IL36A Mouse

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

Description

Introduction to IL36A Mouse

Interleukin-36 alpha (IL36A), also termed IL-1 family member 6 (IL-1F6), is a pro-inflammatory cytokine within the IL-1 family. It plays critical roles in immune regulation, tissue inflammation, and disease pathogenesis. In murine models, IL36A is studied for its involvement in lung inflammation, psoriasis, and autoimmune diseases. This article synthesizes molecular, functional, and preclinical data on IL36A Mouse, emphasizing its structural properties, biological activities, and research applications.

Sequence Homology

Mouse IL36A shares 54% amino acid identity with human IL36A (IL-1F6) and lower homology with other IL-1 family members (e.g., 29% with IL-1ra) .

Pro-Inflammatory Activity in Lung Tissues

IL36A Mouse induces neutrophilic inflammation in the lungs by activating IL-36 receptor (IL-36R)-dependent pathways. Key findings include:

  • Neutrophil Recruitment: Intratracheal instillation of recombinant IL36A triggers neutrophil influx in wild-type and IL-1αβ-deficient mice .

  • Chemokine Production: Upregulates CXCL1 (KC) and CXCL2 (MIP-2) mRNA, critical for neutrophil chemoattraction .

  • Receptor Activation: Binds IL-1 Rrp2 (IL-36R) and recruits IL-1 RAcP for signaling, activating NF-κB and MAPK pathways .

Role in Psoriasis and Skin Inflammation

  • Epidermal Hyperplasia: Transgenic overexpression of IL36A in keratinocytes causes psoriasis-like skin lesions .

  • T Cell Modulation: Enhances CD4+ T cell proliferation and CD40 expression on dendritic cells .

Regulation in Macrophages and Neutrophils

  • LPS Responsiveness: Induced by lipopolysaccharide (LPS) via C/EBPβ binding to a half-CRE- C/EBP element in the Il36a promoter .

  • Neutrophil Source: Neutrophils in LPS-challenged mice produce IL36A, amplifying inflammation .

Lung Inflammation Models

ModelOutcomeMechanismSource
Cigarette Smoke (CS)Reduced neutrophil influx in Il36r−/− miceAttenuated CXCL1, IL-1β, IL-6
CS + H1N1 InfluenzaMitigated alveolar IL-1β and TNF-α in Il36r−/− miceIL-36R dependency in viral exacerbations
IL36A Tracheal InstillationIncreased CXCL1/CXCL2 mRNA in IL-1αβ−/− miceIndependent of IL-1α/β signaling

Psoriasis and Autoimmune Diseases

  • Keratinocyte-Specific Deletion: Il36rΔK mice show reduced IL-36R expression in keratinocytes, mimicking Il36r−/− phenotypes .

  • Osteoarthritis (OA): IL36A overexpression in cartilage exacerbates OA progression, while IL-36Ra (antagonist) mitigates joint damage .

Recombinant Protein Formulations

Recombinant IL36A Mouse is widely used in in vitro and in vivo studies. Key formulations include:

ProductFormulationStabilityApplications
Prospec Bio CYT-181Lyophilized in 1×PBS, pH 7.4Stable at -18°C (lyophilized)Bioactivity assays, ELISA
R&D Systems 7059-MLWith BSA (carrier protein)Store at -20°C (reconstituted)Cell culture, in vivo studies
R&D Systems 7059-ML/CFTrehalose (carrier-free)Ambient shipping, -20°C storageProtein-protein interaction

Functional Assays

  • IL-6 Production: Recombinant IL36A induces IL-6 secretion in NIH-3T3 cells (ED50: 3–18 ng/mL) .

  • NF-κB Activation: Stimulation of macrophages triggers NF-κB signaling, confirmed via luciferase assays .

