IL 33 Human

Interleukin-33 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure and Activation

IL-33 exists in two primary forms:

  • Full-length IL-33 (flIL-33; aa 1–270): Constitutively expressed in endothelial/epithelial cells and sequestered in nuclei via chromatin-binding motifs .

  • Mature IL-33 (mIL-33; aa 95–270, 99–270, or 109–270): Generated via protease cleavage (e.g., neutrophil elastase, mast cell tryptase), exhibiting ~10x higher activity than flIL-33 .

FeatureFull-Length IL-33Mature IL-33
BioactivityModerateHigh
Protease SensitivitySensor domain cleavageStable
Primary SourceNecrotic cellsInflammatory sites

Structural studies reveal a β-trefoil fold critical for binding its receptor ST2 and co-receptor IL-1RAcP . Charge complementarity between IL-33 and ST2 ensures ligand-receptor specificity .

Alarmin Activity

IL-33 is released during cellular necrosis or tissue damage, activating:

  • Group 2 Innate Lymphoid Cells (ILC2s): Drive type 2 cytokine production (IL-5, IL-13) .

  • Mast Cells/Basophils: Amplify allergic inflammation .

  • Th2 Cells: Promote eosinophil recruitment and airway hyperreactivity .

Metabolic Regulation

IL-33 modulates immune cell metabolism via:

  • mTOR/AMPK Pathways: Influence glucose/lipid metabolism in macrophages and T cells .

  • PPAR-γ Activation: Enhances fatty acid oxidation in ILC2s .

Respiratory Diseases

  • Asthma: Elevated IL-33 in inflamed airways drives eosinophilia and mucus hypersecretion . Genetic variants of IL33 and ST2 correlate with asthma susceptibility .

  • COPD/Fibrosis: IL-33 promotes fibroblast activation and collagen deposition .

Cancer Immunology

  • Tertiary Lymphoid Structures (TLSs): IL-33-activated ILC2s recruit LTβR+ myeloid cells to induce TLSs, enhancing anti-tumor immunity in pancreatic ductal adenocarcinoma .

Trauma and Systemic Inflammation

  • Post-traumatic IL-33 surges correlate with injury severity (ISS ≥ 25) and mortality, suggesting endothelial-derived alarmin release .

DiseaseMechanismTherapeutic Target
Allergic AsthmaILC2 activation, Th2 polarizationAnti-ST2 antibodies
Atopic DermatitisKeratinocyte-derived IL-33Protease inhibitors
Pancreatic CancerILC2-dependent TLS formationRecombinant IL-33

Regulatory Mechanisms

  • Oxidative Inactivation: Cysteine residues in flIL-33 are oxidized extracellularly, limiting activity .

  • Epigenetic Modulation: IL-33 alters chromatin accessibility in immune cells via histone methyltransferases (e.g., SUV39H1) .

Therapeutic Prospects

Current strategies focus on:

  • ST2 Blockade: Monoclonal antibodies to disrupt IL-33/ST2 signaling .

  • Protease Inhibitors: Prevent flIL-33 maturation into hyperactive forms .

  • Recombinant IL-33: Engineered variants to boost TLS formation in tumors .

