IL36B Mouse

Interleukin-36 Beta Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

Role in Inflammatory Responses

IL36B amplifies neutrophilic inflammation by upregulating CXCL1, IL-1β, and GM-CSF in macrophages and dendritic cells. In Il36r−/− mice, neutrophil recruitment is reduced by 47–61% in models of cigarette smoke (CS) and H1N1 influenza-induced lung inflammation, with concurrent decreases in IL-1β and CXCL1 .

Metabolic Regulation

Genetic deletion of Il36rn (IL-36 receptor antagonist) in mice suppresses adipose tissue accumulation and improves glucose tolerance, even under high-fat diet (HFD) conditions. This suggests IL-36 signaling promotes obesity and insulin resistance .

ModelOutcomeSource
Chow-fed Il36rn−/−Reduced weight gain, lower epididymal/subcutaneous fat, improved glucose tolerance
HFD-fed Il36rn−/−Attenuated adiposity, enhanced hepatic insulin sensitivity (Akt phosphorylation)

Epithelial Wound Healing

IL36R deficiency accelerates corneal epithelial wound closure in normoglycemic mice, suggesting IL-36 signaling may impede healing by upregulating pro-inflammatory cytokines like lipocalin-2 (LCN2) .

Metabolic Interventions

While Il36rn deficiency improves metabolic outcomes, therapeutic blockade of IL-36R may compromise innate immunity, as IL-1α/β signaling through IL-1RAP is preserved .

Comparative Analysis with Other IL-36 Cytokines

IL36B shares functional redundancy with IL-36α, β, γ, but exhibits distinct tissue-specific roles:

CytokinePrimary SourcesKey FunctionsRelevance to IL36B
IL-36αKeratinocytes, neutrophilsDrives epidermal hyperplasia in psoriasisAmplifies IL36B’s pro-inflammatory effects
IL-36γNeutrophils, macrophagesPromotes neutrophil recruitment in lung inflammationShares receptor (IL-36R)
IL-36RaEpithelial cellsAntagonizes IL-36 signalingTherapeutic target for IL36B-driven diseases

Product Specs

Introduction
Mouse IL-36b, a member of the IL-1 family, shares structural similarities with IL-1b, IL-1a, IL-1ra, IL-18, IL-36ra (IL1F5), IL-36g (IL1F9), IL-37 (IL1F7), and IL-38 (IL-1F10). These family members, characterized by a 12 β-strand, β-trefoil configuration, are believed to have evolved from a common ancestor. Mouse IL-36b/IL-1F8, an 183-amino acid protein, lacks a signal sequence, prosegment, and potential N-linked glycosylation sites. Similar to its human counterpart, Mouse IL-36b is actively secreted. It exhibits 61-74% amino acid sequence homology with human IL-36?/IL-1F8 isoform 2 and rat IL-1F8.
Description
Recombinant Mouse IL36B, produced in E. coli, is a single, non-glycosylated polypeptide chain consisting of 183 amino acids. With a molecular weight of 21.0 kDa, it is purified using proprietary chromatographic techniques.
Physical Appearance
Sterile Filtered White lyophilized powder.
Formulation
Lyophilized from a 0.2µm filtered concentrated solution in 1xPBS, pH 7.4.
Solubility
Reconstitute the lyophilized IL36B in sterile 18M-cm H₂O at a concentration of at least 100µg/ml. This solution can be further diluted in other aqueous solutions.
Stability
Lyophilized Mouse IL36B remains stable at room temperature for 3 weeks. However, it should be stored desiccated below -18°C. After reconstitution, store IL36B at 4°C for 2-7 days. For long-term storage, keep it below -18°C. Avoid repeated freeze-thaw cycles.
Purity
Purity exceeds 95.0% as determined by SDS-PAGE.
Biological Activity
Biological activity is assessed through its binding affinity to recombinant mouse IL-1 Rrp2 in a functional ELISA.
Synonyms
Interleukin 36 beta, interleukin 1 family member 8 (eta), Interleukin-1 homolog 2, IL1F8 (Canonical product IL-1F8a), IL-1F8 (FIL1-eta), Interleukin-1 Superfamily e, IL1H2, MGC126880, MGC126882.
Source
Escherichia Coli.
Amino Acid Sequence
MMAFPPQSCV HVLPPKSIQM WEPNHNTMHG SSQSPRNYRV HDSQQMVWVL TGNTLTAVPA SNNVKPVILS LIACRDTEFQ DVKKGNLVFL GIKNRNLCFC CVEMEGKPTL QLKEVDIMNL YKERKAQKAF LFYHGIEGST SVFQSVLYPG WFIATSSIER QTIILTHQRG KLVNTNFYIE SEK

