IL1RL1 binds IL-33, triggering downstream signaling via MYD88, IRAK1, IRAK4, and TRAF6, leading to MAPK (ERK1/2, p38, JNK) phosphorylation . This pathway modulates immune responses in Th2 cells and mast cells.
IL1RL1’s soluble form acts as a decoy receptor, suppressing Th2 cytokine production in conditions like asthma and sepsis .
The IL1RL1 gene is highly expressed in:
Tissues: Kidney, lung, placenta, stomach, skeletal muscle, colon, and small intestine .
Cell Types: Fibroblasts, mast cells, and activated T helper cells (Th2, Th17) .
Alternative splicing generates a soluble isoform (sST2), which competes with membrane-bound IL1RL1 for IL-33 binding .
IL1RL1 is utilized in:
Immune Response Studies:
Therapeutic Development:
Mechanistic Studies:
IL-33/IL1RL1 Axis in Inflammation:
Role in T Cell Subsets:
Recombinant IL1RL1 protein, expressed in Sf9 Baculovirus cells, is a single, glycosylated polypeptide chain. It comprises 318 amino acids (19-328 a.a.), resulting in a molecular mass of 36.0 kDa. Note that the molecular size on SDS-PAGE may appear between 40-57 kDa.
This IL1RL1 protein is engineered with an 8 amino acid His tag at the C-terminus and purified using proprietary chromatographic techniques.
The IL1RL1 protein solution is provided at a concentration of 0.5 mg/ml and contains 10% glycerol and Phosphate Buffered Saline (pH 7.4).
The biological activity of IL1RL1 is assessed by its ability to inhibit the proliferation of D10.G4.1 mouse helper T cells in the presence of IL-33. The ED50 for this inhibitory effect is determined to be less than or equal to 10 ng/ml in the presence of IL-33.
Interleukin 1 Receptor Like 1, Homolog Of Mouse Growth Stimulation-Expressed, DER4, ST2, T1, Interleukin 1 Receptor-Related Protein, Interleukin-1 Receptor-Like 1, Growth Stimulation-Expressed, Protein ST2, FIT-1, IL33R, ST2L, ST2V, Interleukin-1 receptor-like 1.
KFSKQSWGLE NEALIVRCPR QGKPSYTVDW YYSQTNKSIP TQERNRVFAS GQLLKFLPAA VADSGIYTCI VRSPTFNRTG YANVTIYKKQ SDCNVPDYLM YSTVSGSEKN SKIYCPTIDL YNWTAPLEWF KNCQALQGSR YRAHKSFLVI DNVMTEDAGD YTCKFIHNEN GANYSVTATR SFTVKDEQGF SLFPVIGAPA QNEIKEVEIG KNANLTCSAC FGKGTQFLAA VLWQLNGTKI TDFGEPRIQQ EEGQNQSFSN GLACLDMVLR IADVKEEDLL LQYDCLALNL HGLRRHTVRL SRKNPIDHHS LEHHHHHH.
IL1RL1, also known as Interleukin-1 receptor-like 1 or Protein ST2, is a member of the IL-1 receptor family that functions as a receptor for interleukin-33. When produced in Sf9 Baculovirus cells, IL1RL1 is expressed as a single, glycosylated polypeptide chain containing amino acids 19-328 and is typically fused to an 8 amino acid His Tag at the C-terminus. The complete recombinant protein contains 318 amino acids with a theoretical molecular mass of 36.0kDa, though due to glycosylation it displays multiple bands between 40-57kDa when analyzed by SDS-PAGE under reducing conditions .
The multiple bands observed between 40-57kDa on SDS-PAGE under reducing conditions are primarily attributed to post-translational modifications, specifically variable glycosylation patterns. Sf9 insect cells perform eukaryotic post-translational modifications including glycosylation, but with patterns that differ somewhat from mammalian cells. The heterogeneity in glycosylation results in proteins with slightly different molecular weights, appearing as multiple bands. This is a normal characteristic of glycosylated proteins expressed in insect cell systems and doesn't necessarily indicate protein degradation or contamination .
For producing functional IL1RL1, the Sf9 Baculovirus expression system offers several advantages for researchers. This system supports proper folding and post-translational modifications of complex mammalian proteins. When designing expression constructs, researchers should consider: (1) Codon optimization for insect cell expression; (2) Inclusion of appropriate signal sequences for secretion; (3) Strategic placement of affinity tags (typically C-terminal His-tags) to avoid interference with protein folding or function; (4) Optimization of infection parameters including MOI (multiplicity of infection) and harvest timing to maximize yield of properly folded protein. The resulting recombinant IL1RL1 requires purification by proprietary chromatographic techniques to ensure high purity (>95%) suitable for functional studies .
To maintain IL1RL1 stability, the purified protein should be stored in phosphate-buffered saline (pH 7.4) with 10% glycerol. For short-term storage (2-4 weeks), the protein may be kept at 4°C, but for longer periods, storage at -20°C is recommended. To enhance stability during long-term storage, the addition of carrier proteins (0.1% HSA or BSA) is advisable. Multiple freeze-thaw cycles should be avoided as they can lead to protein degradation and activity loss .
Researchers can evaluate protein integrity through several methods: (1) SDS-PAGE analysis to confirm molecular weight and assess degradation; (2) Western blotting with anti-IL1RL1 or anti-His antibodies; (3) Size-exclusion chromatography to detect aggregation; (4) Functional binding assays with IL-33 to confirm biological activity; and (5) Mass spectrometry for detailed structural analysis and identification of post-translational modifications.