Product Specs

Introduction
Interleukin 33 (IL-33) is a 32 kDa pro-inflammatory cytokine that may play a role in regulating gene transcription within cells that produce it. Structurally similar to IL-1, IL-33 triggers helper T cells to generate type 2 cytokines. It operates through the IL1RL-1 receptor (also known as ST2). When IL-33 binds to this receptor, it activates NF-kappa-B and MAP kinases, prompting Th2 cells to produce cytokines in laboratory settings. In living organisms, IL-33 stimulates the expression of IL-4, IL-5, and IL-13, leading to significant pathological changes in mucosal organs. In vitro, caspase-1 can cleave IL-33 into a 12 kDa N-terminal fragment and an 18 kDa C-terminal fragment.
Description
Recombinant Human Interleukin 33, produced in E. coli, is a single, non-glycosylated polypeptide chain comprised of 160 amino acids. It has a molecular weight of 18,125 Daltons. The purification of IL-33 is achieved through proprietary chromatographic techniques.
Physical Appearance
Sterile Filtered White lyophilized powder.
Formulation
Lyophilized from a 10mM NaP solution at a pH of 7.5.
Solubility
To reconstitute the lyophilized IL-33, it is recommended to dissolve it in sterile 18 MΩ-cm H₂O at a concentration of at least 100 µg/ml. Further dilutions can be made using other aqueous solutions.
Stability
Lyophilized IL-33, while stable at room temperature for up to 3 weeks, should be stored in a dry environment below -18°C. Once reconstituted, IL-33 should be stored at 4°C for no longer than 2-7 days. For long-term storage, adding a carrier protein like 0.1% HSA or BSA is recommended. Avoid repeated freeze-thaw cycles.
Purity
Purity exceeds 95.0% as determined by: (a) RP-HPLC analysis. (b) SDS-PAGE analysis.
Synonyms
Interleukin 33, DVS27, NF-HEV, NKHEV, C9orf26, Interleukin-1 family member 11, IL- 1F11, Nuclear factor from high endothelial venules, NFEHEV, DKFZp586H0523, RP11-575C20.2, IL-33.
Source
Escherichia Coli.
Amino Acid Sequence
MSITGISPIT EYLASLSTYN DQSITFALED ESYEIYVEDL KKDEKKDKVL LSYYESQHPS NESGDGVDGK MLMVTLSPTK DFWLHANNKE HSVELHKCEK PLPDQAFFVL HNMHSNCVSF ECKTDPGVFI GVKDNHLALI KVDSSENLCT ENILFKLSET.

Q&A

What is IL-33 and what are its structural characteristics?

IL-33 is a proinflammatory cytokine that functions as an alarmin when released from damaged cells. Human IL-33 is a 270 amino acid protein composed of three functional domains: a nuclear domain (amino acids 1-65) containing a chromatin-binding motif, a central domain (amino acids 66-111) with protease cleavage sites, and an IL-1-like cytokine domain (amino acids 112-270) . The nuclear domain includes a homeodomain-like HTH domain with a nuclear localization signal that tethers IL-33 to chromatin by binding to the H2A-H2B histone dimer . This nuclear localization prevents unwanted inflammatory responses . When full-length IL-33 (IL-33FL) is cleaved by inflammatory proteases like elastase, neutrophils, and cathepsin G, it produces mature forms with 10-30 times greater biological activity than IL-33FL .

Experimental approach: To study IL-33 structural characteristics, researchers should consider domain-specific antibodies for immunoprecipitation and Western blotting to distinguish between full-length and processed forms. Recombinant expression of domain mutants can help elucidate structure-function relationships.

How is IL-33 expressed in human tissues under normal and inflammatory conditions?

In humans, IL-33 is constitutively and abundantly expressed in various tissues under basal conditions. The primary sources in the respiratory system are bronchial epithelial cells and endothelial cells of high endothelial venules . This differs from mice, where IL-33 is mainly expressed by lung alveolar type II pneumocytes rather than bronchial epithelial cells, representing an important species difference when translating animal studies to humans .

IL-33 expression increases during inflammation or tissue stress. In humans, elevated IL-33 expression has been observed in nasal and lung epithelium of patients with allergic asthma, allergic rhinitis, and chronic obstructive pulmonary disease (COPD) . Inflammation can also induce IL-33 expression in immune cells like macrophages, dendritic cells, B cells, and mast cells, though tissue cell-derived IL-33 appears more essential for allergen-driven airway inflammation .

Methodological considerations: When analyzing IL-33 expression patterns, researchers should:

  • Use cell type-specific markers in immunohistochemistry to identify IL-33-producing cells

  • Consider both nuclear and cytoplasmic staining patterns

  • Compare expression between healthy and diseased tissues using matched samples

  • Validate findings with multiple detection methods (e.g., qPCR, Western blot, immunohistochemistry)

What are the primary receptors and signaling pathways for IL-33?

The primary receptor for IL-33 is ST2 (also known as IL1RL1), which forms a heterodimeric complex with IL-1 receptor accessory protein (IL-1RAcP) . When extracellular IL-33 binds to membrane-bound ST2, the complex recruits IL-1RAcP, initiating a signaling cascade . This heterotrimeric complex formation is essential for signal transduction.

