IL1R1 (Interleukin-1 Receptor Type 1), also known as CD121a, is a membrane-bound receptor critical for mediating immune and inflammatory responses. It binds to IL-1α, IL-1β, and IL-1RA (antagonist), forming a high-affinity complex with the co-receptor IL1RAP to activate downstream signaling pathways like NF-κB and MAPK . The human gene IL1R1 is located on chromosome 2q12 within a cytokine receptor cluster .
IL1R1 is a glycosylated polypeptide containing 324 amino acids (residues 21–336) with a molecular weight of 37.4 kDa. Recombinant IL1R1 is produced in Sf9 baculovirus cells or HEK293 systems, often fused with an 8-amino acid His-tag for purification .
IL1R1 mediates pro-inflammatory responses by:
Binding IL-1α/β: Activates NF-κB, MAPK, and JAK-STAT pathways via recruitment of TOLLIP, MYD88, and IRAK1/2 .
Antagonism by IL-1RA: Blocks IL1RAP interaction, preventing signaling .
Key Interactions (from PubMed and Nature studies) :
Partner | Role |
---|---|
IL1RAP | Co-receptor for high-affinity signaling |
PIK3R1 | Phosphoinositide 3-kinase regulation |
MYD88 | Adapter molecule for TLR/IL-1R signaling |
IL1R1 is expressed in immune and non-immune tissues, with notable presence in:
Tissue | Expression Level | Source |
---|---|---|
Bone marrow | High | Human Protein Atlas |
Liver | Moderate | Human Protein Atlas |
Lung | Moderate | Human Protein Atlas |
Skin | Low | Human Protein Atlas |
Data derived from RNA-seq and IHC analyses .
Type 1 Diabetes: Polymorphisms in the IL1R1 5′UTR correlate with disease susceptibility and altered plasma IL1R1 levels .
Gastric Cancer (GC): High IL1R1 expression predicts poor prognosis and resistance to chemotherapy/immunotherapy, fostering immunosuppressive microenvironments .
Acute Myeloid Leukemia (AML): Reduced IL1RN expression in CD34+ progenitors correlates with IL-1β pathway activation, suggesting therapeutic potential for IL1RN or anti-IL-1β agents .
IL1R1:Fc Fusion Proteins: Recombinant IL1R1 fused to Fc domains (e.g., AdipoGen’s AG-40B-0024) neutralizes IL-1β, blocking inflammatory signaling .
IL-1RA Augmentation: In AML, exogenous IL-1RN suppresses leukemic cell expansion in xenograft models .
Human vs. Mouse Models: Human IL-1β dominates pro-inflammatory responses, while mice show higher IL-1RA production, limiting systemic inflammation .
IL1R1 Deficiency: Mice lacking IL1R1 exhibit reduced cytokine responses (e.g., IL-6, TNF) to stimuli like RNA vaccines or LPS, highlighting its role in amplifying inflammation .
IL1R1 is a key component of the interleukin-1 signaling pathway that belongs to the immunoglobulin superfamily. The gene encoding IL1R1 in humans is located on chromosome 2 and contains multiple exons that can undergo alternative splicing. Expression patterns vary significantly across tissues, with notable expression in immune cells, fibroblasts, and epithelial cells.
In research contexts, IL1R1 expression is typically assessed using RT-PCR, RNA sequencing, or protein-level detection through Western blotting or flow cytometry. Recent single-cell RNA sequencing approaches have revealed that IL1R1 expression can be heterogeneous even within seemingly uniform cell populations, particularly in CD4+ T cells responding to viral antigens . When studying IL1R1 expression, researchers should consider both tissue-specific and cell-type-specific patterns to accurately interpret experimental results.
Several clinically significant SNPs have been identified in the IL1R1 gene that can affect study outcomes and interpretation. When designing genetic association studies involving IL1R1, researchers should consider:
Haplotype analysis rather than isolated SNP analysis, as demonstrated in the Leiden Thrombophilia Study where 18 SNPs were used to tag 25 haplotype groups
Population-specific variation in SNP frequencies
Functional consequences of specific variants
For example, three SNPs of IL1R1 (rs871659, rs3771202, and rs3917238) have been associated with primary knee osteoarthritis, with sex-specific effects observed for the rs871659 allele A showing higher prevalence in females .