IL1RL1 (particularly the transmembrane variant ST2L) mediates signaling upon IL-33 binding by recruiting a cascade of adapter proteins and kinases. Upon ligand binding, IL1RL1 recruits MYD88, IRAK1, IRAK4, and TRAF6, which leads to the phosphorylation of multiple MAP kinases including MAPK3/ERK1, MAPK1/ERK2, MAPK14, and MAPK8 . This signaling cascade ultimately leads to the activation of transcription factors that regulate inflammatory and immune responses.
Experimental approaches to measure IL1RL1 activity include:
Phosphorylation assays to detect activated MAPKs using phospho-specific antibodies
Reporter gene assays with promoters responsive to IL-33/IL1RL1 signaling
Co-immunoprecipitation experiments to detect protein-protein interactions in the signaling complex
Cytokine production measurement in cells expressing IL1RL1 after IL-33 stimulation
Calcium flux assays to detect immediate signaling events
Gene expression profiling to identify downstream transcriptional changes
Membrane-bound IL1RL1 (ST2L) and its soluble form (sST2) play distinct and sometimes opposing roles in inflammation and immune regulation:
ST2L (membrane-bound form):
Functions as the receptor for IL-33, mediating its pro-inflammatory effects
Expressed on various cell types including T helper cells, endothelial cells, epithelial cells, eosinophils, and mast cells
Activates downstream signaling cascades leading to inflammatory responses
Associated with Th2-related immune functions and allergic responses
sST2 (soluble form):
Acts as a decoy receptor that binds free IL-33, preventing it from interacting with ST2L
Elevated in various inflammatory conditions including asthma, sepsis, and myocardial infarction
May have protective effects in certain inflammatory contexts
This dual system allows for fine-tuning of IL-33-mediated responses, with increased sST2 production potentially serving as a regulatory mechanism to control excessive inflammation .
Multiple genetic studies have identified IL1RL1 polymorphisms associated with asthma and respiratory conditions. In European birth cohorts, IL1RL1 rs102082293, rs10204137 (rs4988955), rs13424006, and rs13431828 (rs13048661) variations were associated with asthma development at school age . Additionally, the variant genotype of IL1RL1 rs13408661/13431828 was associated with current ICS (inhaled corticosteroid) use in former bronchiolitis patients at age 5-7 years and with persistent asthma at age 11-13 years .
These genetic associations may be explained by several functional mechanisms:
Altered expression levels of IL1RL1, affecting the balance between membrane-bound and soluble forms
Modified binding affinity to IL-33, changing downstream signaling intensity
Differential regulation of Th2 inflammatory responses, which are central to allergic asthma pathogenesis
Impact on epithelial barrier function and response to environmental triggers
Evidence suggests that IL1RL1 may be particularly relevant in Th2-like asthma phenotypes, as ST2L expression correlates with Th2 biomarkers in bronchial tissue samples . This supports the notion that targeting the IL-33/IL1RL1 pathway might have therapeutic benefits, especially in severe asthma with Th2 inflammation features .
Recent research has revealed important roles for IL1RL1 in cancer biology, particularly in lung cancer. Studies have shown that IL1RL1 expression is significantly downregulated in both lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC) tissues compared to normal lung tissues . Additionally, specific IL1RL1 polymorphisms, particularly rs12479210 and rs1420101, have been associated with increased lung cancer risk in the Chinese Han population .
The mechanistic involvement of IL1RL1 in cancer biology appears to function through several pathways:
Bioinformatics analyses have confirmed significant differences between six SNPs in IL1RL1 and the expression level of IL1RL1 in lung cancer tissues, suggesting that genetic variations may influence cancer risk through altered gene expression and protein function .
For studying IL1RL1 protein-protein interactions and assessing binding affinity to IL-33, researchers should consider these methodological approaches:
Surface Plasmon Resonance (SPR):
Immobilize purified IL1RL1 on a sensor chip using His-tag
Flow IL-33 at different concentrations over the surface
Analyze association and dissociation rates to determine Kd values
Maintain physiological buffer conditions (PBS pH 7.4 with 0.05% surfactant)
Co-Immunoprecipitation (Co-IP):
Express tagged versions of IL1RL1 and potential binding partners
Use anti-tag antibodies to pull down protein complexes
Analyze by Western blot to detect interactions
Include appropriate controls to rule out non-specific binding
Fluorescence Resonance Energy Transfer (FRET):
Generate fluorescently labeled IL1RL1 and IL-33
Monitor energy transfer as an indication of binding
Perform in solution or in cellular contexts for physiological relevance
Cellular Binding Assays:
Use flow cytometry with fluorescently labeled IL-33
Assess binding to cells expressing IL1RL1
Develop competitive binding assays to determine specificity
For all these methods, it's crucial to use properly folded and glycosylated IL1RL1 from Sf9 cells to ensure physiologically relevant results, as glycosylation patterns may influence binding properties and protein-protein interactions .