Product Specs

Introduction
Murine IL-36a, a member of the IL-1 family, shares structural similarities with IL-1b, IL-1a, IL-1ra, IL-18, IL-36ra (IL1F5), IL-36b (IL1F8), IL-36g (IL1F9), IL-37 (IL1F7), and IL-38 (IL-1F10). These family members exhibit a 12 β-strand, β-trefoil configuration, suggesting a common ancestral origin. As a 160-amino acid protein, murine IL-36a exists in both intracellular and secreted forms, lacking a signal sequence, prosegment, and potential N-linked glycosylation sites. The release of IL-36a is triggered by LPS and the activation of the P2X7 receptor induced by cellular ATP. Notably, there is a 54% amino acid sequence homology between the full-length IL-36a/IL-1F6 of mice and humans. IL-36a is predominantly found in the skin and lymphoid tissues, with detectable levels in fetal brain, trachea, stomach, and intestine.
Description
Recombinant IL36A Mouse, produced in E. coli, is a single, non-glycosylated polypeptide chain comprising 160 amino acids, resulting in a molecular weight of 21.0 kDa. The purification of IL36A is achieved through proprietary chromatographic methods.
Physical Appearance
Sterile Filtered White lyophilized (freeze-dried) powder.
Formulation
Lyophilized from a 0.2 μm filtered concentrated solution in 1x PBS, pH 7.4.
Solubility
Reconstitute the lyophilized IL36A in sterile 18 MΩ-cm H₂O to a concentration of at least 100 μg/ml. This solution can be further diluted in other aqueous solutions.
Stability
Lyophilized IL36A Mouse, while stable at room temperature for 3 weeks, should be stored desiccated below -18°C. Upon reconstitution, store IL36A at 4°C for 2-7 days. For long-term storage, keep it below -18°C. Avoid repeated freeze-thaw cycles.
Purity
Greater than 95.0% as determined by SDS-PAGE.
Biological Activity
The biological activity is assessed through its binding ability in a functional ELISA, specifically its capacity to bind to recombinant mouse IL-1 Rrp2.
Synonyms
Interleukin 36 alpha, FIL1E, IL1F6, FIL1, IL1(EPSILON), interleukin 1 family member 6 (epsilon), MGC129552, MGC129553.
Source
Escherichia Coli.
Amino Acid Sequence
MNKEKELRAA SPSLRHVQDL SSRVWILQNN ILTAVPRKEQ TVPVTITLLP CQYLDTLETN RGDPTYMGVQ RPMSCLFCTK DGEQPVLQLG EGNIMEMYNK KEPVKASLFY HKKSGTTSTF ESAAFPGWFI AVCSKGSCPL ILTQELGEIF ITDFEMIVVH

Q&A

What is IL-36α and how does it function within the IL-1 cytokine family in mice?

IL-36α belongs to the IL-1 superfamily and is part of the IL-36 subfamily, which includes three agonists (IL-36α, IL-36β, IL-36γ) and one antagonist (IL-36Ra). All IL-36 isoforms bind to the IL-36 receptor (IL-36R) . When IL-36α binds to IL-36R, it recruits the IL-1 receptor accessory protein (IL-1RAcP) and activates downstream signaling pathways mediated by nuclear transcription factor kappa B and mitogen-activated protein kinase . This activates inflammatory cascades resulting in pro-inflammatory cytokine and chemokine production .

IL-36α functions independently of other IL-1 family members, as demonstrated by studies showing that intratracheal instillation of IL-36α can induce neutrophil influx in both wild-type C57BL/6 mice and IL-1αβ(-/-) mice . This functional independence makes IL-36α an important target for specific therapeutic interventions in inflammatory diseases.

Which cell types express IL-36α and IL-36R in mouse models?

In mouse models, IL-36α is expressed by a diverse range of cell types:

  • Monocytes and B cells

  • T cells (notably, IL-36α is the only novel IL-1 family member expressed on T-cells)

  • Neutrophils (identified as a source in lung inflammation models)

  • Epithelial cells in skin and lung tissues

The IL-36 receptor (IL-36R) expression has been documented in:

  • Bone marrow-derived dendritic cells

  • CD4+ T cells

  • Monocytes and myeloid dendritic cells (mDCs)

  • Monocyte-derived dendritic cells (MDDCs)

  • Keratinocytes

This expression pattern explains the wide-ranging effects of IL-36α across multiple immune and non-immune cell types in inflammatory responses.

How does the regulation of IL-36α differ from other inflammatory cytokines in mice?

IL-36α regulation involves unique transcriptional control mechanisms that distinguish it from other inflammatory cytokines. One notable aspect is the regulation by C/EBPβ (CCAAT/enhancer-binding protein β), which binds specifically to a half-CRE- C/EBP motif in the IL-36α promoter to induce expression upon lipopolysaccharide (LPS) stimulation .