Q&A

What is IL-36β and how does it function in the mouse immune system?

IL-36β is one of three agonistic cytokines within the IL-36 subfamily (alongside IL-36α and IL-36γ) of the larger IL-1 cytokine family. In mice, IL-36β functions as a pro-inflammatory mediator that signals through the IL-36 receptor (IL-36R) and requires IL-1RAcP as a co-receptor. Upon binding, this complex initiates downstream signaling primarily through activation of the NF-κB and MAPK pathways, resulting in the production of inflammatory mediators and chemokines that recruit immune cells to sites of inflammation . The mouse IL36B gene is located on chromosome 2 as part of a genomic cluster that includes other IL-1 family members .

How does mouse IL-36β differ structurally and functionally from other IL-36 family members?

While all three IL-36 agonists (α, β, and γ) signal through the same receptor complex, they exhibit differences in expression patterns and potency. In mouse models, IL-36β shares approximately 65-70% amino acid sequence identity with IL-36α and IL-36γ. All three require N-terminal processing for full biological activity, though the specific proteases responsible may differ. Expression analysis shows that IL-36β and IL-36γ are often co-expressed in response to inflammatory stimuli in epithelial cells, while their relative potency in activating downstream signaling pathways can vary by cell type and context .

What is known about the regulation of IL-36β expression in mice?

Expression of IL-36β mRNA is induced in mouse epithelial cells in response to pro-inflammatory stimuli including IL-1β, TNF, PMA, and bacterial components like flagellin. Notably, a synergistic effect occurs when TNF is combined with either PMA or flagellin, resulting in significantly enhanced IL-36β expression. Unlike some cytokines, IL-36β does not appear to induce its own expression in an autocrine manner in mouse models. At the transcriptional level, regulation involves NF-κB-dependent mechanisms, while post-translational regulation includes proteolytic processing of the inactive precursor to generate the fully active form .

What mouse models are most appropriate for studying IL-36β functions?

Several mouse models have proven valuable for studying IL-36β biology:

For acute inflammation models, cigarette smoke exposure with or without H1N1 influenza virus has shown significant differences between wild-type and IL-36R-deficient mice, making it a useful model to study IL-36 family members including IL-36β .

What are the optimal methods for detecting IL-36β expression in mouse tissues?

Detection of IL-36β in mouse tissues requires specialized approaches due to its relatively low expression levels under baseline conditions:

  • mRNA detection: RT-qPCR remains the gold standard for quantifying IL-36β transcripts, with appropriate normalization to housekeeping genes. Primers should be designed to distinguish between IL-36β and other IL-36 family members due to sequence similarity .

  • Protein detection: Western blotting using specific anti-IL-36β antibodies, often requiring immunoprecipitation to enrich for the protein. Importantly, researchers should consider the form being detected (full-length versus processed) as this affects biological activity .

  • Immunohistochemistry/Immunofluorescence: For tissue localization, though signal amplification may be necessary. Controls using IL-36β knockout tissue are essential to confirm specificity .

  • Reporter systems: When available, reporter mice expressing fluorescent proteins under the IL-36β promoter provide valuable tools for visualizing expression patterns .

For optimal results, stimulation with appropriate inducers (TNF plus PMA or bacterial components) is recommended when studying IL-36β expression in experimental settings .

How can researchers effectively generate and validate recombinant mouse IL-36β for experimental studies?

Generating biologically active recombinant mouse IL-36β requires specific considerations:

  • Expression system selection: Bacterial systems (E. coli) can produce high yields but lack post-translational modifications. Mammalian expression systems may provide better folding but lower yields.