Distinguishing between effects mediated by membrane-bound IL1RL1 (ST2L) versus soluble IL1RL1 (sST2) requires careful experimental design:
Selective Expression Systems:
Generate expression constructs for ST2L (full-length) and sST2 (truncated form)
Use cell lines that do not endogenously express IL1RL1
Create stable cell lines expressing either form for comparative studies
Blocking Antibodies and Reagents:
Utilize antibodies that specifically recognize the extracellular domain (both forms) versus transmembrane domain (ST2L only)
Employ recombinant sST2 to competitively inhibit IL-33 binding to ST2L
Design domain-specific peptides that selectively block protein-protein interactions
Genetic Approaches:
Use siRNA/shRNA targeting exons specific to ST2L or sST2
Employ CRISPR-Cas9 to selectively modify domains unique to each form
Create conditional knockout models for temporal control of expression
Functional Readouts:
Measure downstream signaling events (phosphorylation of MAPK3/ERK1, MAPK1/ERK2, etc.) that occur only with membrane-bound receptor activation
Compare cytokine profiles induced by each form
Analyze differential gene expression patterns specific to each form's activity
In vivo approaches:
Administer recombinant sST2 to neutralize IL-33 in animal models
Generate transgenic animals overexpressing either form selectively
Use tissue-specific promoters to control expression in relevant cell types
These strategies enable researchers to parse the distinct biological roles of each form and understand their complementary functions in inflammatory and immune responses .
The search results identify several significant IL1RL1 SNPs associated with disease phenotypes:
Asthma-associated SNPs:
rs102082293, rs10204137 (rs4988955), rs13424006, and rs13431828 (rs13048661) - associated with asthma at school age in European cohorts
rs13408661/13431828 - associated with current ICS use in former bronchiolitis patients and persistent asthma
Bronchiolitis-associated SNPs:
Lung cancer-associated SNPs:
Other allergic phenotype-associated SNPs:
rs3771180, rs3771175, rs10208293, and rs10197862 - investigated in relation to lung cancer but associations not confirmed
rs1420101 - associated with allergic phenotypes including asthma, eczema, and eosinophilia
For genotyping these SNPs, several methodological approaches are recommended:
TaqMan SNP Genotyping Assays:
High-throughput method suitable for large sample sets
Provides accurate allelic discrimination
Requires specialized equipment but is widely available in research facilities
Sequencing-based approaches:
Next-generation sequencing for comprehensive analysis of the IL1RL1 locus
Sanger sequencing for validation of specific variants
Allows detection of novel variants and haplotype determination
Mass Spectrometry-based methods:
MALDI-TOF-based systems for multiplex SNP analysis
High accuracy and relatively high throughput
PCR-RFLP (Restriction Fragment Length Polymorphism):
Cost-effective method for smaller studies
Allows visual confirmation of genotypes through gel electrophoresis
Digital PCR:
Highly sensitive for detection of allele-specific expression
Useful for quantitative assessment of variant alleles
Researchers should select appropriate methods based on their study size, available resources, and required accuracy level.
The relationship between IL1RL1 genotypes and protein expression levels has important implications for interpreting functional studies:
IL1RL1 disease-associated SNPs correlate with IL1RL1 mRNA and serum protein levels of IL-1RL1a (sST2) . This genotype-phenotype correlation provides a mechanistic link between genetic variation and disease susceptibility. Research has shown that:
Expression Quantitative Trait Loci (eQTL) effects:
Certain IL1RL1 SNPs function as eQTLs, directly influencing gene expression levels
Different alleles can lead to variable promoter activity or mRNA stability
This affects the balance between membrane-bound ST2L and soluble sST2 forms
Tissue-specific expression patterns:
The impact of genotypes on expression may vary across different tissues
For example, expression differences between normal and lung cancer tissues show significant associations with specific SNPs
TCGA database analyses confirm differential IL1RL1 expression between normal tissues and lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC)
Biomarker potential:
These correlations have important implications for interpreting functional studies:
Understanding these genotype-expression relationships helps explain the mechanistic basis of disease associations and provides context for interpreting experimental results across different genetic backgrounds .
Studying IL1RL1 in complex diseases with heterogeneous phenotypes like asthma requires multi-faceted approaches:
Phenotype Stratification:
Categorize patients based on clinical characteristics (age of onset, severity, treatment response)
Use biomarkers to identify specific endotypes (e.g., Th2-high vs. Th2-low asthma)
Apply cluster analysis to identify natural disease groupings based on objective parameters
Multi-omics Integration:
Combine genomics (IL1RL1 SNPs), transcriptomics (gene expression), and proteomics (protein levels)
Integrate with clinical data to correlate genetic variations with disease manifestations
Use systems biology approaches to understand network effects beyond single-gene focus
Functional Validation in Model Systems:
Develop mouse models with specific IL1RL1 variants to recapitulate human phenotypes
Use primary human bronchial epithelial cells in air-liquid interface cultures to study IL1RL1 regulation
Employ precision-cut lung slices to maintain tissue architecture while allowing experimental manipulation
Longitudinal Studies:
Track IL1RL1 expression and sST2 levels over time in relation to disease progression
Correlate with environmental exposures and clinical outcomes
Identify temporal patterns that may indicate disease trajectory or treatment response
Clinical Trial Stratification:
Use IL1RL1 genotypes and/or protein levels as stratification biomarkers in clinical trials
Test for differential treatment response based on IL1RL1 status
Develop targeted therapies against the IL-33/IL1RL1 pathway for specific patient subgroups
These comprehensive approaches allow researchers to address the heterogeneity inherent in complex diseases and identify the specific contexts in which IL1RL1 plays a critical role in pathogenesis .