Interestingly, C/EBPβ binding to this regulatory element is insensitive to CpG methylation, allowing robust expression regardless of the methylation status of the promoter . This differs from many other cytokines whose expression is highly regulated by epigenetic modifications.

Additionally, IL-36α can be regulated by other inflammatory cytokines, creating feedback loops. IL-1α can induce IL-36α expression, and in return, IL-36α may regulate IL-1α in mouse keratinocytes . Similarly, IL-22, IL-17A, and TNF-α can induce the production of IL-36 subfamily members in human systems, suggesting similar regulatory mechanisms may exist in mice .

What are effective mouse models for studying IL-36α-mediated inflammation?

Several experimental mouse models have proven effective for studying IL-36α-mediated inflammation:

Model TypeMethodKey FindingsApplications
Lung inflammationIntratracheal instillation of recombinant IL-36αInduces neutrophil influx and CXCL1/CXCL2 expressionStudies of acute neutrophilic airway inflammation
Cigarette smoke exposureExposure of IL-36R-deficient vs. wild-type mice to cigarette smokeIL-36R-deficient mice show attenuated inflammationModels of chronic obstructive pulmonary disease
Viral-bacterial co-infectionExposure to cigarette smoke and H1N1 influenza virusIL-36 acts as upstream amplifier in combination with viral stimuliStudies of infection-triggered exacerbations
Skin inflammationImiquimod-induced psoriasis with IL-36α injectionIL-36α contributes to severe skin lesion developmentPsoriasis research
Genetic modelsIL-36α knockout or IL-36R knockout miceReduced neutrophil recruitment and chemokine productionMechanistic studies of IL-36α function

How should researchers quantify IL-36α-induced neutrophilic inflammation in mouse lungs?

Quantifying IL-36α-induced neutrophilic inflammation in mouse lungs requires a multi-parameter approach:

  • Bronchoalveolar lavage (BAL) analysis:

    • Collect BAL fluid and perform total and differential cell counts

    • Quantify absolute neutrophil numbers and percentage of total cells

    • Measure neutrophil elastase or myeloperoxidase activity in BAL fluid

  • Lung tissue analysis:

    • Perform histological assessment with H&E staining and immunohistochemistry

    • Score inflammatory infiltrates on a standardized scale

    • Use immunofluorescence to co-localize neutrophils with other cell types

  • Gene expression analysis:

    • Quantify mRNA expression of neutrophil chemokines CXCL1 and CXCL2

    • Measure neutrophil-associated genes (e.g., Ly6G, elastase)

    • Assess IL-36α and IL-36R expression levels to evaluate potential feedback loops

  • Protein measurements:

    • Perform ELISA or multiplex cytokine assays to measure chemokines in BAL fluid

    • Use Western blotting to assess activation of signaling pathways in lung tissue

    • Quantify neutrophil-derived proteins such as calprotectin

  • Flow cytometry:

    • Analyze single-cell suspensions from digested lungs

    • Identify neutrophils using markers such as CD11b and Ly6G

    • Assess activation status with markers like CD66

Research shows that IL-36α significantly increases neutrophil numbers and CXCL1/CXCL2 expression in mouse lungs, making these key parameters to monitor .

What methodological considerations should be taken when using IL-36α or IL-36R knockout mice?

When using IL-36α or IL-36R knockout mice, researchers should consider several methodological aspects:

  • Knockout verification:

    • Confirm gene deletion by genotyping

    • Verify protein absence using Western blot or ELISA

    • Check for compensatory upregulation of related cytokines (IL-36β, IL-36γ)

  • Strain background effects:

    • Use mice backcrossed to a consistent genetic background (typically C57BL/6)

    • Include appropriate wild-type littermate controls

    • Consider potential strain-dependent variations in inflammatory responses

  • Functional redundancy:

    • IL-36R knockout eliminates signaling from all IL-36 family members

    • IL-36α knockout may show partial phenotypes due to compensation by IL-36β/γ

    • Consider using combined knockouts or neutralizing antibodies to address redundancy

  • Developmental effects:

    • Assess whether developmental abnormalities exist in knockout mice

    • Consider using conditional knockout models to avoid developmental adaptations

    • Compare results with pharmacological inhibition approaches

  • Baseline characterization:

    • Evaluate steady-state immune parameters before experimental manipulation

    • Check for pre-existing differences in cell populations

    • Document any spontaneous phenotypes in unchallenged knockout mice

Research has shown that IL-36α knockout mice display reduced neutrophil recruitment to the epidermis and dermis, along with downregulated CXCL1 generation in inflammatory skin conditions, demonstrating the value of these models for understanding IL-36α function .