The signaling pathway activates MyD88, IRAK1/4, and TRAF6, leading to NF-κB and MAPK pathway activation. This results in the production of type 2 cytokines and promotes allergic inflammatory responses . The IL-33/ST2 axis plays critical roles in both innate immunity (particularly through group 2 innate lymphoid cells) and adaptive immunity (through effects on dendritic cells, Th2 cells, and regulatory T cells) .

Experimental approaches:

  • Use phospho-specific antibodies to monitor activation of downstream signaling molecules

  • Employ pharmacological inhibitors of specific pathway components to dissect signaling mechanisms

  • Utilize CRISPR/Cas9 to generate knockout or knockin cell lines for mechanistic studies

  • Consider reporter cell lines that express luciferase under NF-κB-responsive promoters

Which immune cells respond to IL-33 and what are their functions?

Multiple immune cell types express the ST2 receptor and respond to IL-33 stimulation:

  • Group 2 innate lymphoid cells (ILC2s): These cells are major responders to IL-33 and rapidly produce large amounts of type 2 cytokines (IL-5, IL-13) upon stimulation . ILC2s provide an essential axis for rapid immune responses and tissue homeostasis in innate immunity.

  • T cells: IL-33 acts directly on several T cell subsets including:

    • Th2 cells: Enhances production of IL-5 and IL-13

    • Regulatory T cells: Promotes their stability and function in some contexts

    • Follicular T cells: Influences their development and function

  • Mast cells and basophils: Respond to IL-33 with degranulation and cytokine production

  • Dendritic cells: IL-33 can modulate their activation and cytokine production, influencing subsequent T cell responses

  • Eosinophils: Express ST2 and respond to IL-33 with increased survival and activation

Methodological considerations:

  • Single-cell technologies (flow cytometry, scRNA-seq) can identify specific responder populations

  • Bone marrow chimeras or conditional knockout models help distinguish cell-intrinsic effects

  • Multiplex cytokine assays capture the range of mediators produced in response to IL-33

What are the mechanisms of IL-33 release from cells?

IL-33 release occurs through both passive and active mechanisms:

Passive release: During cellular necrosis, the nuclear membrane breaks down, allowing nuclear IL-33 to be released into the extracellular space as a damage-associated molecular pattern (DAMP) .

Active release: IL-33 can also be actively secreted without cell death through mechanisms that remain incompletely understood. Airway epithelial cells exposed to allergens like Alternaria or cockroach extract show translocation of IL-33 from the nucleus to the cytoplasm followed by extracellular release without apparent cell death . This pathway involves:

  • ATP release and purinergic receptor activation: Allergen exposure causes extracellular ATP accumulation, which activates P2 purinergic receptors .

  • Calcium signaling: P2 receptor activation increases intracellular Ca2+ concentrations, critical for IL-33 release. Calcium chelators or P2 receptor antagonists inhibit allergen-induced IL-33 release .

  • NADPH oxidase activation: In airway epithelial cells, NADPH oxidase dual oxidase 1 (DUOX1) is activated following P2 receptor engagement and mediates IL-33 secretion .

  • Potential inflammasome involvement: The NLRP3 inflammasome may contribute to IL-33 secretion in certain contexts .

Experimental approaches:

  • Live cell imaging with fluorescently tagged IL-33 to track translocation

  • ELISA assays of supernatants combined with cell viability assessments

  • Pharmacological inhibitors of calcium signaling, purinergic receptors, and NADPH oxidases

  • Genetic approaches (siRNA, CRISPR) targeting specific pathway components

How do post-translational modifications affect IL-33 bioactivity?

IL-33 bioactivity is significantly regulated by post-translational modifications:

  • Proteolytic processing: Full-length IL-33 has relatively weak cytokine activity compared to processed forms. Cleavage by inflammatory proteases from neutrophils, mast cells, and other sources generates mature forms with substantially enhanced bioactivity (10-30 times stronger than IL-33FL) . The central domain of IL-33 contains multiple protease cleavage sites that are sensitive to these inflammatory proteases .