Leveraging IL1RL1 as a therapeutic target requires understanding both its biology and appropriate experimental models for predicting clinical efficacy:
Therapeutic Targeting Strategies:
Blocking antibodies: Develop antibodies against IL1RL1 to prevent IL-33 binding
Soluble decoy receptors: Engineer optimized versions of sST2 with enhanced IL-33 binding
Small molecule inhibitors: Design molecules that disrupt IL1RL1 signaling complexes
Anti-sense oligonucleotides: Reduce IL1RL1 expression in specific tissues
Gene editing approaches: Modify expression or function of IL1RL1 in relevant cell types
Experimental Models for Efficacy Prediction:
In vitro models:
Primary human bronchial epithelial cells (HBECs) in air-liquid interface cultures to assess IL1RL1 regulation in response to therapeutic intervention
Co-culture systems incorporating multiple cell types (epithelial cells, immune cells) to capture complex cellular interactions
Precision-cut lung slices from human donors to maintain tissue architecture and evaluate responses in a more physiological context
Ex vivo models:
Human lung explants to evaluate therapeutic effects on intact human tissue
Bronchial biopsies from patients cultured with potential therapeutics
Patient-derived organoids representing disease-specific phenotypes
In vivo models:
Humanized mouse models expressing human IL1RL1 variants
Mouse models with human-relevant IL1RL1 polymorphisms
Challenge models that recapitulate specific disease triggers (allergen, viral, etc.)
Translational approaches:
Early-phase clinical trials with extensive biomarker analysis
Patient stratification based on IL1RL1 genotype or protein expression levels
Proxy endpoints that correlate with clinical outcomes (e.g., changes in inflammatory biomarkers)
Predictive Biomarkers for Response:
Baseline sST2 levels may predict response to anti-IL-33/IL1RL1 therapies
IL1RL1 genotype could identify likely responders to pathway-targeted interventions
Expression patterns of ST2L on specific immune cell populations might indicate therapeutic susceptibility
Evidence suggests that targeting this pathway may have particular therapeutic benefits in severe asthma, especially in phenotypes characterized by Th2-like inflammation . Both pathologic and genetic approaches support a role for IL1RL1 in severe and Th2-like asthma, making it a promising target for precision medicine approaches .
Several emerging technologies have potential to substantially advance our understanding of IL1RL1 biology:
Single-cell genomics and proteomics:
Single-cell RNA sequencing to identify cell-specific expression patterns of IL1RL1 and its signaling partners
Single-cell proteomics to detect protein-level variation not captured by transcriptomic approaches
Spatial transcriptomics to map IL1RL1 expression within tissue microenvironments
CRISPR-based technologies:
CRISPR activation/interference systems to modulate IL1RL1 expression with high specificity
Base editing to introduce specific SNPs associated with disease risk
Prime editing for precise genomic modifications of regulatory regions
CRISPR screens to identify novel regulators of IL1RL1 expression and function
Advanced imaging technologies:
Super-resolution microscopy to visualize IL1RL1 distribution and clustering at the cell membrane
Live-cell imaging to track receptor dynamics in real-time
Intravital microscopy to observe IL1RL1 function in intact tissues
Computational approaches:
Machine learning algorithms to predict functional consequences of IL1RL1 genetic variants
Network analysis to understand how IL1RL1 integrates with broader signaling systems
Molecular dynamics simulations to model IL-33/IL1RL1 interactions at atomic resolution
Bioengineered model systems:
Organ-on-chip platforms incorporating primary human cells expressing IL1RL1
Microfluidic systems to analyze IL1RL1-mediated cell-cell communication
3D bioprinted tissues with controlled expression of IL1RL1 variants
These technologies will help address critical knowledge gaps, including: understanding cell-type specific roles of IL1RL1, elucidating the functional consequences of genetic variants, mapping temporal dynamics of IL1RL1 signaling, and identifying novel therapeutic strategies targeting this pathway in various disease contexts .
Current limitations in IL1RL1 research present several challenges that future studies must address:
Technical and Methodological Limitations:
Protein heterogeneity: The multiple bands (40-57kDa) observed with recombinant IL1RL1 due to glycosylation patterns complicate structural and functional analyses
Isoform-specific reagents: Limited availability of antibodies and tools that can specifically distinguish between membrane-bound ST2L and soluble sST2 forms
Model systems: Incomplete representation of human disease complexity in current animal and cellular models
Knowledge Gaps:
Regulatory mechanisms: Incomplete understanding of how IL1RL1 expression is regulated in different tissues and disease states
Signaling complexity: Limited knowledge of how IL1RL1 signaling integrates with other pathways in different cellular contexts
Genetic variation: Functional consequences of many IL1RL1 SNPs remain poorly characterized
Tissue-specific roles: The function of IL1RL1 beyond immune and epithelial cells remains understudied
Future Approaches to Address These Challenges:
Develop improved reagents and tools:
Generate isoform-specific antibodies and detection methods
Create reporter systems for tracking ST2L vs. sST2 expression
Design assays to measure specific post-translational modifications
Enhance model systems:
Develop humanized mouse models with relevant IL1RL1 polymorphisms
Create patient-derived organoids that maintain genetic background and disease phenotypes
Establish co-culture systems that recapitulate complex cellular interactions
Apply systems biology approaches:
Integrate multi-omics data to build comprehensive models of IL1RL1 function
Use network analysis to identify key regulators and interaction partners
Develop computational methods to predict functional consequences of genetic variants
Expand clinical correlations:
Conduct larger, well-characterized cohort studies with comprehensive genotyping
Incorporate longitudinal sampling to capture dynamic changes in IL1RL1 expression
Correlate IL1RL1 genetics and expression with detailed clinical phenotyping
Collaborative research initiatives:
Establish biobanks with well-characterized samples from diverse populations
Create shared resources for IL1RL1 reagents and model systems
Develop standardized protocols for measuring IL1RL1 expression and function
By addressing these limitations, future research can provide a more comprehensive understanding of IL1RL1 biology and accelerate the development of targeted therapies for conditions including asthma, bronchiolitis, and potentially cancer .