How does IL-36α signaling drive neutrophilic inflammation in mouse models?

IL-36α drives neutrophilic inflammation through multiple interconnected mechanisms in mouse models:

  • Direct chemokine induction: IL-36α stimulation increases expression of neutrophil-specific chemokines CXCL1 and CXCL2 in the lungs, creating a chemotactic gradient for neutrophil recruitment .

  • Activation of CD11c+ cells: IL-36α stimulates dendritic cells and macrophages to produce neutrophil-specific chemokines and TNFα, amplifying the inflammatory response. Additionally, IL-36α enhances expression of the co-stimulatory molecule CD40 on these cells .

  • Positive feedback loops: Intratracheal IL-36α enhances expression of its own receptor (IL-36R) in the lungs, creating a self-amplifying inflammatory circuit .

  • Synergistic effects: IL-36α cooperates with GM-CSF and viral mimics like poly(I:C) to promote activation of neutrophils, macrophages, and fibroblasts, further enhancing inflammation .

  • NF-κB activation: Stimulation with IL-36α activates NF-κB in mouse macrophage cell lines, triggering pro-inflammatory gene expression .

  • Cross-talk with adaptive immunity: IL-36α enhances the ability of dendritic cells to induce CD4+ T cell proliferation, linking innate and adaptive immune responses .

This multi-faceted role positions IL-36α as a crucial upstream amplifier of neutrophilic inflammation, particularly in lung diseases characterized by neutrophil infiltration.

What is known about the transcriptional regulation of IL-36α in mouse models?

Transcriptional regulation of IL-36α in mouse models involves several key mechanisms:

  • C/EBPβ-mediated regulation: The transcription factor C/EBPβ binds specifically to a half-CRE- C/EBP motif in the IL-36α promoter to induce expression following LPS stimulation . This binding occurs regardless of CpG methylation status, allowing robust expression in different cellular contexts.

  • LPS-induced expression: Lipopolysaccharide is a potent inducer of IL-36α transcription in mouse macrophages, suggesting a role in bacterial infection responses .

  • Cytokine networks: In mouse keratinocytes, IL-1α can induce IL-36α expression, creating interconnected cytokine networks. The levels of IL-36α from inflamed IL-1R1−/− skin were significantly lower than those of wild-type mice, suggesting IL-1 receptor signaling contributes to IL-36α regulation .

  • Cell type-specific mechanisms: Studies show differential methylation of the IL-36α promoter in the RAW264.7 macrophage cell line compared to primary murine macrophages, yet similar IL-36α mRNA and protein expression levels were observed following stimulation .

  • Viral and bacterial stimuli: Beyond LPS, other pathogen-associated molecular patterns likely regulate IL-36α expression, as evidenced by its role in models combining cigarette smoke exposure and H1N1 influenza virus infection .

Understanding these regulatory mechanisms provides insights into how IL-36α expression is controlled during inflammatory responses and potential points for therapeutic intervention.

How does IL-36α interact with other cytokines in inflammatory cascades?

IL-36α engages in complex interactions with other cytokines within inflammatory cascades:

  • IL-1 family cross-regulation: IL-36α and IL-1α appear to regulate each other in a feedback loop. IL-1α can induce IL-36α expression, and conversely, IL-36α stimulation leads to increased IL-1α production in mouse keratinocytes .

  • Synergy with GM-CSF: IL-36α cooperates with GM-CSF to promote activation of neutrophils, macrophages, and fibroblasts, amplifying inflammatory responses in the lung .

  • Enhancement of viral responses: IL-36α collaborates with the viral mimic poly(I:C) to potentiate inflammatory responses, suggesting an important role during viral infections .

