa:27179

STRING: 9606.ENSP00000259211

UniGene: Hs.278910

Protein Families
IL-1 family
Subcellular Location
Cytoplasm. Secreted.
Tissue Specificity
Expressed in immune system and fetal brain, but not in other tissues tested or in multiple hematopoietic cell lines. Predominantly expressed in skin keratinocytes but not in fibroblasts, endothelial cells or melanocytes. Increased in lesional psoriasis sk

Q&A

What is the structural composition of recombinant human IL-36α protein?

Recombinant human IL-36α protein is typically produced as a truncated form (amino acids 6-158) that exhibits enhanced biological activity compared to the full-length protein. The most common commercially available recombinant forms are produced in E. coli expression systems. The protein shares 57-68% amino acid sequence identity with mouse, rabbit, equine, and bovine IL-36α homologs, and 27-57% amino acid sequence identity with other IL-1 family members .

The active form of IL-36α is not the full-length protein but rather a processed form. IL-36 cytokines are expressed as inactive precursors that require proteolytic processing to become fully active . This processing is crucial for the protein's ability to bind effectively to its receptor and initiate downstream signaling cascades.

What are the primary expression patterns and cellular sources of IL-36α?

IL-36α demonstrates tissue-specific expression patterns primarily in:

  • Epithelial barriers (skin, bronchial epithelium, intestine)

  • Lymphoid tissues

  • Fetal brain

  • Trachea

  • Stomach

At the cellular level, IL-36α is expressed by:

  • Monocytes

  • B cells

  • T cells

  • Keratinocytes (particularly when activated)

Importantly, IL-36α expression can be induced by various stimuli. For instance, IL-17 and TNF can induce IL-36α expression in keratinocytes, with IL-22 synergizing this induction. Epidermal growth factor (EGF) also regulates IL-36α expression in the skin . In immune cells, Toll-like receptor (TLR) activation through various ligands can trigger IL-36α expression in a cell type-specific manner .

What is the receptor-mediated signaling pathway for IL-36α?

IL-36α signals through a receptor complex composed of:

  • IL-36 receptor (IL-36R, also known as IL-1Rrp2 or IL-1RL2) - primarily expressed in epithelial cells and keratinocytes

  • IL-1 receptor accessory protein (IL-1RAcP) - widely expressed co-receptor

Upon binding to this receptor complex, IL-36α activates:

  • Nuclear factor kappa B (NF-κB) signaling

  • Mitogen-activated protein kinase (MAPK) pathways

These signaling cascades ultimately lead to the induction of pro-inflammatory genes and cytokine production. The effective concentration (EC50) for IL-36α-induced responses varies by cell type and readout, typically ranging from 0.4-24 ng/mL .

How should researchers determine optimal concentrations for in vitro experiments with recombinant IL-36α?

When designing in vitro experiments with recombinant IL-36α, researchers should consider:

  • Cell type-specific sensitivity: Different cell types respond to IL-36α at varying concentrations. For example:

    • Human epidermoid carcinoma cells (A-431) respond to IL-36α with IL-8 production at an ED50 of 0.4-2.4 ng/mL

    • Other cell systems may require concentrations of 4-24 ng/mL for observable effects

  • Experimental readout: The concentration required may vary based on the specific endpoint being measured:

    • For NF-κB activation assays: Start with concentrations of 1-10 ng/mL

    • For cytokine induction studies: 5-50 ng/mL is typically effective

    • For cellular phenotypic changes: Higher concentrations (10-100 ng/mL) may be necessary

  • Methodology for determining optimal concentration:

    • Perform dose-response curves using 2-5 fold serial dilutions

    • Include positive controls (such as IL-1β or TNFα) for comparison

    • Assess multiple time points (4, 8, 24 hours) as response kinetics may vary

    • Measure multiple outputs when possible (e.g., both mRNA using qRT-PCR and protein using ELISA)

What cellular and molecular assays are most appropriate for evaluating IL-36α activity?

