IL33 Mouse, His

Interleukin-33 Mouse Recombinant, His Tag
Shipped with Ice Packs
In Stock

Description

Introduction to IL-33 Mouse, His

Mouse IL-33 with histidine tag refers to recombinant mouse interleukin-33 protein that has been engineered to include a histidine tag, typically at the C-terminus. This modification facilitates protein purification through metal affinity chromatography while maintaining the biological activity of the native protein. Recombinant mouse IL-33 is a valuable research tool for investigating immune responses, inflammatory pathways, and potential therapeutic applications in various disease models .

The mouse IL-33 gene (Il33) is located on chromosome 19qC1 and exhibits unique transcriptional regulation. Unlike many cytokines, IL-33 functions both as a nuclear factor and as a secreted cytokine, making it a fascinating subject for immunological research . His-tagged mouse IL-33 preserves these dual functional properties while providing practical advantages for laboratory applications.

Primary Structure and Domains

Mouse IL-33 protein consists of 266 amino acids and shares approximately 55% homology with its human counterpart (270 amino acids) . The protein can be divided into three functional domains:

  1. Nuclear domain (N-terminal region): Contains a chromatin-binding motif that facilitates nuclear localization and interaction with histone complexes

  2. Central domain: Contains protease cleavage sites sensitive to neutrophil and mast cell-derived proteases

  3. IL-1-like cytokine domain (C-terminal region): Mediates binding to the ST2 receptor and is responsible for cytokine activities

Most commercially available His-tagged mouse IL-33 proteins include the mature form (amino acids 109-266), which encompasses the biologically active IL-1-like cytokine domain .

Gene Expression and Regulation

Mouse IL-33 exhibits a unique pattern of gene expression, with the Il33 gene utilizing two alternative promoters that generate distinct transcripts (Il33a and Il33b). These transcripts differ in their 5' untranslated regions but encode identical proteins. The expression of these transcripts is cell type-specific and stimulus-dependent .

Table 1: Comparison of Il33a and Il33b Transcript Expression Patterns

Tissue/Cell TypeIl33a ExpressionIl33b ExpressionResponse to Stimuli
StomachConstitutiveConstitutiveBoth increase after LPS
LungConstitutiveLow/AbsentBoth increase after LPS
SpleenConstitutiveLow/AbsentBoth increase after LPS
BrainConstitutiveLow/AbsentBoth increase after LPS
BMDCsConstitutiveLowIl33a increases with poly(I:C); both increase with LPS
BMMsNot expressedNot expressedIl33b selectively induced by LPS
MEFsHigh constitutiveHigh constitutiveVariable response to stimuli

BMDCs: Bone marrow-derived dendritic cells; BMMs: Bone marrow-derived macrophages; MEFs: Mouse embryonic fibroblasts; LPS: Lipopolysaccharide; poly(I:C): Polyinosinic:polycytidylic acid

Expression Systems

Recombinant mouse IL-33 with histidine tag is primarily produced using two expression systems:

  1. Escherichia coli expression system: Produces high yields of protein with >98% purity. The recombinant protein typically includes amino acids Ser109-Ile266 with a C-terminal histidine tag. This system is cost-effective but may have limitations in post-translational modifications .

  2. HEK 293 expression system: Produces mouse IL-33 (typically amino acids 109-266) with >95% purity. This mammalian expression system may provide more appropriate post-translational modifications than bacterial systems .

Receptor Binding and Signal Transduction

His-tagged mouse IL-33, like native IL-33, binds to the IL1RL1/ST2 receptor, initiating a cascade of signaling events that activate NF-kappa-B and MAPK signaling pathways in target cells . This interaction is critical for various immune responses, particularly those involving type 2 immunity.