  • N-terminal processing: Critical for activity, as full-length IL-36β has significantly lower biological activity. Express truncated forms beginning at key residues (equivalent to human n6 or n5 positions) or perform controlled proteolytic processing of full-length protein .

  • Purification strategy: Affinity tags (His or GST) followed by size exclusion chromatography to ensure homogeneity.

  • Activity validation: Functional testing using reporter cell lines expressing the mouse IL-36 receptor and an NF-κB reporter construct. The HT-29 cell line stably transfected with an NF-κB reporter gene has been used successfully for this purpose .

  • Endotoxin removal: Critical for in vivo applications to prevent confounding inflammatory responses.

EC50 determination using dose-response curves is essential for comparing potency between different preparations and forms of IL-36β .

How do neutrophil-derived proteases regulate IL-36β activity in mouse models of inflammation?

Neutrophils play a dual role in IL-36β biology in mouse inflammation models - both as sources of IL-36 cytokines and as providers of proteases that can process and activate IL-36β. Research suggests that neutrophil serine proteases, including cathepsin G, elastase, and proteinase-3, can cleave the N-terminus of IL-36β, potentially at different sites than those identified through in vitro studies .

Methodologically, researchers investigating this interaction should:

  • Use neutrophil depletion models (anti-Ly6G antibody treatment) to assess impact on IL-36β processing and activity

  • Employ specific protease inhibitors in vivo and ex vivo to identify which neutrophil proteases are primarily responsible

  • Generate site-directed mutagenesis variants of mouse IL-36β with altered predicted cleavage sites to determine processing specificity

  • Use mass spectrometry to identify precise cleavage products in inflammatory exudates from mouse models

This research direction is particularly relevant for understanding the amplification of neutrophilic inflammation in conditions like COPD, where IL-36β processing may create positive feedback loops .

What are the molecular mechanisms by which IL-36β synergizes with other inflammatory mediators in mouse models?

IL-36β exhibits significant synergistic effects with other inflammatory mediators, particularly GM-CSF and viral mimics like poly(I:C), to amplify inflammatory responses in mouse models . The molecular basis for this synergy involves:

  • Receptor cross-talk: IL-36R signaling may enhance expression or signaling capacity of receptors for other cytokines through shared adapter molecules like MyD88

  • Convergent downstream pathways: Both IL-36β and GM-CSF activate overlapping but distinct aspects of the NF-κB and MAPK pathways, potentially leading to enhanced and sustained activation

  • Cooperative transcription factor activation: Combined stimulation may recruit additional transcription factors or coactivators to inflammatory gene promoters

To investigate these mechanisms, researchers should employ:

  • Phospho-proteomic analysis to identify shared and distinct signaling nodes

  • ChIP-seq to map transcription factor binding patterns under single versus combined stimulation

  • Proximity ligation assays to detect physical interactions between receptor components

  • Single-cell RNA-seq to identify cell populations that are particularly responsive to the synergistic effects

Understanding this synergy is particularly important in mixed inflammatory environments such as viral-bacterial co-infections of the lung .

What is the temporal relationship between IL-36β expression and neutrophil recruitment in acute versus chronic mouse inflammation models?

The temporal dynamics between IL-36β expression and neutrophil recruitment represent a complex relationship with potential feedback loops. In acute inflammation models:

  • Initial inflammatory triggers (pathogens, tissue damage) induce IL-36β expression primarily from epithelial cells

  • IL-36β signaling promotes chemokine production (particularly CXCL1 and CXCL2 in mice)

  • Neutrophils are recruited to the inflammatory site

  • Recruited neutrophils can both produce additional IL-36β and process existing IL-36β to more active forms

  • This creates a potential amplification circuit

In chronic models, such as repeated cigarette smoke exposure, this relationship becomes more complex, with:

  • Sustained IL-36β expression from structural cells

  • Persistent neutrophil recruitment and activation

  • Tissue remodeling that may alter the cellular sources and responsiveness to IL-36β

Appropriate experimental approaches include:

  • Time-course analyses with frequent sampling in both acute and chronic models

  • Cell-specific conditional knockouts to determine the relative contribution of epithelial versus neutrophil-derived IL-36β

  • Neutrophil transfer experiments using labeled cells to track their activation and cytokine production

How can researchers effectively distinguish between the biological activities of different IL-36 family members in mouse models?

