IL1RAP is a member of the Interleukin-1 receptor family. Mature human IL1RAP consists of a 347 amino acid extracellular domain (ECD) with three Ig-like domains, a 21 amino acid transmembrane segment, and a 182 amino acid cytoplasmic domain . The recombinant form typically consists of the extracellular domain spanning from Ser21 to Glu359 . IL1RAP exists in multiple isoforms, including membrane-bound (mIL1RAP) and soluble (sIL1RAP) forms, with the latter being formed by the extracellular domain of mIL1RAP .
The functional architecture of IL1RAP contains several critical domains that determine its signaling capabilities:
Extracellular Region: Contains a small hydrophobic patch formed by three amino acid residues: Ile 135, Leu 180, and Ile 181, which is essential for binding to IL-1Rs .
D2 and D3 Domains: These extracellular domains interact with corresponding D2 and D3 domains of primary receptors . While initially characterized for IL-36R interactions, similar binding patterns likely occur with other IL-1 family receptors due to conserved cysteine residues that form disulfide bonds promoting the 3D structure of receptors .
Transmembrane Domain: Anchors the protein in the cell membrane.
Cytoplasmic Domain: Critical for downstream signaling through recruitment of adaptor proteins like MyD88 and IRAK-4, leading to MAPK pathway activation .
The functional importance of these domains is evident in experiments where disruption of IL1RAP interactions inhibits signaling through multiple pathways, including IL-1, IL-33, and IL-36 signaling cascades.
IL1RAP is ubiquitously expressed across multiple tissues including the liver, placenta, and white blood cells . In the hematopoietic system, IL1RAP is expressed at lower levels on normal hematopoietic stem and progenitor cells (HSPCs) compared to their malignant counterparts . The soluble form (sIL1RAP) is found in circulation at relatively high concentrations, with a reported median of approximately 300 ng/mL (range: 92–600 ng/mL) in healthy individuals . Expression varies across different immune cell types, with differential expression observed on monocytes, neutrophils, T cells, and macrophages as demonstrated by flow cytometry .
IL1RAP is significantly upregulated in various hematological malignancies, particularly in leukemic stem cells, compared to normal counterparts. Expression analysis across different leukemia subtypes reveals distinct patterns:
Gene expression correlation analyses show that high IL1RAP expression in AML is associated with "mitochondrial translation elongation and termination," "energy production via oxidative phosphorylation," and a "leukemic granulocyte-monocyte progenitor signature," whereas low IL1RAP expression correlates with "regulation of RNA metabolic processes," "gene expression," and a glycolysis-enriched HSC-like signature .
Multiple complementary techniques provide comprehensive analysis of IL1RAP expression:
Flow Cytometry: Quantifies surface protein expression using fluorescently-labeled antibodies, measuring mean fluorescence intensity (MFI). This approach can detect co-expression with other surface markers (CD135, CD123, CD117) .
RNA Sequencing: Measures transcript levels and enables correlation with genome-wide expression patterns. Methods include batch correction, normalization via LogNormalize method, and scaling by a factor of 10,000 followed by log1p-transformation .
Quantitative Proteomics: Assesses protein abundance, with the advantage of detecting post-translational modifications.
Gene-Based Clustering: Employs modularity optimization methods using graph-based approaches to identify gene expression patterns throughout disease progression .
Western Blotting: Evaluates protein expression in cell lysates, particularly useful for detecting different IL1RAP isoforms.
For comprehensive analysis, researchers should combine multiple methods to verify consistency across transcriptional and protein levels.
Production of high-quality recombinant human IL1RAP involves several critical steps:
Expression Construct Design: Most commonly involves cloning the extracellular domain (ECD, Ser21-Glu359) with a C-terminal 6-His tag for purification .
Expression System Selection: Mammalian expression systems (e.g., ExpiCHO cells) are preferred to ensure proper folding and post-translational modifications .
Purification Strategy: Affinity chromatography using the His-tag, followed by additional purification steps to achieve >90% purity.
Quality Control: Size exclusion chromatography (SEC) and SDS-PAGE to verify purity and molecular weight .
Formulation and Storage: Typically lyophilized from a 0.2 μm filtered solution in PBS and reconstituted at 200 μg/mL in PBS .
For carrier-free preparations (without BSA), special consideration must be given to protein stability during storage and handling .
Several complementary assays provide robust assessment of IL1RAP functionality:
Binding Assays:
Signaling Assays:
Functional Cellular Assays:
Co-immunoprecipitation Studies:
Each assay should include appropriate positive and negative controls to ensure specificity and reproducibility of results.
IL1RAP plays multifaceted roles in hematological malignancies beyond its canonical function in IL-1 signaling:
Co-receptor Function Expansion: IL1RAP physically interacts with and mediates signaling through FLT3 and c-KIT, two receptor tyrosine kinases with established roles in AML pathogenesis .
Leukemic Stem Cell Support: IL1RAP is consistently overexpressed on leukemic stem cells across various genetic subtypes of AML, CML, and high-risk MDS .