Several complementary assays can effectively measure IL-36α activity:

Gene Expression Assays:

  • Quantitative RT-PCR targeting downstream genes including IL-8, IL-6, CXCL1, CXCL2, CXCL10, S100A8/A9, and β-defensins

  • RNA-seq for genome-wide assessment of transcriptional responses

Protein Production Assays:

  • ELISA for quantification of induced cytokines/chemokines

  • Multiplexed cytokine arrays for broader profiling

  • Western blotting for signaling protein phosphorylation (p-p38, p-ERK, p-JNK, IκB degradation)

Functional Cellular Assays:

  • Cell migration assays to assess chemotactic responses

  • Reporter cell lines with NF-κB or AP-1 responsive elements

  • Flow cytometry to assess surface marker changes

Ex Vivo Tissue Models:

  • Human skin biopsies or reconstructed 3D skin models can be used to evaluate IL-36α effects in a complex tissue environment

  • Gene expression analysis using skin biopsies can effectively demonstrate IL-36 pathway activation or inhibition

How can researchers effectively validate IL-36α specificity in their experiments?

To ensure experimental results are specifically attributable to IL-36α activity:

  • Include appropriate controls:

    • Use heat-inactivated IL-36α protein

    • Include the IL-36 receptor antagonist (IL-36Ra) to block specific signaling

    • Use isotype-matched irrelevant recombinant proteins

  • Receptor validation approaches:

    • Employ IL-36R knockdown/knockout models (siRNA, CRISPR-Cas9)

    • Use blocking antibodies against IL-36R

    • Test newly developed small molecule IL-36R antagonists like 36R-P192 or 36R-D481

  • Genetic validation:

    • Compare responses in wild-type vs. IL-36R-deficient cells

    • Use cells from different sources with confirmed IL-36R expression levels

  • Downstream signaling verification:

    • Monitor NF-κB and MAPK pathway activation

    • Compare signaling kinetics with other IL-1 family members

How does IL-36α contribute to inflammatory skin conditions, particularly psoriasis?

IL-36α plays a critical role in inflammatory skin pathologies:

  • Expression pattern in psoriasis:

    • IL-36α is significantly upregulated in psoriatic lesions

    • Activated keratinocytes are a primary source of IL-36 cytokines in inflamed skin

    • Creates an amplification loop where IL-36α stimulates further cytokine production

  • Mechanistic contributions:

    • Activates keratinocytes to produce additional pro-inflammatory mediators

    • Recruits and activates immune cells including neutrophils and T cells

    • Promotes tissue remodeling through induction of matrix metalloproteinases

    • Synergizes with other cytokines (IL-17, TNF) to enhance inflammatory responses

  • Therapeutic implications:

    • Targeting the IL-36 pathway may provide benefit in psoriasis treatment

    • Small molecule IL-36R antagonists like 36R-P192 effectively suppress IL-36-induced gene expression in human skin biopsies

    • Combines with existing psoriasis treatments may offer synergistic benefits

What roles does IL-36α play in lung inflammation and respiratory diseases?

IL-36α is increasingly recognized as an important mediator in pulmonary inflammation:

  • Expression in respiratory tissues:

    • Bronchial epithelial cells and pulmonary macrophages produce IL-36α in response to pathogen-associated molecular patterns

    • Toll-like receptor (TLR) stimulation induces IL-36α expression in lung cells

  • Functional effects in the lung:

    • Intratracheal administration of IL-36α induces CXCL1 and CXCL2 expression

    • Promotes neutrophil recruitment to the lungs

    • Activates lung fibroblasts

    • May contribute to both protective anti-microbial responses and pathological inflammation

  • Research implications:

    • IL-36α levels may serve as biomarkers for specific lung pathologies

    • Targeting IL-36 signaling could provide therapeutic benefit in certain pulmonary inflammatory conditions

    • Understanding the balance between IL-36α-mediated protection versus pathology is crucial for therapeutic development

What is the current understanding of IL-36α's role in kidney inflammation and injury?