Immunological Functions

Mouse IL-33 exhibits diverse immunological functions that are preserved in the His-tagged recombinant protein:

  1. Th2 Cell Regulation: Induces maturation of T helper type 2 (Th2) cells and stimulates secretion of Th2-associated cytokines

  2. Innate Immune Cell Activation: Activates mast cells, basophils, eosinophils, and natural killer cells, contributing to innate immune responses

  3. Chemoattraction: Acts as a chemoattractant for Th2 cells, facilitating their recruitment to sites of inflammation

  4. Alarmin Function: Functions as an "alarmin" that amplifies immune responses during tissue injury, serving as a bridge between innate and adaptive immunity

  5. Regulatory Functions: Regulates the functions of dendritic cells and influences the development of various T cell subsets, including T follicular helper (Tfh) cells and regulatory T (Treg) cells

Tissue Distribution and Expression

The expression pattern of endogenous mouse IL-33 provides important context for research applications of His-tagged recombinant protein:

Table 3: Tissue Distribution of Endogenous IL-33 in Mice

Tissue/OrganCell Types Expressing IL-33LocalizationExpression Level
Lymphoid OrgansFibroblastic reticular cellsNuclearHigh
LungEpithelial cellsNuclearHigh
SkinEpithelial cellsNuclearHigh
VaginaEpithelial cellsNuclearHigh
StomachEpithelial cellsNuclearHigh
Salivary GlandsEpithelial cellsNuclearHigh
EyeMüller glial cells, ciliary body epithelial cellsNuclearHigh
BrainVarious cell typesNuclearDetectable
Embryonic TissuesOlfactory epithelium, mesenchymal cellsNuclearHigh

IL-33 is consistently localized to the nucleus of producing cells, with no evidence for cytoplasmic localization under homeostatic conditions .

Knockout Mouse Studies

Studies utilizing IL-33 knockout mice (Il33−/− mice) have provided valuable insights into the physiological functions of IL-33:

  1. Behavioral Effects: Il33−/− mice exhibit reduced anxiety-like behaviors in elevated plus maze and open field tests, as well as deficits in social novelty recognition despite intact sociability

  2. Neuronal Activity: Altered c-Fos immunoreactivity (an indicator of neuronal activity) in brain regions implicated in anxiety-related behaviors, such as the medial prefrontal cortex, amygdala, and piriform cortex

  3. Developmental Role: IL-33 may regulate the development and/or maturation of neuronal circuits rather than directly controlling neuronal activities in adult brains

Role in Tertiary Lymphoid Structures

Recent research has identified crucial roles for IL-33 in the formation of tertiary lymphoid structures (TLSs):

  1. TLS Induction: IL-33 deficiency severely attenuates inflammation- and lymphotoxin β receptor (LTβR)-activation-induced TLSs in models of colitis and pancreatic ductal adenocarcinoma (PDAC)

  2. Group 2 Innate Lymphoid Cells: The alarmin domain of IL-33 activates group 2 innate lymphoid cells (ILC2s) expressing lymphotoxin that engage LTβR+ myeloid organizer cells to initiate tertiary lymphoneogenesis

  3. Therapeutic Potential: Engineered recombinant human IL-33 protein expands intratumoural lymphoneogenic ILC2s and TLSs, demonstrating enhanced anti-tumor activity in PDAC mouse models

Quality Control Considerations

When selecting His-tagged mouse IL-33 for research applications, several quality parameters should be considered:

  1. Purity: Higher purity (>95%) ensures reliable experimental results by minimizing interference from contaminants

  2. Endotoxin Levels: Low endotoxin levels (typically <0.1 EU/µg) are essential for in vitro and in vivo applications to prevent non-specific immune activation

  3. Biological Activity: Confirmation of biological activity through cell-based assays ensures the functionality of the recombinant protein

  4. Storage and Stability: Proper storage at -20°C to -80°C and avoidance of repeated freeze-thaw cycles are recommended to maintain protein integrity