</thinking>This comprehensive FAQ collection addresses key research considerations regarding IL1RL1 (Interleukin-1 receptor-like 1) human protein expressed in Sf9 systems. The questions are structured to support both fundamental understanding and advanced research applications, with emphasis on methodological approaches for academic researchers.
IL1RL1, also known as Interleukin-1 receptor-like 1 or Protein ST2, is a member of the IL-1 receptor family that functions as a receptor for interleukin-33. When produced in Sf9 Baculovirus cells, IL1RL1 is expressed as a single, glycosylated polypeptide chain spanning amino acids 19-328 and typically fused to an 8 amino acid His Tag at the C-terminus. The complete recombinant protein contains 318 amino acids with a theoretical molecular mass of 36.0kDa, though due to glycosylation it displays multiple bands between 40-57kDa on SDS-PAGE under reducing conditions .
The multiple bands observed between 40-57kDa on SDS-PAGE under reducing conditions are primarily attributed to post-translational modifications, specifically variable glycosylation patterns. Sf9 insect cells perform eukaryotic post-translational modifications including glycosylation, but with patterns that differ from mammalian cells. The heterogeneity in glycosylation results in proteins with slightly different molecular weights, appearing as multiple bands. For research applications, this heterogeneity should be documented when characterizing the protein preparation, but doesn't necessarily indicate degradation or compromised functionality .
For producing functional IL1RL1, the Sf9 Baculovirus expression system offers several advantages for researchers. This system requires careful optimization across multiple parameters:
Expression construct design:
Codon optimization for insect cell expression
Inclusion of appropriate signal sequences for secretion
Strategic placement of affinity tags (typically C-terminal His-tags)
Selection of promoters for optimal expression timing
Infection parameters:
MOI (multiplicity of infection) optimization
Time-course analysis to determine peak expression
Cell density at infection time
Purification considerations:
Multiple chromatographic steps typically required
Careful buffer selection to maintain stability
Monitoring of glycosylation patterns during purification
The resulting recombinant IL1RL1 requires purification to ensure high purity (>95%) suitable for functional studies .
Evaluating functional integrity of IL1RL1 requires multiple orthogonal approaches:
Binding assays:
Surface plasmon resonance (SPR) with immobilized IL-33
ELISA-based binding assays
Cell-based binding assays using IL-33-responsive cells
Structural analysis:
Circular dichroism to assess secondary structure
Limited proteolysis to evaluate folding integrity
Thermal shift assays to determine stability
Mass spectrometry to confirm post-translational modifications
Signaling activity:
Measuring downstream phosphorylation of MAPK3/ERK1, MAPK1/ERK2, MAPK14, and MAPK8 in appropriate cell lines
Analyzing recruitment of MYD88, IRAK1, IRAK4, and TRAF6 in cell-based systems
Reporter gene assays to monitor transcriptional activation
Researchers should maintain proper storage conditions in phosphate-buffered saline (pH 7.4) with 10% glycerol at -20°C for long-term storage with carrier proteins (0.1% HSA or BSA) while avoiding multiple freeze-thaw cycles .
IL1RL1 (particularly the transmembrane variant ST2L) initiates a complex signaling cascade upon IL-33 binding. This process involves:
Receptor activation: Upon binding IL-33, IL1RL1 recruits MYD88, IRAK1, IRAK4, and TRAF6
Kinase activation: This complex leads to phosphorylation of multiple MAP kinases including MAPK3/ERK1, MAPK1/ERK2, MAPK14, and MAPK8
Transcriptional regulation: Activated kinases modulate transcription factors controlling inflammatory and immune responses
Experimental approaches to measure this activity include:
Phospho-specific Western blotting to detect activated MAPK signaling
Co-immunoprecipitation to analyze protein complex formation
Luciferase reporter assays with IL-33-responsive promoters
Quantitative RT-PCR to measure induction of downstream target genes
Flow cytometry to assess phospho-protein levels at single-cell resolution
Proximity ligation assays to detect protein-protein interactions in situ
These methods can be combined to provide a comprehensive understanding of IL1RL1 signaling dynamics in different cellular contexts .
Membrane-bound IL1RL1 (ST2L) and its soluble form (sST2) have distinct and sometimes opposing functions in inflammation and immune regulation:
ST2L (membrane-bound form):
Functions as the receptor for IL-33, mediating its pro-inflammatory effects
Expressed on various cell types including T helper cells, endothelial cells, epithelial cells, eosinophils, and mast cells
Activates downstream signaling cascades leading to inflammatory responses
Associated with Th2-related immune functions and allergic responses
sST2 (soluble form):
Acts as a decoy receptor that binds free IL-33, preventing it from interacting with ST2L
Elevated in various inflammatory conditions including asthma, sepsis, and myocardial infarction
Functions as an immunomodulatory molecule with potential protective effects
This dual system allows for fine-tuning of IL-33-mediated responses, with increased sST2 production potentially serving as a regulatory mechanism to control excessive inflammation. Researchers investigating IL1RL1 should carefully distinguish between these forms when designing experiments and interpreting results .