  • Independence from IL-1α/β: Studies in IL-1αβ(-/-) mice demonstrated that IL-36α can induce neutrophil influx and chemokine expression independently of IL-1α and IL-1β, highlighting its non-redundant role in inflammation .

  • Th17 pathway connections: IL-36α can induce expression of IL-17A signaling-related genes, suggesting cross-talk with the Th17 inflammatory axis which is crucial in several autoimmune conditions .

  • Pro-inflammatory cytokine induction: In mouse monocytes, IL-36 stimulation significantly upregulates expression of IL-1α, IL-1β, and IL-6, creating a broader cytokine response .

These interactions position IL-36α as an important node in inflammatory networks, with the ability to amplify and sustain inflammatory responses through multiple cytokine pathways.

How does IL-36α contribute to psoriasis-like inflammation in mouse models?

IL-36α plays several critical roles in psoriasis-like inflammation in mouse models:

  • Disease exacerbation: In the imiquimod-induced psoriasis mouse model, IL-36α injection contributes to the development of severe skin lesions, demonstrating its pathogenic role .

  • Neutrophil recruitment: IL-36α knockout (-/-) mice show reduced neutrophil infiltration in the epidermis and dermis following inflammatory stimuli, indicating its importance in the neutrophilic component of psoriasiform inflammation .

  • Chemokine regulation: IL-36α deficiency results in downregulated CXCL1 generation, a key neutrophil-attracting chemokine in psoriatic lesions .

  • Therapeutic target validation: Psoriatic mice treated with IL-36R-blocking antibodies show improved psoriatic dermatitis, providing evidence for IL-36 pathway inhibition as a therapeutic approach .

  • Genetic associations: IL-36 signaling-related genes are enriched within psoriasis susceptibility loci, supporting a genetic basis for IL-36α involvement in psoriasis .

  • Cytokine network amplification: IL-36α participates in inflammatory loops with other psoriasis-relevant cytokines. For example, IL-22, IL-17A, and TNF-α induce IL-36 production in keratinocytes, while IL-36α can promote IL-17A signaling pathways .

These findings from mouse models have helped establish IL-36α as a key mediator in psoriasis pathogenesis and have supported the development of targeted therapies against the IL-36 pathway, particularly for pustular forms of psoriasis.

What is the role of IL-36α in mouse models of lung inflammation?

IL-36α serves as a critical mediator in mouse models of lung inflammation:

  • Key upstream amplifier: Research has identified IL-36α as a key upstream amplifier of neutrophilic lung inflammation, promoting activation of neutrophils, macrophages and fibroblasts .

  • Direct neutrophil recruitment: Intratracheal instillation of recombinant IL-36α induces neutrophil influx in the lungs of both wild-type C57BL/6 mice and IL-1αβ(-/-) mice .

  • Chemokine induction: IL-36α stimulation increases mRNA expression of neutrophil-specific chemokines CXCL1 and CXCL2 in the lungs, driving neutrophil infiltration .

  • Synergistic pathways: IL-36α cooperates with GM-CSF and viral mimic poly(I:C) to amplify lung inflammation, suggesting important roles during respiratory infections .

  • Cigarette smoke responses: IL-36 receptor deficient mice exposed to cigarette smoke show attenuated lung inflammation compared with wild-type controls, implicating IL-36α in smoking-related lung diseases .

  • Viral exacerbations: IL-36 receptor deficient mice exposed to cigarette smoke and H1N1 influenza virus have attenuated lung inflammation, suggesting a role in viral exacerbations of chronic lung diseases .

  • Self-amplification: Intratracheal IL-36α enhances mRNA expression of its own receptor (IL-36R) in the lungs, creating a positive feedback loop that magnifies inflammatory responses .

These findings provide a rationale for targeting IL-36α in various neutrophilic lung diseases, including chronic obstructive pulmonary disease (COPD), severe asthma, and respiratory infections.

How can IL-36α antagonism be achieved in mouse models and what therapeutic effects have been observed?

Several approaches to IL-36α antagonism have been implemented in mouse models, yielding significant therapeutic effects:

  • IL-36R blocking antibodies: Treatment of psoriatic mice with IL-36R-blocking antibodies improves psoriatic dermatitis, demonstrating the potential of this approach .