Distinguishing the specific contributions of IL-36β from other family members presents a significant challenge due to shared receptors and similar biological activities. Advanced methodological approaches include:

  • Selective neutralization: Using highly specific monoclonal antibodies against mouse IL-36β that don't cross-react with IL-36α or IL-36γ

  • Cytokine replacement studies: Using IL-36β knockout mice reconstituted with recombinant IL-36β versus other family members

  • Receptor mutants: Developing receptor variants with altered affinity for specific IL-36 family members

  • Differential expression analysis: Comprehensive temporal profiling of all IL-36 family members during inflammatory responses to identify unique expression patterns

  • Conditional deletion models: Cell-type specific deletion of IL-36β to determine tissue-specific roles

What are the most reliable protocols for studying IL-36β processing in mouse tissues and cell cultures?

Studying IL-36β processing in biological samples requires specialized techniques due to the low abundance of processed forms and technical challenges in their detection:

  • Ex vivo processing assays: Incubate recombinant full-length mouse IL-36β with cellular fractions or purified enzymes, then analyze by:

    • Western blotting with antibodies specific to different regions

    • Mass spectrometry to identify precise cleavage sites

    • Functional assays using reporter cell lines to assess activity

  • In vivo processing detection:

    • Immunoprecipitation with N-terminal versus C-terminal antibodies

    • Differential extraction protocols to enhance recovery of processed forms

    • Analysis of biological fluids (BAL, inflammatory exudates) by western blotting or mass spectrometry

  • Live-cell imaging approaches:

    • Split fluorescent protein constructs that change localization or FRET properties upon processing

    • Activity-based probes that detect active proteases capable of processing IL-36β

A common methodological error is attempting to induce processing with inappropriate stimuli. Research indicates that classical inflammatory stimuli (TNF, IL-1) induce expression but not necessarily processing of IL-36β, while certain apoptotic stimuli (cycloheximide, staurosporine) may induce mobility changes suggesting processing .

How should researchers address the variability in IL-36β responses across different mouse strains?

Strain-dependent variability in IL-36β responses represents an important consideration in experimental design. Key methodological recommendations include:

  • Strain selection considerations:

    • C57BL/6 mice: Most commonly used, with well-characterized IL-36 pathway components

    • BALB/c mice: May exhibit different T helper cell polarization affecting IL-36β responses

    • Background matching: Ensure knockout and control mice are on identical genetic backgrounds

  • Experimental controls and reporting:

    • Always report complete strain information including substrain (e.g., C57BL/6J versus C57BL/6N)

    • Include wild-type littermate controls whenever possible

    • Consider using multiple strains for critical findings to establish generalizability

  • Technical approaches to address variability:

    • Increased sample sizes based on power calculations from preliminary data

    • Mixed-effects statistical models that account for strain as a variable

    • Ex vivo validation using cells derived from different strains

  • Mechanistic investigation of strain differences:

    • Sequence the IL-36β gene and its promoter regions across strains to identify polymorphisms

    • Compare IL-36 receptor expression levels and signaling responses

    • Evaluate strain-dependent differences in protease expression that might affect processing

Understanding strain variability not only improves experimental reproducibility but may also provide insights into genetic factors influencing IL-36β biology relevant to human disease heterogeneity.

What are the key differences between mouse and human IL-36β that researchers should consider when designing translational studies?