Multiple genetic studies have identified IL1RL1 polymorphisms associated with respiratory conditions:
Asthma associations:
European birth cohorts identified rs102082293, rs10204137 (rs4988955), rs13424006, and rs13431828 (rs13048661) as significantly associated with asthma development at school age
The variant genotype of IL1RL1 rs13408661/13431828 showed association with current ICS (inhaled corticosteroid) use in former bronchiolitis patients at age 5-7 years and with persistent asthma at age 11-13 years
Evidence suggests IL1RL1 may be particularly relevant in Th2-like asthma phenotypes, as ST2L expression correlates with Th2 biomarkers in bronchial tissue samples
Bronchiolitis associations:
The IL1RL1 rs1921622 SNP was associated with severe bronchiolitis compared with controls
Severe bronchiolitis was associated with higher soluble IL1RL1-a concentrations in nasopharyngeal aspirates
Functionally, these genetic variations can impact:
Understanding these genetic associations helps identify at-risk populations and may guide development of targeted therapies for patients with specific IL1RL1 variants .
Recent research has revealed important roles for IL1RL1 in cancer biology:
These findings suggest IL1RL1 may serve as both a biomarker and potential therapeutic target in certain cancers, particularly lung cancer, though mechanistic details require further investigation .
Distinguishing between effects mediated by membrane-bound IL1RL1 (ST2L) versus soluble IL1RL1 (sST2) requires sophisticated experimental design:
Selective Expression Systems:
Generate expression constructs for ST2L (full-length) and sST2 (truncated form)
Use cell lines that do not endogenously express IL1RL1
Create stable cell lines expressing either form for comparative studies
Genetic Approaches:
Use siRNA/shRNA targeting exons specific to ST2L or sST2
Employ CRISPR-Cas9 to selectively modify domains unique to each form
Design splice-switching oligonucleotides to modulate relative expression of isoforms
Blocking Reagents:
Utilize antibodies that specifically recognize the extracellular domain (both forms) versus transmembrane domain (ST2L only)
Employ recombinant sST2 to competitively inhibit IL-33 binding to ST2L
Design domain-specific peptides that selectively block protein-protein interactions
Functional Readouts:
Measure downstream signaling events (phosphorylation cascades) that occur only with membrane-bound receptor activation
Compare cytokine profiles induced by each form
Analyze differential gene expression patterns specific to each form's activity
Experimental Controls:
Include appropriate negative controls lacking IL-33 stimulation
Use IL-33 mutants with differential binding to ST2L vs. sST2
Control for transfection/transduction efficiency in expression systems
These strategies enable researchers to parse the distinct biological roles of each form and understand their complementary functions in inflammatory and immune responses .
Studying IL1RL1 polymorphisms in relation to disease phenotypes requires comprehensive methodological approaches:
Study Design Considerations:
Prospective cohort studies to assess polymorphism impact on disease development
Case-control studies to evaluate associations with established disease
Family-based designs to control for population stratification
Longitudinal follow-up to capture age-dependent phenotypes
Adequate sample size calculations based on expected effect sizes
Genotyping Methods:
TaqMan SNP genotyping assays for targeted polymorphism analysis
Next-generation sequencing for comprehensive variant detection
Custom genotyping arrays for high-throughput analysis
Digital PCR for highly sensitive allele discrimination
Validation with secondary methods to confirm results
Phenotyping Approaches:
Standardized clinical assessments (e.g., spirometry for asthma)
Biomarker measurements (serum sST2 levels, IL-33 levels)
Detailed clinical characterization to identify disease subtypes
Objective measures of disease severity and progression
Response to specific treatments or challenges
Statistical Analysis:
Appropriate adjustment for multiple testing
Haplotype analysis rather than single SNP associations
Gene-environment interaction assessments
Mediation analysis to determine mechanisms of effect
Meta-analysis to combine results across studies
Functional Validation:
Expression quantitative trait loci (eQTL) analysis
In vitro reporter assays to assess variant impact on expression
CRISPR-based editing to introduce specific variants
Protein-level assessments (Western blotting, ELISA)
Signaling studies to determine functional consequences
These methodological approaches have successfully identified significant associations, such as IL1RL1 rs13408661/13431828 with persistent asthma and rs12479210/rs1420101 with lung cancer risk .
Multi-omics approaches provide powerful frameworks for understanding IL1RL1 in complex diseases:
Integrated Genomics:
Whole genome/exome sequencing to identify rare variants beyond common SNPs
Epigenomic profiling (methylation, chromatin accessibility) to understand regulatory mechanisms
Targeted sequencing of the entire IL1RL1 locus including regulatory regions
Analysis of structural variants and copy number variations affecting IL1RL1
Transcriptomics:
RNA-seq to identify disease-specific expression patterns
Single-cell RNA-seq to define cell-type specific IL1RL1 expression
Alternative splicing analysis to quantify ST2L vs. sST2 isoform ratios
Long-read sequencing to identify novel IL1RL1 isoforms
Proteomics:
Mass spectrometry to analyze IL1RL1 post-translational modifications
Targeted protein quantification of ST2L and sST2 in various tissues
Interactome mapping to identify novel IL1RL1 binding partners
Phosphoproteomics to characterize IL1RL1 signaling dynamics
Metabolomics:
Identification of metabolic signatures associated with IL1RL1 activation
Analysis of lipid mediators impacted by IL-33/IL1RL1 signaling
Correlation of metabolic profiles with sST2 levels
Data Integration Approaches:
Network analysis to position IL1RL1 within broader disease pathways
Machine learning to identify patterns across multi-omic datasets
Bayesian approaches to infer causal relationships
Systems biology modeling of IL1RL1 signaling networks
This integrated approach can reveal mechanisms by which IL1RL1 variants influence disease risk and progression across conditions like asthma, bronchiolitis, and lung cancer, potentially identifying novel biomarkers and therapeutic targets .