  • Oxidation: IL-33 contains cysteine residues that can form disulfide bonds upon oxidation, which inactivates the cytokine. This oxidation-dependent inactivation serves as a natural regulatory mechanism to limit IL-33's inflammatory potential .

  • Nuclear sequestration: Though not a classical post-translational modification, the nuclear localization of IL-33 effectively prevents it from engaging its receptor and functions as a regulatory mechanism .

Methodological considerations:

  • Western blot analysis under reducing and non-reducing conditions to assess oxidation state

  • Mass spectrometry to identify specific cleavage sites and oxidation sites

  • Functional assays comparing activity of full-length versus processed forms

  • Site-directed mutagenesis of key residues to determine their importance for bioactivity

What are the challenges in measuring endogenous IL-33 levels in human samples?

Researchers face several technical challenges when measuring IL-33 in human samples:

  • Interference from binding partners: IL-33 binds to soluble ST2 (sST2) and other proteins in biological samples, masking epitopes and preventing antibody binding in standard immunoassays . Commercial IL-33 detection kits often suffer from poor reliability due to this interference, resulting in under-quantitation .

  • Low circulating levels: IL-33 is typically present at very low concentrations in circulation, often below the detection limit of standard assays.

  • Redox state considerations: The oxidation state of IL-33 affects its bioactivity and potentially its detectability. Measurements that don't account for oxidation state may not accurately reflect bioactive IL-33 levels .

Methodological solutions:

  • Acid dissociation treatment disrupts IL-33 binding to endogenous partners like sST2, greatly improving detection sensitivity

  • Simultaneous addition of detection reagent with the capture step in immunoassays enhances detection

  • Optimized assays can achieve lower limits of quantification around 6.25 pg/ml for reduced (active) IL-33 in human serum

  • Specific assay conditions to maintain IL-33 in its reduced state during measurement

Researchers should interpret IL-33 measurements from studies using standard commercial kits cautiously, as they may significantly underestimate actual IL-33 levels due to binding partner interference.

How does IL-33 contribute to disease pathogenesis in different contexts?

IL-33 exhibits context-dependent roles in disease pathogenesis:

Allergic airway diseases: IL-33 promotes type 2 inflammation by activating ILC2s and Th2 cells to produce IL-5 and IL-13, leading to eosinophilia, mucus hypersecretion, and airway hyperresponsiveness . Genome-wide association studies have identified both IL33 and ST2/IL1RL1 genes as being associated with asthma and allergic rhinitis across diverse ethnic groups .

Tissue repair vs. fibrosis: IL-33 can promote tissue repair after acute injury, but chronic IL-33 signaling may contribute to pathological tissue remodeling and fibrosis in various organs .

Autoimmunity: IL-33 has been implicated in autoimmune diseases like rheumatoid arthritis and systemic lupus erythematosus, though its precise roles remain incompletely understood .

Interestingly, despite the strong genetic associations and increased local tissue expression in respiratory diseases, studies have found that serum IL-33 levels may not differ significantly between healthy individuals and people with respiratory disease, or between individuals with different smoking statuses . This suggests that local tissue IL-33 levels and responsiveness, rather than systemic levels, may be more relevant to disease pathogenesis.

Experimental approaches:

  • Tissue-specific conditional knockout models to distinguish local from systemic effects

  • Temporal blockade of IL-33 at different disease stages to determine stage-specific roles

  • Analysis of downstream mediators to identify disease-specific pathways

  • Correlation of disease severity with IL-33 levels in affected tissues

How can pathogens modulate IL-33 signaling to evade immune responses?

Some pathogens have evolved sophisticated mechanisms to interfere with the IL-33 pathway as an immune evasion strategy. The gastrointestinal nematode Heligmosomoides polygyrus bakeri provides an instructive example by secreting an effector protein called Alarmin Release Inhibitor (HpARI2) that suppresses protective immune responses by inhibiting IL-33 signaling .

Structural studies reveal that HpARI2 contains three CCP-like domains and binds directly to mouse IL-33, primarily through the second and third domains . A large loop emerging from the CCP3 domain directly contacts IL-33 at a site that overlaps with the binding site for the ST2 receptor . This competitive binding prevents formation of the IL-33/ST2 signaling complex, effectively blocking IL-33-dependent immune responses.