Product Specs

Introduction
Interleukin 33 (IL-33) is a 32kDa protein that causes inflammation and may also control gene expression within cells that produce it. IL-33 shares structural similarities with IL-1, a molecule known to stimulate helper T cells to generate type 2 cytokines. IL-33 exerts its effects by interacting with the IL1RL-1 receptor (also called ST2). This interaction triggers the activation of NF-kappa-B and MAP kinases, prompting Th2 cells in laboratory settings to produce cytokines. In living organisms, IL-33 stimulates the production of IL-4, IL-5, and IL-13, leading to substantial pathological alterations in mucosal organs. Notably, IL-33 can be broken down into a 12kDa N-terminal fragment and an 18kDa C-terminal fragment through caspase-1 cleavage.
Description
Recombinant Mouse IL33, produced in E. coli, is a single, non-glycosylated polypeptide chain comprising 181 amino acids (specifically, residues 109-266). With a molecular weight of 18.1kDa, this IL33 variant features a 23-amino acid His-tag attached to its N-terminus. Its purification is achieved through proprietary chromatographic methods.
Physical Appearance
A clear solution that has undergone sterile filtration.
Formulation
The IL33 protein solution is provided at a concentration of 0.5mg/ml. It is formulated in a buffer consisting of phosphate-buffered saline (PBS), 10% glycerol, and 1mM DTT.
Stability
For short-term storage (up to 2-4 weeks), the product should be kept at 4°C. For extended storage, it is recommended to freeze the product at -20°C. To further enhance long-term stability, consider adding a carrier protein like HSA or BSA (0.1% concentration) to the solution. It is crucial to minimize repeated freezing and thawing cycles.
Purity
The purity of the IL33 protein is greater than 85%, as assessed by SDS-PAGE analysis.
Biological Activity
The biological activity of IL33 is evaluated through a cell proliferation assay using D10.G4.1 mouse helper T cells. The ED50, representing the concentration at which IL33 achieves half-maximal activity in this assay, is less than or equal to 0.1ng/ml.
Synonyms
9230117N10Rik, Il-33, Il1f11, NF-HEV, Interleukin-33.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSSIQGTSL LTQSPASLST YNDQSVSFVL ENGCYVINVD DSGKDQEQDQ VLLRYYESPC PASQSGDGVD GKKLMVNMSP IKDTDIWLHA NDKDYSVELQ RGDVSPPEQA FFVLHKKSSD FVSFECKNLP GTYIGVKDNQ LALVEEKDES CNNIMFKLSK I.

Q&A

What is IL-33 and what are its primary functions in mice?

IL-33 is an epithelial-derived cytokine that belongs to the IL-1 family. It acts as an alarmin that is released upon tissue damage, stress, or infection to alert the immune system. In mice, IL-33 has been shown to modulate various immune responses, particularly Th2-type responses, but can also stimulate Th1-type immunity. It binds to its specific receptor ST2, which is expressed on most immune cell populations. IL-33 plays roles in multiple biological processes including inflammatory responses, tissue homeostasis, and neuronal function .

How does IL-33 expression differ across mouse tissues and developmental stages?

IL-33 is predominantly expressed in epithelial barrier tissues, endothelial cells, and tissue-resident immune cells in mice. During development, IL-33 expression patterns change dynamically, with particularly important roles in brain development and maturation. Altered IL-33 expression during development can lead to behavioral changes in adult mice, as evidenced by studies with Il33−/− mice that exhibit reduced anxiety-like behaviors and deficits in social novelty recognition .

What mouse models are commonly used to study IL-33 function?

Several mouse models are used to investigate IL-33 function:

  • IL-33 knockout mice (Il33−/−): These mice have complete absence of IL-33 expression and show altered behaviors, including reduced anxiety and deficits in social novelty recognition .

  • ST2 (IL-33 receptor) knockout mice: These models help distinguish receptor-dependent effects.

  • D-galactose (D-gal)-induced aging models: Used to study IL-33's effects on age-related conditions, these mice exhibit features of accelerated aging and respond to IL-33 treatment with improved cognitive function and bone health .

  • Conditional knockout models: Allow tissue-specific or inducible deletion of IL-33 to examine context-dependent functions.

How should researchers design experiments to discern the direct versus indirect effects of IL-33 in mouse models?

To distinguish direct from indirect effects of IL-33, researchers should:

  • Use cell-specific conditional knockouts to determine which cell types mediate IL-33 responses

  • Implement time-course experiments with acute and chronic IL-33 administration

  • Compare local versus systemic IL-33 delivery methods

  • Employ bone marrow chimeras to distinguish stromal versus hematopoietic IL-33 effects

  • Use ex vivo cell culture systems alongside in vivo models to validate direct cellular effects

For example, in studies examining IL-33's effects on age-related bone loss, researchers used both in vitro osteoblast cultures and in vivo D-galactose-induced aging models to determine that IL-33 directly affects osteoblast function while also modulating T cell populations that indirectly impact bone homeostasis .