Translating IL1RL1 research into clinical applications faces several significant challenges:
Biological Complexity:
Dual roles of membrane-bound versus soluble forms with potentially opposing functions
Tissue and cell-type specific expression patterns requiring targeted approaches
Complex post-translational modifications affecting protein function
Multiple genetic variants with different functional consequences
Technical Limitations:
Lack of standardized assays for measuring ST2L versus sST2 in clinical samples
Heterogeneity in glycosylation patterns affecting protein function and detection
Challenges in developing specific antibodies distinguishing protein isoforms
Difficulties in maintaining proper protein folding in recombinant systems
Clinical Translation Barriers:
Phenotypic heterogeneity within disease categories (e.g., asthma subtypes)
Population differences in genetic variant frequencies and functional impacts
Need for large, well-characterized cohorts for validating genetic associations
Determining optimal timing for therapeutic interventions targeting this pathway
Therapeutic Development Challenges:
Specificity in targeting IL-33/IL1RL1 interaction without affecting other pathways
Achieving tissue-specific delivery of therapeutics
Identifying appropriate patient populations most likely to benefit
Developing suitable biomarkers for monitoring treatment response
Regulatory and Practical Considerations:
Standardizing IL1RL1/sST2 as a clinical biomarker across laboratories
Cost-effectiveness of genotyping for clinical decision-making
Integration with existing clinical algorithms and practice guidelines
Demonstrating improved outcomes compared to standard treatments
Addressing these challenges requires coordinated efforts across basic science, translational research, and clinical studies to fully realize the potential of IL1RL1 as a therapeutic target and biomarker in conditions ranging from asthma and bronchiolitis to potentially lung cancer .
Several innovative experimental models show promise for advancing IL1RL1 research:
Advanced Cell Culture Systems:
Air-liquid interface cultures of primary human bronchial epithelial cells for studying IL1RL1 regulation in a physiologically relevant context
Co-culture systems incorporating multiple cell types (epithelial, immune, stromal) to recapitulate complex cellular interactions
Microfluidic organ-on-chip platforms to study IL1RL1 function under physiological flow conditions
Organoid Technologies:
Patient-derived lung organoids for studying IL1RL1 in disease-specific contexts
Immune-competent organoid systems incorporating both structural and immune cells
Brain organoids to study neuroinflammatory aspects of IL1RL1 signaling
Advanced Animal Models:
Humanized mouse models expressing human IL1RL1 variants
CRISPR-engineered mice harboring specific human IL1RL1 polymorphisms
Conditional and inducible IL1RL1 knockout/knockin models for temporal control
Tissue-specific IL1RL1 models to delineate organ-specific functions
Ex Vivo Human Systems:
Precision-cut lung slices from human donors with defined IL1RL1 genotypes
Explant cultures from surgical specimens for pharmacological studies
Human lung-on-chip microfluidic devices incorporating IL1RL1 variants
Computational and In Silico Models:
Machine learning approaches to predict functional consequences of IL1RL1 variants
Molecular dynamics simulations of IL-33/IL1RL1 interactions
Network models integrating IL1RL1 signaling with broader inflammatory pathways
Virtual patient cohorts for in silico clinical trials of IL1RL1-targeted therapies
These advanced models can help address key knowledge gaps, including understanding cell type-specific functions, elucidating the consequences of genetic variations, and identifying optimal therapeutic targeting strategies .
The IL-33/IL1RL1 pathway presents several promising therapeutic targeting strategies:
Direct Pathway Inhibition:
Anti-IL1RL1 monoclonal antibodies to block IL-33 binding
Recombinant sST2 proteins as decoy receptors
Small molecule inhibitors of IL1RL1 signaling
IL-33 neutralizing antibodies or traps
Peptide inhibitors targeting critical interaction domains
Genetic and RNA-based Approaches:
Antisense oligonucleotides to modulate ST2L vs. sST2 splicing ratios
siRNA/shRNA to selectively knock down IL1RL1 expression
mRNA therapeutics to deliver modified sST2 as a decoy receptor
CRISPR-based gene editing for patients with high-risk variants
Pathway-Specific Targeting:
Inhibitors of downstream signaling components (IRAK1/4, TRAF6)
Modulators of IL1RL1 expression (targeting transcriptional regulators)
Agents affecting IL1RL1 trafficking and membrane localization
Compounds influencing IL1RL1 post-translational modifications
Precision Medicine Applications:
IL1RL1 genotype-guided therapy selection
sST2 levels as biomarkers for patient stratification and response monitoring
Combination therapies targeting IL1RL1 along with other pathways
Tissue-specific delivery approaches for localized effects
Disease-Specific Approaches:
For asthma: Inhaled formulations targeting bronchial epithelium
For lung cancer: Combination with immune checkpoint inhibitors
For bronchiolitis: Early intervention in genetically susceptible infants
For allergic conditions: Desensitization protocols with IL1RL1 modulation
The strongest evidence currently supports targeting this pathway in Th2-driven inflammatory conditions like severe asthma . Both pathologic and genetic approaches confirm IL1RL1's role in these conditions, suggesting that these therapeutic strategies may benefit specific patient populations identified through biomarker and genetic screening .