  • Genetic deletion models: IL-36α knockout mice show reduced neutrophil recruitment and chemokine production in inflammatory conditions, validating IL-36α as a therapeutic target .

  • IL-36Ra administration: Recombinant IL-36 receptor antagonist (IL-36Ra) administration can inhibit IL-36 signaling. IL-36Ra binds to IL-36R but inhibits recruitment of IL-1RAcP, blocking downstream signaling pathways .

  • Small molecule inhibitors: While still in development, targeting downstream signaling components such as MyD88 or IRAK proteins can inhibit IL-36α-mediated responses.

  • Combined pathway inhibition: Simultaneous targeting of IL-36α and synergistic pathways (e.g., GM-CSF) has shown enhanced anti-inflammatory effects in lung inflammation models .

Therapeutic effects observed with these approaches include:

  • Reduced neutrophilic infiltration in skin and lung tissues

  • Decreased pro-inflammatory cytokine and chemokine production

  • Attenuated tissue damage and remodeling

  • Improved disease scores in psoriasis models

  • Protection from cigarette smoke-induced lung inflammation

These findings from mouse models have translated to human studies, with monoclonal antibodies against IL-36R showing promise in clinical trials for pustular psoriasis patients .

What are the optimal methods for detecting and quantifying IL-36α in mouse samples?

Several complementary methods can be used for detecting and quantifying IL-36α in mouse samples:

MethodApplicationAdvantagesConsiderations
ELISAQuantification of IL-36α protein in tissue homogenates, cell culture supernatants, or biological fluidsHigh specificity and sensitivity; quantitative measurementsRequires validated antibodies; limited spatial information
Western blottingDetection of IL-36α protein and processing formsIdentifies specific protein bands; detects protein modificationsSemi-quantitative; requires tissue disruption
Quantitative PCR (qPCR)Measurement of IL-36α mRNA expressionHigh sensitivity; specific primer designReflects transcription, not protein levels; requires reference genes
Immunohistochemistry (IHC)Visualization of IL-36α in tissue sectionsPreserves tissue architecture; identifies expressing cellsSemi-quantitative; dependent on antibody quality
ImmunofluorescenceCo-localization of IL-36α with cellular markersAllows multi-parameter analysis; high-resolution imagingRequires careful controls for autofluorescence
RNA in situ hybridizationVisualization of IL-36α mRNA in tissuesSpecific detection of transcript; works when antibodies are lackingComplex protocol; requires specialized equipment
Flow cytometryIntracellular IL-36α in single cellsSingle-cell resolution; combinable with surface markersRequires tissue dissociation; potential artifacts

For comprehensive analysis, researchers should use multiple complementary approaches. The Mouse IL-36 alpha/IL-1F6 DuoSet ELISA from R&D Systems provides a validated tool for quantitative measurements in mouse samples .

What are the best approaches for studying IL-36α signaling mechanisms in mouse cells?

Studying IL-36α signaling mechanisms in mouse cells requires several specialized approaches:

  • Phosphorylation assays:

    • Western blotting for phosphorylated signaling proteins (p38 MAPK, JNK, ERK)

    • Phospho-flow cytometry for single-cell analysis of signaling activation

    • Phospho-proteomics to identify novel signaling targets

  • Transcriptional regulation:

    • Chromatin immunoprecipitation (ChIP) to study transcription factor binding to IL-36α-regulated genes

    • Reporter assays with promoter constructs to identify regulatory elements

    • Analysis of histone modifications at IL-36α-responsive genes

  • Receptor studies:

    • Binding assays with labeled recombinant IL-36α

    • Co-immunoprecipitation of IL-36R with IL-1RAcP following stimulation

    • CRISPR/Cas9 modification of receptor components

  • Gene expression profiling:

    • RNA-sequencing of IL-36α-stimulated versus unstimulated cells

    • Pathway analysis to identify signaling networks

    • Comparison of wild-type versus IL-36R-deficient cells

  • Genetic approaches:

    • Knockdown studies using siRNA targeting specific signaling components

    • Overexpression of dominant-negative mutants of signaling molecules

    • Pharmacological inhibitors of specific signaling pathways

  • Cell-specific analyses:

    • Isolation of specific cell populations from IL-36α-treated mice

    • Single-cell RNA-seq to identify cell-specific responses

    • Conditional knockout of signaling components in specific cell types

Research has shown that IL-36α signaling activates NF-κB in mouse macrophage cell lines and induces p38-MAPK signaling-related genes, making these important pathways to investigate .