What are the critical considerations when generating recombinant IL-33 for mouse studies?

When generating recombinant IL-33 for mouse studies, researchers should consider:

  • Expression construct design: The mature peptide sequence (S109 to I266) of murine IL-33 should be used, as full-length IL-33 is a nuclear protein that may not be properly released. Consider adding a signal sequence (e.g., human CD8α signal sequence) to ensure proper secretion .

  • Protein purification strategy: His-tagged constructs are common for purification, but researchers should be aware that tags might affect protein function.

  • Protein validation: Functional assays should confirm that recombinant IL-33 binds ST2 receptor and activates appropriate signaling pathways.

  • Quality control: Endotoxin testing is essential as contamination can confound immune response studies.

  • Storage conditions: Proper aliquoting and storage at -80°C is recommended to maintain protein activity.

  • Dosing determination: Pilot dose-response studies should establish effective concentrations for specific experimental endpoints.

What methodological approaches provide the most reliable assessment of IL-33 levels in mouse tissues and fluids?

For reliable assessment of IL-33 levels in mouse specimens:

  • ELISA: Use validated antibody pairs with appropriate standards. For example, purified anti-mouse IL-33 antibody (Goat Polyclonal IgG, Poly5165, Biolegend) as the capturing antibody and Biotin anti-mouse IL-33 antibody for detection, with recombinant mouse IL-33 as standard .

  • Immunohistochemistry: Optimize fixation protocols as IL-33 is predominantly nuclear in non-stressed cells.

  • Flow cytometry: For intracellular IL-33 detection in specific cell populations.

  • qPCR: For mRNA expression analysis, though post-transcriptional regulation means protein levels may not correlate with mRNA.

  • Western blotting: To distinguish between full-length and cleaved forms of IL-33.

  • Multiple method validation: Combining techniques provides more robust data than relying on a single approach.

How does IL-33 modulate T cell populations in mouse models, and what are the implications for disease intervention?

IL-33 significantly affects T cell populations with important disease implications:

  • Th17/Treg balance: IL-33 reduces the proliferation of proinflammatory Th17 cells while enhancing regulatory T cell (Treg) numbers. In D-galactose-induced aging mice, IL-33 treatment decreased IL-17A+ cells, suppressed ROR-γt and STAT-3 expression (Th17 transcription factors), and increased Foxp3 expression (Treg transcription factor) .

  • Transcriptional regulation: IL-33 downregulates mRNA expression of ROR-γt and STAT-3 while enhancing Foxp3 expression, shifting the T cell balance toward an anti-inflammatory state .

  • Cytokine modulation: IL-33 treatment reduces proinflammatory cytokines like TNF-α in aged mice .

  • Disease implications: These immunomodulatory effects suggest therapeutic potential for:

    • Age-related bone loss (osteoporosis): Through suppression of inflammatory Th17 cells and enhancement of bone-protective Tregs

    • Neurodegenerative conditions: By reducing inflammatory processes in the brain

    • Allergic diseases: Context-dependent effects where IL-33 blockade may be beneficial

What explains the contradictory pro- and anti-tumorigenic effects of IL-33 in different mouse cancer models?

The dual role of IL-33 in cancer is context-dependent and can be explained by:

  • Tumor microenvironment composition: The dominant immune cell populations present determine whether IL-33 promotes anti-tumor immunity or tumor-supporting inflammation.

  • Specific immune cell activation: IL-33 can stimulate various anti-tumor immune effectors:

    • NK cells: IL-33 enhances NK cell activation markers (NKG2D, CD69), cytotoxic mediators, and tumor-killing capacity .

    • CD8+ T cells: IL-33 can promote CD8+ T cell functions in some contexts.

    • Eosinophils: IL-33 activates eosinophils to exert tumor cytotoxic functions through contact-dependent degranulation .

  • Timing of IL-33 exposure: Early exposure may promote anti-tumor immunity, while chronic exposure might favor immunosuppressive mechanisms.