Rigorous quality control is critical when working with recombinant IL1RL1 from Sf9 cells:
Purity Assessment:
SDS-PAGE analysis with Coomassie or silver staining (>95% purity standard)
Size-exclusion chromatography to detect aggregates and multimers
Western blot with specific antibodies to confirm identity
Mass spectrometry for accurate molecular weight determination
Endotoxin testing to ensure preparation is endotoxin-free (<1 EU/mg)
Structural Integrity:
Circular dichroism spectroscopy to assess secondary structure
Thermal shift assays to determine stability profiles
Limited proteolysis to evaluate folding quality
Dynamic light scattering to assess homogeneity and aggregation state
N-terminal sequencing to confirm correct processing
Glycosylation Analysis:
Lectin blotting to characterize glycan patterns
PNGase F treatment to assess contribution of N-glycans
Mass spectrometry to map glycosylation sites
Comparison of glycoform patterns between batches for consistency
Functional impact assessment of different glycoforms
Functional Validation:
IL-33 binding assays with quantitative affinity measurements
Cell-based activity assays measuring downstream signaling
Comparative analysis with mammalian-expressed IL1RL1
Lot-to-lot consistency testing with reference standards
Stability studies under various storage conditions
Documentation and Reporting:
Detailed batch records including expression conditions
Certificate of analysis with all quality parameters
Stability data under recommended storage conditions
Validation of performance in standard assay systems
Transparent reporting of heterogeneity in publications
These quality control measures ensure that experimental results using IL1RL1 are reliable and reproducible, which is essential for advancing our understanding of its biology and therapeutic potential .
Optimal experimental designs for studying IL1RL1 genetic variants require careful consideration of multiple factors:
Cohort Selection and Design:
Prospective cohorts with longitudinal follow-up
Enables assessment of variant impact on disease development
Allows evaluation of age-dependent effects
Provides opportunity for repeated biomarker measurements
Nested case-control studies within established cohorts
Efficient use of resources
Matching on important confounders
Availability of pre-disease samples
Family-based designs
Controls for population stratification
Enables transmission disequilibrium testing
Allows study of rare variants in familial cases
Sample Size Considerations:
Power calculations based on expected effect sizes from previous studies
Adjustment for multiple testing when analyzing multiple variants
Consideration of gene-environment interactions requiring larger samples
Planning for replication cohorts to validate findings
Genotyping Strategy:
Targeted approach focusing on known IL1RL1 SNPs:
Comprehensive sequencing approach:
Capture of regulatory regions beyond coding sequences
Identification of rare variants not covered in genotyping arrays
Detection of structural variants affecting IL1RL1 locus
Phenotyping Depth:
Detailed clinical characterization to identify disease subtypes
Molecular phenotyping (IL1RL1 expression levels, ST2L vs. sST2 ratio)
Biomarker panels to capture pathway activity
Treatment response as a phenotypic outcome
Environmental exposure assessment for interaction studies
Analysis Considerations:
Haplotype analysis rather than single SNP approach
Proper adjustment for ancestry and population stratification
Assessment of gene-environment interactions
Integrative analysis incorporating expression data (eQTL)
Meta-analysis across multiple cohorts for robust findings
Interleukin-1 Receptor Like-1 (IL1RL1), also known as ST2, is a member of the interleukin-1 receptor family. It is a transmembrane protein that plays a crucial role in the immune system by acting as a receptor for interleukin-33 (IL-33). The IL1RL1 gene encodes this protein, which is involved in various immune responses and has been implicated in several diseases.
IL1RL1 is a transmembrane protein with a structure similar to IL-1R1. It is composed of a single, glycosylated polypeptide chain and is fused to an 8 amino acid His Tag at the C-terminus. The protein has a total of 318 amino acids and a molecular mass of approximately 36.0 kDa . When expressed in Sf9 Baculovirus cells, IL1RL1 shows multiple bands between 40-57 kDa on SDS-PAGE under reducing conditions .
IL1RL1 functions as a receptor for IL-33. Upon binding to IL-33, IL1RL1 recruits several signaling molecules, including MYD88, IRAK1, IRAK4, and TRAF6. This recruitment leads to the phosphorylation of MAPK3/ERK1, MAPK1/ERK2, MAPK14, and MAPK8, which are involved in various signaling pathways . IL1RL1 is thought to play a role in helper T-cell function and is highly expressed in tissues such as the kidney, lung, placenta, stomach, skeletal muscle, colon, and small intestine .
IL1RL1 is involved in both innate and adaptive immunity. It contributes to tissue homeostasis and responses to environmental stresses. The receptor is also implicated in the function of helper T cells and may be induced by proinflammatory stimuli . Additionally, IL1RL1 has been shown to act as a negative regulator of Th2 cytokine production. High levels of soluble IL1RL1 have been reported in several disease states, including asthma, sepsis, and myocardial infarction .
Recombinant IL1RL1 produced in Sf9 Baculovirus cells is a valuable tool for research. It is available as a sterile filtered colorless solution and is formulated with phosphate-buffered saline (pH 7.4) and 10% glycerol . The protein is highly pure, with a purity greater than 95.0% as determined by SDS-PAGE . It is recommended to store the protein at 4°C if used within 2-4 weeks, or at -20°C for longer periods. For long-term storage, adding a carrier protein such as 0.1% HSA or BSA is advised to avoid multiple freeze-thaw cycles .
Recombinant IL1RL1 is used in various research applications, including studies on immune responses, signaling pathways, and disease mechanisms. It is particularly useful for investigating the role of IL-33 and its receptor in different physiological and pathological conditions.