When conducting genetic studies involving IL1R1, sample size calculation should account for expected effect sizes. One study calculated that with a power of 80% (β = 0.2 and α = 0.05), appropriate sample sizes were 100 patients for the control group and 130 for the case group .
Multiple techniques can be employed to quantify IL1R1 protein levels in human samples, each with methodological considerations:
Technique | Sample Type | Sensitivity | Advantages | Limitations |
---|---|---|---|---|
ELISA | Serum, plasma, tissue lysates | 10-15 pg/ml | Quantitative, high-throughput | Cannot distinguish cell-specific expression |
Flow cytometry | Cells, blood | Cell-level detection | Single-cell resolution, multiparameter | Requires fresh or properly preserved samples |
Mass cytometry | Cells, tissues | High sensitivity | Multiparameter, no spectral overlap | Expensive, complex data analysis |
Immunohistochemistry | Tissue sections | Moderate | Spatial context preserved | Semi-quantitative |
Western blot | Tissue/cell lysates | Moderate | Size verification | Not high-throughput |
Researchers should be aware that plasma concentrations of IL1R1 protein have been shown to correlate with specific genotypes, as demonstrated in studies examining associations between gene variants such as rs871659 (A/A), rs3771202 (G/G), and rs3917238 (T/T) and plasma IL1R1 levels .
Recent high-dimensional single-cell analyses have revealed that viral antigen-specific CD4+ T cells, including those specific for SARS-CoV-2 spike protein, express elevated levels of IL1R1 in humans . This expression pattern has significant functional consequences for immune responses.
Methodologically, researchers investigating this phenomenon should consider:
Using multiplexed approaches (single-cell RNA-seq, mass cytometry, and flow cytometry) to characterize IL1R1+ T cell populations
Correlating IL1R1 expression with functional markers of T cell activation and cytokine production
Conducting ex vivo IL1R1 triggering or blocking experiments to assess functional outcomes
In vivo studies using COVID-19 mRNA vaccine mouse models demonstrated that neutralizing IL1R1 decreased IFN-γ expression by spike protein-specific CD4+ T cells and reduced the development of anti-spike protein IgG antibodies . This suggests a critical role for IL1R1 in coordinating T cell-dependent humoral immunity.
When designing experiments to investigate IL1R1 in T cell responses, researchers should include appropriate controls for both IL1R1 stimulation (using recombinant IL-1β) and blocking (using IL-1 receptor antagonists like Anakinra), with careful attention to dosing and timing relative to antigenic stimulation.
IL1R1+ cancer-associated fibroblasts (CAFs) have emerged as key modulators of the tumor microenvironment with significant implications for cancer progression. Single-cell sequencing of colorectal cancer patient samples has identified a pro-tumorigenic IL1R1+, IL-1-high-signaling subtype of fibroblasts that is associated with T cell and macrophage suppression .
For researchers studying IL1R1+ CAFs, methodological considerations include:
Isolation and characterization of CAF subtypes using single-cell approaches
3D co-culture systems with cancer cells to assess functional impacts
In vivo models using fibroblast-specific IL1R1 knockout or pharmacological intervention with IL-1 receptor antagonists
An emerging area of IL1R1 research concerns its regulation by telomere length, particularly in cancer contexts. Recent studies have demonstrated that IL-1 signaling is telomere-length dependent in cancer cells, with a mechanism involving non-telomeric TRF2 (telomeric repeat binding factor 2) binding at the IL1R1 promoter .