IL-36α has emerged as a significant factor in renal pathology:

  • Expression and regulation in kidney tissue:

    • IL-36R is expressed in kidney tissue, particularly in proximal tubules

    • IL-36α expression increases significantly following kidney injury

  • Experimental evidence from kidney disease models:

    • In renal ischemia-reperfusion models, IL-36R depletion protects against kidney inflammation

    • IL-36α expression increases within 24 hours of unilateral ureter obstruction (UUO)

    • IL-36α levels correlate with kidney dysfunction in folic acid-induced acute kidney injury models

    • In chronic glomerulonephritis models, IL-36α levels correlate with tubular damage severity and renal interstitial fibrosis

  • Cellular mechanisms:

    • In renal tubular epithelial cells, IL-36α treatment increases NF-κB activity and ERK phosphorylation

    • Contributes to inflammatory cytokine production in the kidney microenvironment

  • Clinical implications:

    • IL-36α is consistently upregulated in various mouse models of kidney disease

    • May serve as a biomarker for earlier detection of kidney injuries

    • Represents a potential therapeutic target for kidney inflammatory conditions

How does the proteolytic processing of IL-36α affect its bioactivity, and what are the key methodological considerations?

The proteolytic processing of IL-36α is critical for its full biological activity:

  • Processing requirements:

    • IL-36 cytokines are expressed as inactive precursors requiring proteolytic processing to attain full activity

    • Proper N-terminal processing significantly enhances receptor binding and biological activity

    • Commercial recombinant preparations typically use the truncated form (aa 6-158) to maximize activity

  • Methodological implications for researchers:

    • When designing experiments, researchers should:

      • Verify the form of IL-36α being used (full-length vs. truncated)

      • Consider that full-length IL-36α may show substantially less activity

      • Account for endogenous processing that may occur in complex biological systems

      • Consider co-expression or treatment with relevant proteases if working with full-length protein

  • Research directions:

    • Identification of specific proteases responsible for IL-36α processing in different tissues

    • Development of processing-resistant forms for mechanistic studies

    • Investigation of how processing is regulated in different disease states

What are the molecular mechanisms underlying the differential effects of IL-36α versus other IL-36 family members?

Despite signaling through the same receptor complex, IL-36α, IL-36β, and IL-36γ exhibit distinct biological effects:

  • Sequence and structural differences:

    • IL-36α shares only partial sequence homology with IL-36β and IL-36γ

    • These differences likely affect receptor binding kinetics and downstream signaling intensity

  • Expression pattern variations:

    • Cell type-specific expression of different IL-36 family members

    • Differential regulation of IL-36α vs other family members:

      • TLR stimulation induces different IL-36 family members depending on cell type and specific TLR ligand

      • IL-36α and IL-36γ, but not IL-36β, are induced in bronchial epithelial cells by various TLR ligands

  • Functional distinctions:

    • While all IL-36 agonists activate NF-κB and MAPK pathways, they may do so with different kinetics or magnitude

    • Tissue-specific responses may vary due to differential receptor expression or co-factors

  • Research design considerations:

    • Comparative studies should include multiple IL-36 family members

    • Dose-matching is essential when comparing effects between family members

    • Analysis of signaling kinetics rather than single time points may reveal differences

How can researchers effectively study IL-36α in the context of complex disease models?