IL-1 Pathway Activation: In CML, primitive (CD34+CD38−) cells express functional IL-1 receptor complexes and respond with NF-κB activation and marked proliferation in response to IL-1, effects that can be blocked by IL1RAP antibodies .
Growth Advantage Mechanism: Knockdown of IL1RAP significantly reduces colony-forming capacity of primary MDS/AML cells, indicating its role in promoting leukemic cell growth and survival .
Flow cytometry analysis of 124 primary AML patients revealed that IL1RAP is frequently co-expressed with other important signaling receptors:
21% of patients were CD135+IL1RAP+
4% expressed IL1RAP without CD135
29.8% were IL1RAP+CD117+
These co-expression patterns suggest potential synergistic effects between IL1RAP and other oncogenic signaling pathways.
Beyond hematological malignancies, IL1RAP is implicated in several inflammatory and metabolic conditions:
Atherosclerosis: IL1RAP blockade limits plaque development and inflammation in apolipoprotein E–deficient (Apoe−/−) mice, suggesting its role in atherosclerotic disease progression .
Myocarditis: Studies in mouse models of coxsackievirus B3 (CVB3)–mediated and experimental autoimmune myocarditis (EAM) showed that IL1RAP blockade reduced disease severity and preserved cardiac function .
Non-alcoholic Fatty Liver Disease (NAFLD): The QSOX1/IL1RAP ratio has been identified as a novel biomarker for NAFLD severity, with elevated QSOX1 and reduced IL1RAP levels associated with increasing disease severity .
Systemic Sclerosis: IL1RAP has been implicated in fibrotic processes, with therapeutic antibodies targeting IL1RAP under development for treatment of inflammatory and fibrotic diseases .
In these conditions, IL1RAP appears to function as an amplifier of inflammatory signaling by mediating the effects of multiple IL-1 family cytokines simultaneously.
Several approaches for targeting IL1RAP have been developed and evaluated in preclinical and clinical settings:
Blocking Antibodies:
ADCC-Mediating Antibodies:
Genetic Approaches:
T Cell Engagers:
These approaches have shown efficacy in various preclinical models, with several advancing to clinical development. The first-in-human phase 1 dose-escalation study of CAN04 (an IL1RAP-targeting antibody) demonstrated that it can be safely administered to patients up to 10.0 mg/kg weekly .
Comprehensive evaluation of IL1RAP-targeting therapies requires multi-parameter assessment:
In Vitro Assays:
In Vivo Disease Models:
Efficacy Parameters:
Disease-specific endpoints: Leukemic burden, plaque development, cardiac function.
Molecular endpoints: Changes in signaling pathway activation, gene expression profiles.
Cellular endpoints: Immune cell infiltration, differentiation status.
Advanced Analytical Methods:
For myocarditis models, IL1RAP blockade demonstrated efficacy without impeding viral clearance from the heart while significantly reducing the numbers of infiltrating immune cells (CD4+ T cells, Ly6C+CCR2+ monocytes, neutrophils, and eosinophils) . In atherosclerosis models, treatment with an IL1RAP-blocking antibody limited plaque development and inflammation .
Optimal handling of recombinant IL1RAP requires attention to several factors:
Storage Conditions:
Reconstitution Protocol:
Carrier Protein Considerations:
Quality Control Testing:
Functional testing using binding assays to confirm activity before use in experiments.
Assessment of aggregation state by SEC or dynamic light scattering.
Experimental Design Factors:
Development of effective IL1RAP-targeting antibodies requires careful consideration of several factors:
Epitope Selection:
Antibody Generation Methods:
Engineering Considerations:
Functional Characterization:
Confirmation of specific binding to IL1RAP.
Assessment of blocking capacity for IL-1, IL-33, and IL-36 signaling.
Evaluation of downstream effects on cellular phenotypes.
Production and Quality Control:
Successful antibodies have demonstrated efficacy through multiple mechanisms, including signaling blockade and immune-mediated elimination of target cells .
Several cutting-edge research directions are expanding our understanding of IL1RAP:
Novel Signaling Partnerships:
Tissue-Specific Functions:
Biomarker Development:
Combination Therapies:
Exploration of synergistic effects between IL1RAP targeting and other therapeutic modalities (e.g., tyrosine kinase inhibitors, immune checkpoint inhibitors).
Development of dual-targeting approaches to address multiple pathways simultaneously.
Structural Biology:
Detailed structural characterization of IL1RAP interactions with various binding partners to facilitate structure-based drug design.
Investigation of conformational changes induced by ligand binding and receptor complex formation.
These emerging areas represent promising directions for researchers seeking to expand the frontiers of IL1RAP biology and therapeutic applications.
Integration of multiple omics approaches provides comprehensive insights into IL1RAP biology:
Transcriptomics-Proteomics Integration:
Spatial Transcriptomics:
Phosphoproteomics:
Characterization of phosphorylation cascades downstream of IL1RAP activation to map signaling networks.
Identification of novel phosphorylation targets affected by IL1RAP modulation.
Single-Cell Analysis:
Systems Biology Approaches:
Network analysis to identify hub genes and pathways connected to IL1RAP function.
Mathematical modeling of IL1RAP-mediated signaling dynamics across different cellular contexts.