For researchers investigating telomere-IL1R1 interactions, important methodological approaches include:
Chromatin immunoprecipitation (ChIP) to assess TRF2 binding and histone modifications at the IL1R1 promoter
Comparative studies between cells with artificially manipulated telomere lengths
Assessment of downstream effectors, including NF-κB signaling and cytokine production
Specifically, enhanced TRF2 binding at the IL1R1 promoter in cells with short telomeres directly recruits the histone-acetyl-transferase p300, leading to H3K27 acetylation and IL1R1 activation . This mechanism alters NF-κB signaling and affects downstream cytokines including IL6, IL8, and TNF.
In triple-negative breast cancer (TNBC) clinical samples, IL1R1 expression correlates with telomere length, and infiltration of tumor-associated macrophages shows sensitivity to tumor cell telomere length . This relationship provides a novel framework for understanding immune microenvironment dynamics in cancers with varying telomere biology.
When investigating IL1R1 genetic variants in disease association studies, researchers should implement several key methodological strategies:
Haplotype-based analysis rather than isolated SNP studies
Appropriate control selection and matching for demographic variables
Adequate statistical power through proper sample size calculation
Assessment of gene-environment interactions
The Leiden Thrombophilia Study provided a robust methodological framework by genotyping 18 SNPs to tag 25 haplotype groups, enabling the identification of haplotype 5 of IL1RN (tagged by SNP 13888T/G, rs2232354) as a risk factor for venous thrombosis when present in homozygous form (Odds ratio=3.9; 95% confidence interval: 1.6 to 9.7; P=0.002) .
For analyzing genetic association data, specialized statistical tools like Haplo.stats can be employed to assess haplotype effects without requiring definitive haplotype assignment to individuals . This approach is particularly valuable when multiple linked polymorphisms are present within the gene region.
When designing interventional studies targeting IL1R1 signaling, researchers should consider:
Selection of appropriate IL1R1 antagonists (small molecules vs. biologics)
Dosing strategies based on pharmacokinetic/pharmacodynamic models
Assessment of on-target effects through biomarker evaluation
Potential compensatory mechanisms through related pathways
In experimental settings, the IL-1 receptor antagonist Anakinra has been successfully employed to block IL1R1 signaling, with studies demonstrating efficacy in reducing tumor growth in vivo and restricting M2 macrophage infiltration in TNBC tumor organoids .
Researchers face several challenges when interpreting IL1R1 expression data across disease contexts:
Cell type-specific expression patterns that may be obscured in bulk tissue analyses
Context-dependent regulation by factors including telomere length , inflammatory stimuli, and genetic variants
Post-transcriptional regulation affecting protein levels
Heterogeneity in expression even within defined cell populations
To address these challenges, researchers should employ multiple complementary techniques, including:
Single-cell approaches to resolve cell type-specific expression
Correlation of expression with functional outcomes
Integration of genetic, epigenetic, and expression data
Longitudinal sampling where feasible to capture dynamic changes
For example, in TNBC studies, IL1R1 expression varies with telomere length and correlates with tumor-associated macrophage infiltration, highlighting the importance of considering both intrinsic cellular factors and microenvironmental interactions .
IL1R1 genotyping can provide valuable information for clinical risk assessment across several disease contexts:
Venous thrombosis risk assessment: Homozygous carriers of haplotype 5 of IL1RN (tagged by SNP 13888T/G, rs2232354) have an increased thrombotic risk (OR=3.9)
Osteoarthritis susceptibility: IL1R1 SNPs rs871659, rs3771202, and rs3917238 associate with primary knee osteoarthritis, with sex-specific effects
Cancer prognosis: IL1R1 expression patterns in cancer-associated fibroblasts correlate with immune infiltration and patient survival in colorectal cancer
For clinical implementation, researchers should consider:
Development of clinically validated genotyping panels
Integration with established risk factors and biomarkers
Prospective validation in diverse populations
Cost-effectiveness and accessibility of testing approaches
As IL1R1 emerges as a potential therapeutic target, researchers developing immunotherapeutic approaches should consider:
Cell type-specific targeting strategies to avoid systemic inflammatory effects
Combination approaches with established immunotherapies
Biomarker development for patient stratification
Monitoring of immune cell phenotypes and function during treatment
Experimental evidence supports the potential efficacy of IL1R1 targeting in cancer contexts. In TNBC tumor organoids, both IL-1 Receptor antagonist (IL1RA) and IL1R1 targeting ligands demonstrated ability to abrogate M2 macrophage infiltration . The finding that infiltration of tumor-associated macrophages is sensitive to tumor cell telomere length and highly correlated with IL1R1 expression suggests that patient stratification based on these parameters might improve therapeutic outcomes .