Understanding the species-specific differences in IL-36β biology is crucial for translational research. Key comparative aspects include:

FeatureMouse IL-36βHuman IL-36βResearch Implications
Amino acid sequence100% (reference)~65-70% identity to mouseMay affect cross-reactivity of reagents
Optimal N-terminal processing siteSimilar to human but may have species-specific preferencesActive form begins at K6 (n6-IL-36β)Different proteases may be involved in processing
Expression patternBroadly similar to human but with some tissue-specific differencesHighest in skin, lung, gut epitheliaConsider tissue relevance in model selection
Receptor binding affinitySimilar potency rangeEC50 in nanomolar rangeComparable dose-response relationships
Gene regulationIL36B induced by TNF, flagellin, PMASimilar inducers but may have human-specific factorsStimulus selection for experiments

For translational research, humanized mouse models expressing human IL-36 pathway components provide valuable tools to bridge these species differences. Additionally, parallel studies using both mouse and human primary cells are recommended to validate key findings across species .

How can mouse models of IL-36β biology inform therapeutic targeting strategies for human inflammatory diseases?

Mouse models have provided valuable insights for therapeutic targeting of the IL-36 pathway, with several key translational implications:

  • Target identification and validation:

    • IL-36R antagonism: Mouse models of psoriasiform inflammation and neutrophilic lung inflammation support IL-36R as a therapeutic target

    • Processing inhibition: Understanding of mouse IL-36β processing suggests targeting specific proteases may prevent IL-36β activation

    • Signaling modulation: Mouse studies reveal key nodes in IL-36β signaling that might be targetable

  • Therapeutic modality selection:

    • Biologics: Monoclonal antibodies against IL-36β or IL-36R validated in mice have translational potential

    • Small molecules: Inhibitors of processing enzymes or downstream signaling components

    • Cell-based therapies: Approaches targeting IL-36β-producing or responsive cells

  • Biomarker development:

    • Mouse studies suggest processed forms of IL-36β or downstream products as potential biomarkers

    • Neutrophil activation markers correlate with IL-36 activity in mouse models

    • Combined biomarker panels may predict responsiveness to IL-36 pathway inhibition

  • Potential clinical applications:

    • Pustular psoriasis: Strong genetic evidence links IL-36 pathway to human disease

    • COPD/neutrophilic asthma: Mouse models suggest IL-36β as an amplifier of neutrophilic inflammation

    • Inflammatory bowel disease: Emerging evidence for IL-36β in intestinal inflammation

The role of IL-36β as an upstream amplifier of neutrophilic inflammation in mouse models provides particular rationale for targeting this pathway in human neutrophil-mediated diseases .

What are the appropriate considerations when attempting to replicate human IL-36β-associated disease phenotypes in mouse models?

Effective disease modeling requires careful consideration of species differences and methodological approaches:

  • Anatomical and physiological considerations:

    • Skin differences: Mouse skin is thinner with different hair follicle density affecting psoriasiform models

    • Lung architecture: Fewer airway generations in mice affecting respiratory disease models

    • Immune system: Different baseline neutrophil percentages and inflammatory mediator profiles

  • Disease induction strategies:

    • Genetic approaches: Overexpression of processed IL-36β or deletion of IL-36Ra

    • Environmental triggers: Cigarette smoke, TLR ligands, or combined stimuli that better recapitulate complex human exposures

    • Humanized models: Reconstitution with human immune cells or expression of human IL-36 pathway components

  • Assessment parameters:

    • Comprehensive phenotyping: Beyond gross pathology to molecular and cellular parameters

    • Temporal dynamics: Acute versus chronic phase analysis

    • Therapeutic response: Testing standard-of-care human treatments as benchmarks

  • Limitations and interpretations:

    • Acknowledge differences in disease time course between mice and humans

    • Consider that successful treatment in mice may overpredict human efficacy

    • Validate key findings across multiple model systems when possible

Combined environmental exposures (such as cigarette smoke plus viral infection) often better recapitulate the complexity of human disease than single-trigger models, particularly for IL-36β-mediated conditions with multiple inflammatory inputs .

What emerging technologies are most promising for advancing our understanding of IL-36β biology in mouse models?