  • Cancer type: Different tumors have varying sensitivities to immune effector mechanisms modulated by IL-33.

  • Expression pattern: Tumoral expression of IL-33 may inhibit tumor growth by modifying the tumor microenvironment differently than systemic IL-33 .

Researchers should carefully control for these variables when designing experiments to study IL-33 in cancer models.

How does IL-33 signaling interact with helminth infection models in mice?

IL-33 plays a critical role in anti-helminth immunity:

  • Susceptibility: Mice deficient in IL-33 or its receptor ST2 show increased susceptibility to various helminth infections .

  • Immune evasion: Some helminths actively suppress the IL-33 pathway to avoid ejection by the host immune system. For instance, certain mouse-infective helminths secrete the Alarmin Release Inhibitor HpARI2, which suppresses IL-33 signaling .

  • Molecular mechanisms: HpARI2 binds to mouse IL-33 with high affinity (KD ~48 pM) through its CCP domains, preventing IL-33 from interacting with its receptor ST2 .

  • Structural basis: The binding interface between HpARI2 and IL-33 has been characterized, showing that HpARI2 contains three CCP-like domains that contact IL-33 primarily through the second and third domains. This interaction prevents the formation of a signaling complex between IL-33 and ST2 .

  • Allergic connections: The suppression of IL-33 by helminths not only helps parasites evade immunity but may also reduce allergic responses, potentially explaining the observed reduction in asthma prevalence in helminth-endemic regions .

What neurobiological mechanisms explain the behavioral changes observed in IL-33 deficient mice?

IL-33 deficiency leads to notable behavioral alterations through several neurobiological mechanisms:

  • Altered neuronal activity: IL-33−/− mice show changed patterns of c-Fos immunoreactivity (a marker of neuronal activation) in brain regions critical for anxiety-related behaviors, including the medial prefrontal cortex (mPFC), amygdala, and piriform cortex (PCX) .

  • Neurodevelopmental effects: IL-33 appears to play a role in brain development and maturation, with its absence potentially altering neuronal connectivity patterns during critical developmental periods .

  • Social cognition circuits: While IL-33−/− mice maintain intact sociability, they exhibit deficits in social novelty recognition in the three-chamber social interaction test, suggesting specific effects on neural circuits involved in social memory rather than general social motivation .

  • Anxiety-related circuitry: IL-33 deficient mice show reduced anxiety-like behaviors in the elevated plus maze (EPM) and open field test (OFT), indicating altered function in anxiety-processing neural pathways .

  • Neurotransmitter systems: Changes in excitatory/inhibitory balance or monoaminergic signaling may underlie the behavioral phenotypes, though these mechanisms require further investigation.

How should researchers design cognitive and behavioral tests to assess IL-33's effects on mouse brain function?

For robust assessment of IL-33's effects on cognition and behavior:

  • Comprehensive test battery: Include tests for multiple domains:

    • Memory assessment: Novel object recognition test (measures recognition memory)

    • Spatial learning and memory: Morris water maze (evaluates spatial learning and memory retention)

    • Anxiety-like behaviors: Elevated plus maze and open field test

    • Social behaviors: Three-chamber social interaction test (distinguishes sociability from social novelty recognition)

    • Motor function: Analysis of locomotor activity

  • Controlled testing conditions:

    • Use age and sex-matched controls

    • Conduct tests at consistent times of day

    • Control environmental variables (lighting, noise, handling)

    • Include appropriate habituation periods

  • Complementary molecular analyses:

    • Assess c-Fos immunoreactivity to map activated brain regions following behavioral tests

    • Examine neuronal markers in regions showing altered activity

    • Investigate changes in neurotransmitter systems

  • Quantitative assessment methods:

    • For Morris water maze: Track escape latency time across sessions, path length, and time spent in target quadrant

    • For novel object recognition: Calculate discrimination index and total investigation time

How does IL-33 treatment mitigate age-related pathologies in mouse models, and what are the translational implications?