For T cell-directed immunotherapies, the high expression of IL1R1 on viral antigen-specific CD4+ T cells provides a rationale for targeting this pathway to enhance vaccine efficacy or antiviral responses . Experimental approaches should include assessment of IL1R1 manipulation on both T cell effector function and memory formation.
Several cutting-edge technologies are advancing our understanding of IL1R1 signaling dynamics:
Live-cell imaging of IL1R1 trafficking and signaling using fluorescent protein fusions
CRISPR-based genetic screens to identify novel regulators of IL1R1 expression and function
Proteomics approaches to comprehensively map IL1R1 signaling complexes
Spatial transcriptomics to resolve IL1R1 expression patterns within complex tissues
These technologies enable researchers to move beyond static measurements to capture the temporal and spatial dynamics of IL1R1 signaling in physiologically relevant contexts.
Integration of multiple -omics approaches provides powerful insights into IL1R1 biology:
Omics Approach | Information Provided | Integration Value |
---|---|---|
Genomics | Genetic variants, regulatory elements | Identifies heritable risk factors |
Transcriptomics | Expression patterns, alternative splicing | Reveals regulatory mechanisms |
Epigenomics | Chromatin accessibility, histone modifications | Uncovers context-specific regulation |
Proteomics | Protein levels, post-translational modifications | Connects genotype to functional phenotype |
Metabolomics | Downstream metabolic effects | Links signaling to cellular physiology |
The power of multi-omics is exemplified in studies demonstrating that telomere length affects TRF2 binding and histone acetylation at the IL1R1 promoter, altering gene expression and downstream signaling cascades . This mechanistic insight required integration of genomic, epigenomic, and transcriptomic data.
Future research should leverage these integrated approaches to build comprehensive models of IL1R1 function across different physiological and pathological contexts, enabling more precise therapeutic targeting of this key inflammatory pathway.
Interleukin 1 Receptor Type I (IL-1RI) is a cytokine receptor that belongs to the interleukin-1 receptor family. It plays a crucial role in mediating immune and inflammatory responses by binding to interleukin 1 alpha (IL-1α), interleukin 1 beta (IL-1β), and interleukin 1 receptor antagonist (IL-1RA) . This receptor is expressed on various cell types, including T cells, B cells, monocytes, and fibroblasts .
IL-1RI is a transmembrane protein composed of a single polypeptide chain. It binds both IL-1α and IL-1β, acting as a potent antagonist of IL-1 action . The receptor complex mediates all known IL-1 biological responses, including the activation of immune cells and the promotion of inflammatory processes .
Recombinant human IL-1RI is produced using DNA sequences encoding the amino-terminal residues of the extracellular domain of the human IL-1 receptor Type I protein . This recombinant form is used in various research and therapeutic applications, particularly in studying the role of IL-1 in diseases such as rheumatoid arthritis .
IL-1RI is a key player in the pathogenesis of several inflammatory diseases. In rheumatoid arthritis, for example, IL-1 stimulates the production of prostaglandin E2, nitric oxide, and matrix metalloproteases, which promote joint degradation . Additionally, IL-1 suppresses joint repair by inhibiting collagen synthesis . The receptor’s involvement in these processes makes it a target for therapeutic interventions aimed at reducing inflammation and tissue damage.
The recombinant form of IL-1RI has been developed and tested in animal models for arthritis . It binds to IL-1R type I with the same affinity as IL-1β, making it a valuable tool for studying the receptor’s role in disease and for developing new treatments . One such treatment is anakinra (Kineret™), a recombinant human IL-1 receptor antagonist used to treat rheumatoid arthritis .