Functional validation through truncation experiments confirmed this mechanism: HpARI2 variants lacking the large loop from CCP3 failed to block IL-33-mediated signaling in both cell-based assays and in vivo mouse models of asthma .

Research implications:

  • Parasite-derived inhibitors provide structural insights for therapeutic design

  • Competitive receptor binding represents an effective strategy for pathway inhibition

  • Understanding immune evasion mechanisms reveals essential components of protective immunity

  • Natural antagonists can serve as templates for biomimetic therapeutic development

What are the optimal methods for detecting and quantifying IL-33 in biological samples?

Accurate IL-33 measurement requires careful consideration of several factors:

Sample preparation:

  • Implement acid dissociation treatment to release IL-33 from binding partners like soluble ST2

  • Standardize the acid treatment protocol (typically using a low pH buffer followed by neutralization)

  • Process samples rapidly after collection and minimize freeze-thaw cycles

  • Consider adding protease inhibitors to prevent degradation of IL-33 by sample proteases

Immunoassay optimization:

  • Select antibodies that recognize relevant epitopes not masked by binding partners

  • For serum or plasma samples, use methods that account for matrix effects

  • Verify assay detection of both full-length and processed forms if total IL-33 measurement is desired

  • Ensure assay specificity for reduced (active) IL-33 if biological activity assessment is important

Controls and validation:

  • Include spike recovery experiments to assess potential interference

  • Use appropriate positive controls (e.g., samples from allergen challenge models)

  • Run samples at multiple dilutions to identify potential hook effects or inhibition

Alternative approaches:

  • Mass spectrometry-based methods for unbiased detection and characterization

  • Proximity ligation assays to detect IL-33 in complex with binding partners

  • Digital ELISA (Simoa) technologies for ultra-sensitive detection of low abundance IL-33

Researchers should note that measurements of IL-33 in clinical samples have shown that serum IL-33 concentrations appear similar between healthy individuals and people with respiratory disease, despite local tissue differences . This highlights the importance of tissue-specific analysis when possible.

How can researchers effectively distinguish between different forms of IL-33?

Different forms of IL-33 (full-length, processed, oxidized) have varying biological activities. Researchers can distinguish these forms using several approaches:

Biochemical differentiation:

  • Western blotting with antibodies specific to different domains can distinguish full-length from processed forms

  • Use of reducing vs. non-reducing conditions can reveal oxidation state

  • Size exclusion chromatography can separate IL-33 monomers from complexes with binding partners

Functional bioassays:

  • Reporter cell lines expressing ST2 and responsive elements (e.g., NF-κB-luciferase)

  • Primary cell stimulation assays measuring downstream cytokine production

  • Comparison of activity before and after treatment with reducing agents to assess oxidation state

Specific detection methods:

  • Antibody pairs that preferentially detect specific forms of IL-33

  • Mass spectrometry to identify specific cleavage sites and post-translational modifications

  • Protease digestion patterns to distinguish different structural conformations

Implementation strategy:

  • First identify total IL-33 protein levels using antibodies against conserved epitopes

  • Then characterize the forms present using form-specific detection methods

  • Finally, correlate with functional bioactivity using appropriate cellular assays

  • Consider comparing results from multiple detection methods to build a comprehensive profile

What experimental designs best elucidate IL-33's role in complex disease models?

Given IL-33's pleiotropic functions, experimental designs must carefully address temporal, spatial, and cellular complexity:

Temporal considerations:

  • Include multiple time points to capture both early inflammatory and later repair phases

  • Use inducible genetic systems for temporal control of IL-33 or ST2 expression

  • Consider kinetic analysis of IL-33 release, processing, and inactivation

  • Compare acute versus chronic models to distinguish initial versus sustained effects

Cell type-specific approaches:

  • Utilize cell-specific conditional knockout models rather than global deletion

  • Employ bone marrow chimeras to distinguish hematopoietic from non-hematopoietic sources/targets

  • Use adoptive transfer of specific cell populations to determine their contribution

  • Apply single-cell technologies to identify responding cell subsets with high resolution

Intervention strategies:

  • Compare prophylactic versus therapeutic blockade to determine stage-specific roles

  • Use domain-specific blocking antibodies to target specific functions of IL-33

  • Combine IL-33 pathway modulators with treatments targeting other pathways to assess interactions

  • Test dose-dependent effects, as IL-33 may have different outcomes at varying concentrations

Readout selection:

  • Include both inflammatory markers and tissue repair/remodeling indicators

  • Measure functional parameters (e.g., airway hyperresponsiveness) alongside molecular readouts

  • Examine tissue-specific responses across multiple organ systems

  • Consider long-term outcomes to assess chronic effects and potential compensatory mechanisms

Implementing these multifaceted experimental designs provides a more comprehensive understanding of IL-33's context-dependent roles in complex disease states.

What considerations are important when targeting IL-33 therapeutically?

Development of IL-33-targeting therapeutics requires careful consideration of several factors:

Target selection strategies:

  • Direct IL-33 neutralization using antibodies or soluble receptors

  • Receptor antagonism to block IL-33/ST2 interaction

  • Inhibition of IL-33 processing to prevent generation of highly active forms

  • Stabilization of oxidized (inactive) forms of IL-33

  • Blockade of release mechanisms rather than the cytokine itself

Delivery optimization:

  • For respiratory diseases, consider inhaled delivery to maximize local concentration

  • Evaluate tissue penetration capabilities, particularly for antibody-based therapeutics

  • Assess pharmacokinetics in target tissues versus circulation

  • Consider the need for chronic versus acute administration

Patient selection biomarkers:

  • Develop companion diagnostics to measure local or systemic IL-33 activity

  • Identify genetic variants (IL33 or IL1RL1) that predict treatment response

  • Consider disease endotypes that might be particularly IL-33-dependent

  • Establish tissue or blood biomarkers that correlate with IL-33 pathway activation

Safety considerations:

  • Assess potential impact on beneficial IL-33 functions in tissue repair

  • Evaluate risk of compromising anti-parasitic immunity, particularly in endemic regions

  • Monitor effects on regulatory T cell populations that may depend on IL-33 signaling

  • Consider potential compensatory upregulation of related pathways

Learning from natural antagonists:

  • Study parasite-derived inhibitors like HpARI2 that have evolved to block IL-33

  • Apply structural insights from these molecules to therapeutic design

  • Consider biomimetic approaches based on natural antagonist mechanisms

This comprehensive approach to therapeutic development acknowledges both the pathogenic and physiological roles of IL-33, maximizing efficacy while minimizing potential adverse effects.

Product Science Overview

Structure and Expression

IL-33 is an 18 kDa protein composed of 169 amino acid residues . It exists in both an un-cleaved (full-length) and a cleaved form, although the proteins responsible for IL-33 maturation are still under investigation . The recombinant form of human IL-33 is optimized for use in cell culture, differentiation studies, and functional assays .

Function and Mechanism

When IL-33 binds to its receptor, ST2L (also known as IL1RL1), it activates mast cells and innate lymphoid cells, leading to the production of IL-5 and IL-13 . These cytokines are essential for allergic inflammation and type-2 innate immunity . IL-33 also acts as an alarm signal, alerting the immune system to cellular damage .

Biological Activity

IL-33 is involved in regulating type 2-associated innate immune responses . It enhances the activation of Tʜ2 cells, mast cells, and eosinophils, sustaining the humoral response and contributing to adverse Tʜ2 reactions such as dermatitis, asthma, and allergies . Additionally, IL-33 is expressed in blood epithelium and smooth muscle, playing a role in bone protection .

Applications

Recombinant human IL-33 is used in various applications, including:

  • Tʜ2 cell activation and maintenance
  • Investigation of intracellular signaling of IL-1 family and apoptotic/necrotic pathways
  • In vitro inhibition of osteoclast differentiation
Storage and Stability

Recombinant human IL-33 is typically lyophilized and should be reconstituted in sterile water to a concentration of not less than 200 μg/mL . It is stable for 12 months when stored at -20°C in its lyophilized state and for one month under sterile conditions after reconstitution . It is important to avoid repeated freeze/thaw cycles to maintain its stability .

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.