IL-33 treatment shows remarkable efficacy in addressing multiple age-related pathologies:

  • Oxidative stress reduction: IL-33 significantly decreases oxidative stress markers in aging mice:

    • Increases glutathione (GSH) content in cortex and hippocampus

    • Reduces malondialdehyde (MDA) levels and nitrite content

    • Enhances antioxidant defense systems in the brain

  • Cognitive function improvement:

    • Enhances recognition memory (novel object recognition test)

    • Improves spatial learning and memory retention (Morris water maze)

    • Reverses D-galactose-induced memory deficits

  • Amyloid pathology modulation:

    • Downregulates BACE1 protein levels in cortex and hippocampus

    • BACE1 inhibition reduces Aβ burden, a key pathological feature in Alzheimer's disease

  • Tau pathology reduction:

    • Diminishes tau phosphorylation at Ser 396 in cortex and hippocampus

    • Abnormal tau phosphorylation is associated with neuronal death and dementia

  • Immune system modulation:

    • Restores Th17/Treg balance

    • Reduces proinflammatory cytokines like TNF-α

Translational implications include potential therapeutic applications for age-related conditions like osteoporosis and dementia, though further research is needed to determine optimal dosing, administration routes, and safety profiles for human applications.

What methodological approaches best capture IL-33's multisystem effects in age-related pathologies?

To comprehensively evaluate IL-33's effects across multiple physiological systems:

  • Integrated experimental design:

    • Use age-appropriate models (naturally aged mice or accelerated aging models like D-galactose-induced aging)

    • Include both male and female mice to capture sex differences

    • Employ longitudinal studies to track progression of intervention effects

  • Multi-tissue analysis approach:

    • Brain: Assess cognitive function, oxidative stress markers, and protein aggregation

    • Bone: Evaluate bone density, microarchitecture, and cellular composition

    • Immune system: Analyze T cell populations and inflammatory markers

    • Metabolism: Monitor metabolic parameters and body composition

  • Molecular and cellular methodologies:

    • Flow cytometry to analyze immune cell populations (Th17, Treg)

    • Biochemical assays for oxidative stress markers (MDA, nitrite, GSH)

    • Immunoblotting for protein expression (BACE1, phosphorylated tau)

    • RT-PCR for transcription factor analysis (ROR-γt, STAT-3, Foxp3)

  • Functional assessments:

    • Cognitive tests (novel object recognition, Morris water maze)

    • Bone strength and microarchitecture (micro-CT)

    • Physiological parameters (metabolism, activity levels)

  • Tissue-specific intervention comparisons:

    • Compare systemic versus tissue-targeted IL-33 delivery

    • Use tissue-specific IL-33 receptor knockouts to determine primary sites of action

What are the critical quality control steps when preparing and administering IL-33 in mouse experiments?

Ensuring reliable and reproducible IL-33 administration requires:

  • Protein preparation:

    • Use the mature form of mouse IL-33 (S109 to I266) rather than full-length protein

    • Consider adding a signal sequence for secreted expression systems

    • Validate protein structure and function through binding assays

  • Purity assessment:

    • Perform SDS-PAGE and Western blotting to confirm size and identity

    • Test for endotoxin contamination, which can confound immune response studies

    • Confirm protein concentration using validated methods (BCA assay, spectrophotometry)

  • Activity validation:

    • Conduct cell-based assays to confirm biological activity before in vivo use

    • Test receptor binding using surface plasmon resonance (SPR) or similar techniques

  • Administration considerations:

    • Optimize dose through pilot dose-response studies

    • Select appropriate administration route based on experimental goals

    • Maintain consistent timing of administration relative to outcome measurements

    • Use vehicle controls with identical composition minus IL-33

  • Storage and handling:

    • Aliquot protein to avoid freeze-thaw cycles

    • Store at recommended temperatures (typically -80°C)

    • Follow validated reconstitution protocols

How can researchers effectively measure IL-33 signaling activity in different mouse tissues?