Studying IL-36α in complex disease systems requires sophisticated approaches:

  • Animal model selection and development:

    • Consider species-specific differences in IL-36 signaling

    • Humanized mouse models may be necessary for certain applications

    • Tissue-specific conditional knockout models provide advantages over global knockouts

  • Combination treatments and pathway interactions:

    • IL-36α rarely acts alone in disease states

    • Consider combination treatments with other cytokines (IL-17, TNF, IL-22) that synergize with IL-36α

    • Study pathway crosstalk, particularly between IL-36 and other IL-1 family members

  • Translational approaches:

    • Ex vivo culture of patient-derived samples with IL-36α

    • Correlation of IL-36α levels with disease parameters in patient cohorts

    • Evaluation of genetic variations in IL-36 pathway components

  • Opposing roles of IL-36 pathway components:

    • The opposing effects of IL-1Ra and IL-36Ra must be considered in experimental design

    • In corneal inflammation models, IL-1Ra improves outcomes while IL-36Ra worsens them

    • Administration of IL-36γ stimulates expression of innate defense molecules but suppresses IL-1β expression

What is the current understanding of the role of IL-36α in cancer biology?

The role of IL-36α in cancer development represents an emerging research area:

  • Expression patterns in cancer:

    • Decreased IL-36α expression correlates with poor prognosis in hepatocellular carcinoma

    • Expression patterns vary across cancer types, with some showing upregulation and others downregulation

  • Potential mechanisms in tumor biology:

    • May influence tumor microenvironment through recruitment and activation of immune cells

    • Limited studies suggest potential anti-tumor effects

    • IL-36α may affect cancer cell proliferation, invasion, and response to therapy

    • Could have context-dependent pro- or anti-tumor effects depending on cancer type and stage

  • Research opportunities:

    • Characterizing IL-36α expression across different cancer types and stages

    • Investigating the impact of IL-36α on tumor-infiltrating immune cells

    • Exploring potential applications in cancer immunotherapy

    • Studying the interaction between IL-36α and established cancer-associated inflammatory pathways

  • Methodological approaches:

    • Cancer cell line studies with recombinant IL-36α treatment

    • IL-36α overexpression or knockout in tumor models

    • Analysis of patient tumor samples for IL-36 pathway component expression

    • Correlation of IL-36α levels with response to immunotherapy

How is IL-36α gene expression regulated at the transcriptional level?

Understanding the transcriptional regulation of IL-36α is crucial for experimental design and interpretation:

  • Transcription factor involvement:

    • C/EBPβ binds specifically to an essential half-CRE- C/EBP motif in the IL-36α promoter

    • This binding is crucial for promoter activation in the cellular context

  • Epigenetic regulation:

    • DNA methylation influences IL-36α expression

    • Different cell types show varying methylation levels at the half-CRE- C/EBP site

    • This may explain cell type-specific expression patterns

  • Induction mechanisms:

    • Lipopolysaccharide (LPS) stimulation induces IL-36α expression in multiple cell types

    • In RAW264.7 cells and bone marrow-derived macrophages (BMDMs), LPS triggers IL-36α expression despite different methylation levels at regulatory sites

    • Multiple TLR ligands can induce IL-36α expression with cell type-specific patterns

  • Research applications:

    • Promoter analysis can help identify key regulatory elements

    • Understanding transcriptional regulation provides insight into targeting IL-36α expression therapeutically

    • Cell-specific expression patterns should inform experimental design

What are the latest therapeutic approaches targeting the IL-36 pathway?

Recent advances in targeting the IL-36 pathway include:

  • Small molecule inhibitors:

    • Development of low molecular weight (<1000 Da) IL-36R antagonists

    • Compound 36R-D481, identified through DNA encoded libraries (DEL) screening, effectively inhibits IL-36 signaling

    • X-ray crystallography shows these molecules bind to the IL-36R's D1 domain, potentially disrupting IL-36 cytokine binding

  • Macrocyclic peptides:

    • 36R-P138, identified through mRNA-based display technique, blocks IL-36R signaling

    • Optimized analog 36R-P192 effectively suppresses expression of marker genes induced by IL-36 in human skin biopsies

  • Receptor antagonist applications:

    • Recombinant IL-36Ra for therapeutic applications

    • Understanding the opposing effects of IL-1Ra and IL-36Ra in different disease models

  • Combination approaches:

    • Targeting multiple inflammatory pathways simultaneously

    • Combining IL-36 pathway inhibition with established therapies

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.