How should researchers design IL-36α dose-response studies in mouse models?

Designing effective IL-36α dose-response studies in mouse models requires careful methodological considerations:

  • Dose range determination:

    • For intratracheal administration, test a range from 0.1-10 μg per mouse

    • Include at least four different doses to establish a complete dose-response curve

    • Use both sub-threshold and saturating doses to capture the full response range

  • Time course considerations:

    • Perform both acute (hours) and extended (days) timepoints

    • Include measurements at 2h, 6h, 24h, and 48h post-administration

    • Consider multiple administrations for chronic models

  • Readout selection:

    • Primary readouts: neutrophil counts, chemokine levels (CXCL1, CXCL2)

    • Secondary readouts: tissue histology, gene expression profiles

    • Functional measures: lung function tests, behavioral assessments

  • Controls and validation:

    • Include inactive protein controls (heat-inactivated IL-36α)

    • Use IL-36R knockout mice to confirm specificity

    • Compare with positive controls (LPS, TNF-α) at established doses

  • Route of administration considerations:

    • For lung studies: intratracheal, intranasal, or aerosolized delivery

    • For skin studies: intradermal or topical application

    • For systemic effects: intraperitoneal or intravenous administration

  • Synergy assessment:

    • Combine IL-36α with other stimuli (GM-CSF, poly(I:C))

    • Test multiple combinations of concentrations

    • Calculate combination indices to quantify synergistic effects

  • Statistical analysis:

    • Use appropriate statistical methods for dose-response curves (e.g., four-parameter logistic regression)

    • Calculate EC50 values to compare potency across different readouts

    • Consider area-under-curve analyses for time-dependent responses

Research has shown that intratracheal instillation of IL-36α induces dose-dependent neutrophil influx and chemokine expression in mouse lungs, providing a foundation for dose-response study designs .

Product Science Overview

Introduction

Interleukin-36 alpha (IL-36α), also known as IL-1F6, is a member of the interleukin-1 (IL-1) cytokine family. This family includes several pro-inflammatory cytokines that play crucial roles in the regulation of immune responses. IL-36α is particularly significant due to its involvement in both innate and adaptive immunity.

Structure and Expression

IL-36α is a 160 amino acid protein that lacks a signal sequence, prosegment, and potential N-linked glycosylation sites . It is produced by various cells, including epithelial and immune cells, and is released in response to stimuli such as lipopolysaccharides (LPS) and ATP-induced activation of the P2X7 receptor . The protein is expressed and purified from E. coli for recombinant use .

Signaling Pathways

IL-36α functions by binding to its receptor, IL-36R, which then recruits the accessory protein IL-1RAcP to form a ternary complex . This complex activates intracellular signaling pathways, including NF-κB and MAPK, leading to the production of pro-inflammatory mediators . The activation of these pathways is crucial for the cytokine’s role in immune responses.

Biological Functions

IL-36α plays a significant role in the inflammatory response. It is involved in the activation of MAPK and NF-κB pathways, which are essential for the production of inflammatory cytokines and chemokines . This cytokine is also implicated in the pathophysiology of several diseases, including psoriasis and other inflammatory conditions .

Role in Disease

Dysregulation of IL-36α signaling can lead to various inflammatory diseases. For instance, in the skin, IL-36α contributes to host defense through inflammatory responses. However, when dysregulated, it can stimulate keratinocytes and immune cells, enhancing the Th17/Th23 axis and inducing psoriatic-like skin disorders . Genetic mutations in the antagonist IL-36Ra are associated with generalized pustular psoriasis, a severe skin disease .

Recombinant IL-36α

Recombinant mouse IL-36α is produced for research purposes to study its biological functions and potential therapeutic applications. The recombinant protein is typically lyophilized and can be reconstituted for use in various experimental setups . It is essential to store the protein under sterile conditions at -20°C to -80°C to maintain its stability and avoid repeated freeze-thaw cycles .

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.