Multiple complementary approaches provide comprehensive assessment of IL-33 signaling:

  • Receptor expression analysis:

    • Immunohistochemistry or flow cytometry to quantify ST2 receptor expression

    • RT-PCR for ST2 mRNA levels in different tissues

    • Single-cell RNA sequencing to identify ST2-expressing cell populations

  • Signaling pathway activation:

    • Western blotting for phosphorylated signaling proteins (NF-κB, p38 MAPK, JNK)

    • Immunoprecipitation to detect protein-protein interactions in the signaling cascade

    • Transcriptional reporter assays in cell culture models

  • Downstream gene expression:

    • RT-PCR arrays for IL-33-responsive genes

    • RNA-seq to comprehensively profile transcriptional changes

    • ChIP-seq to identify transcription factor binding sites in regulated genes

  • Functional readouts:

    • Cell-type specific activation markers

    • Cytokine production profiles (ELISA, multiplex assays)

    • Cell population changes (flow cytometry)

  • In vivo reporter systems:

    • Transgenic reporter mice with fluorescent proteins driven by IL-33-responsive elements

    • Bioluminescence imaging for real-time monitoring of signaling activity

How should researchers interpret contradictory findings regarding IL-33's effects across different mouse disease models?

When encountering contradictory findings about IL-33:

  • Context-dependent factors to consider:

    • Disease model specifics (acute vs. chronic, infection vs. sterile inflammation)

    • Genetic background of mice (C57BL/6 vs. BALB/c responses can differ significantly)

    • Sex and age of mice (IL-33 effects may vary with age and between sexes)

    • Dose and timing of IL-33 administration or manipulation

  • Methodological evaluation:

    • Different forms of IL-33 used (full-length vs. mature form)

    • Administration routes (local vs. systemic)

    • Assessment timepoints (early vs. late responses)

    • Knockout strategies (global vs. conditional knockouts)

  • Mechanistic resolution approaches:

    • Cell-specific conditional knockouts to identify key responding cell types

    • Temporal manipulation using inducible systems

    • Dose-response studies across model systems

    • Comprehensive immune profiling to identify divergent downstream mechanisms

  • Translational relevance assessment:

    • Compare findings to human data when available

    • Evaluate whether contradictions reflect genuine biological complexity or methodological issues

    • Consider evolutionary differences in IL-33 signaling between species

What are the most effective experimental approaches to study IL-33's role in neurodegenerative diseases using mouse models?

For studying IL-33 in neurodegeneration:

  • Model selection considerations:

    • Use well-established models relevant to specific diseases (e.g., 5xFAD for Alzheimer's, MPTP for Parkinson's)

    • Consider naturally aged mice alongside genetic models

    • D-galactose-induced aging models are useful for preliminary studies

  • Intervention design:

    • Compare preventive versus therapeutic IL-33 administration

    • Test central (intracerebroventricular) versus peripheral delivery

    • Include dose-response studies to establish optimal concentrations

  • Comprehensive outcome assessment:

    • Cognitive function: Novel object recognition, Morris water maze, Y-maze

    • Neuropathology: Amyloid plaque and tau pathology quantification

    • Neuroinflammation: Microglial and astrocyte activation markers

    • Oxidative stress: MDA, nitrite, and GSH measurements in brain regions

    • Neuronal function: Electrophysiology, synaptic protein expression

  • Molecular mechanism investigation:

    • Assess BACE1 protein levels, which IL-33 can downregulate

    • Measure tau phosphorylation at Ser 396, which IL-33 can diminish

    • Analyze effects on antioxidant defense systems in brain

  • Cell-specific manipulations:

    • Target IL-33 or ST2 in specific cell populations (neurons, microglia, astrocytes)

    • Use cell-specific Cre lines with floxed IL-33 or ST2 alleles

    • Employ bone marrow chimeras to distinguish central vs. peripheral effects

What emerging technologies will advance our understanding of IL-33 biology in mouse models?

Cutting-edge approaches poised to transform IL-33 research include:

  • Single-cell technologies:

    • Single-cell RNA sequencing to identify specific cell populations responding to IL-33

    • Single-cell proteomics to profile signaling pathways at cellular resolution

    • Spatial transcriptomics to map IL-33 and ST2 expression patterns in tissues

  • Advanced imaging:

    • Intravital microscopy to visualize IL-33 responses in living tissues

    • PET imaging with radiolabeled IL-33 to track tissue distribution

    • Optogenetic control of IL-33 expression for precise temporal manipulation

  • Genome editing advances:

    • CRISPR-Cas9 screens to identify novel components of IL-33 signaling

    • Base editing to introduce specific mutations in IL-33 or ST2

    • Tissue-specific and inducible CRISPR systems for precise manipulation

  • Structural biology approaches:

    • Cryo-electron microscopy of IL-33/ST2 complexes in different states

    • Hydrogen-deuterium exchange mass spectrometry to map protein dynamics

    • Structural studies of IL-33 antagonist interactions to guide therapeutic design

  • Systems biology integration:

    • Multi-omics approaches combining transcriptomics, proteomics, and metabolomics

    • Machine learning analysis of complex IL-33-dependent phenotypes

    • Network modeling of IL-33 signaling across tissues and disease states

How can mouse models of IL-33 function inform human therapeutic development?

Translating findings from mouse models to human applications requires:

  • Comparative biology considerations:

    • Analyze structural and functional differences between mouse and human IL-33/ST2

    • Compare expression patterns across species in relevant tissues

    • Evaluate conservation of downstream signaling pathways

  • Humanized mouse models:

    • Mice expressing human IL-33 or ST2 to test species-specific interactions

    • Humanized immune system mice to study human immune cell responses to IL-33

    • Patient-derived xenograft models to test IL-33 therapies in human disease contexts

  • Therapeutic development approaches:

    • Testing recombinant IL-33 for conditions where enhancement is beneficial (neurodegeneration, osteoporosis)

    • Developing IL-33 antagonists or ST2-Fc fusion proteins for allergic or inflammatory conditions

    • Exploring cell-specific delivery systems to target particular tissues

  • Biomarker identification:

    • Correlate mouse responses to IL-33 with potential human biomarkers

    • Develop companion diagnostics to identify patients likely to respond to IL-33-targeted therapies

    • Establish imaging or blood-based markers that predict treatment efficacy

  • Safety assessment strategies:

    • Comprehensive toxicology studies addressing IL-33's pleiotropic effects

    • Long-term studies to identify delayed effects on multiple organ systems

    • Dose-finding studies to establish therapeutic windows

Product Science Overview

Introduction

Interleukin-33 (IL-33) is a cytokine belonging to the IL-1 superfamily. It is known by several synonyms, including C9orf26, DKFZp586H0523, DVS27, NF-HEV, NFEHEV, and RP11-575C20.2 . IL-33 plays a crucial role in the immune system by inducing helper T cells, mast cells, eosinophils, and basophils to produce type 2 cytokines .

Structure and Expression

The recombinant mouse IL-33 protein, tagged with a polyhistidine (His) tag, is expressed in human 293 cells (HEK293). The protein consists of amino acids Serine 109 to Isoleucine 266, with a calculated molecular weight of 19.5 kDa . Due to glycosylation, the protein migrates as 20-30 kDa under reducing conditions in SDS-PAGE .

Biological Function

IL-33 mediates its biological effects by interacting with the receptors ST2 (also known as IL1RL1) and IL-1 Receptor Accessory Protein (IL1RAP). This interaction activates intracellular molecules in the NF-κB and MAP kinase signaling pathways, driving the production of type 2 cytokines such as IL-5 and IL-13 from polarized Th2 cells . In vivo, IL-33 induces the expression of IL-4, IL-5, and IL-13, leading to severe pathological changes in mucosal organs .

Preparation and Purity

The recombinant mouse IL-33 protein is lyophilized from a 0.22 μm filtered solution in PBS, pH 7.4, with trehalose added as a protectant before lyophilization . The protein has a purity of greater than 90% as determined by SDS-PAGE . It is recommended to follow the reconstitution protocol provided in the Certificate of Analysis for optimal performance .

Storage and Stability

For long-term storage, the lyophilized product should be stored at -20°C or lower. It is stable for 12 months in the lyophilized state at -20°C to -70°C and for 3 months under sterile conditions after reconstitution at -70°C . Repeated freeze-thaw cycles should be avoided to maintain protein integrity .

Applications

Recombinant mouse IL-33 protein is used in various research applications, including studying the immune response, cytokine signaling pathways, and the role of IL-33 in diseases. It is also utilized in SDS-PAGE and mass spectrometry (MS) analyses .

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.