Several cutting-edge technologies offer significant potential for advancing IL-36β research:

  • Single-cell multi-omics:

    • scRNA-seq to identify specific cellular sources and responders to IL-36β

    • Single-cell proteomics to detect low-abundance processed forms of IL-36β

    • Spatial transcriptomics to map IL-36β expression in tissue microenvironments

  • CRISPR-based technologies:

    • Base editing for precise modification of processing sites or regulatory elements

    • CRISPRi/CRISPRa for temporal control of IL-36β expression

    • CRISPR screening to identify novel regulators of IL-36β processing or signaling

  • Advanced imaging approaches:

    • Intravital microscopy with fluorescent reporter systems to track IL-36β expression in live animals

    • Multiplexed imaging mass cytometry to simultaneously visualize multiple IL-36 pathway components

    • Label-free imaging techniques to assess tissue structural changes in response to IL-36β

  • Organoid and microphysiological systems:

    • Mouse-derived epithelial organoids for studying IL-36β in controlled tissue-like environments

    • Organ-on-chip systems incorporating multiple cell types for complex interaction studies

    • Co-culture systems with defined cellular components to dissect intercellular communication

These technologies promise to provide unprecedented resolution in understanding the spatiotemporal dynamics of IL-36β expression, processing, and signaling in complex inflammatory environments .

How might the study of IL-36β in mouse models contribute to our understanding of inflammatory disease heterogeneity?

The IL-36 pathway represents an important axis for understanding inflammatory disease heterogeneity. Mouse models can contribute in several ways:

  • Identification of inflammatory endotypes:

    • IL-36β-high versus IL-36β-low inflammatory responses may define distinct disease subsets

    • Different processing mechanisms may contribute to disease variation

    • Distinct cellular sources of IL-36β might characterize different disease phenotypes

  • Genetic and environmental interaction studies:

    • Models examining how IL-36β interacts with other genetic risk factors

    • Investigation of how environmental exposures modulate IL-36β responses

    • Development of combinatorial models that better reflect human disease complexity

  • Response prediction:

    • Identification of biomarkers that predict responsiveness to IL-36 pathway inhibition

    • Characterization of compensatory mechanisms in IL-36β-deficient conditions

    • Understanding of resistance mechanisms to IL-36 pathway targeting

  • Precision medicine approaches:

    • Definition of IL-36β-dependent versus IL-36β-independent disease variants

    • Stratification markers for patient selection in clinical trials

    • Combination therapy approaches based on mechanistic understanding

Mouse models incorporating genetic diversity (such as the Collaborative Cross or Diversity Outbred mice) may be particularly valuable for capturing the heterogeneity relevant to human disease .

Product Science Overview

Introduction

Interleukin-36 beta (IL-36β), also known as IL-1F8, is a member of the interleukin-1 (IL-1) superfamily, which includes several cytokines involved in inflammatory and immune responses. IL-36β is one of the three agonists in the IL-36 family, alongside IL-36α and IL-36γ, and it plays a crucial role in the regulation of immune responses and inflammation .

Structure and Function

IL-36β is a protein composed of 183 amino acids and lacks a signal sequence, prosegment, and potential N-linked glycosylation sites . The IL-1 family members, including IL-36β, display a 12 β-strand, β-trefoil configuration, which is thought to have evolved from a common ancestral gene . IL-36β is actively secreted by cells such as resting and activated monocytes and B cells .

The receptor for IL-36β is a combination of IL-1 receptor-related protein 2 (IL-1Rrp2) and IL-1 receptor accessory protein (IL-1RAcP) . Upon binding to its receptor, IL-36β activates intracellular signaling pathways involving nuclear factor-kappa B (NF-κB) and mitogen-activated protein kinase (MAPK), leading to the regulation of target genes and the induction of inflammatory responses .

Biological Role

IL-36β is involved in various physiological and pathological processes. It is expressed at low levels under normal conditions but is upregulated in response to inflammatory stimuli . IL-36β acts on a variety of cells, including epithelial and immune cells, and contributes to host defense through the inflammatory response . In the skin, IL-36β plays a role in the pathogenesis of psoriasis by stimulating keratinocytes and immune cells, enhancing the Th17/Th23 axis, and inducing psoriatic-like skin disorders .

In addition to its role in the skin, IL-36β is also expressed in other organs such as the lungs, intestines, joints, and brain upon stimulation . However, its physiological and pathological roles in these organs are less well-defined compared to its role in the skin .

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.