IL1R1 Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

The IL1R1 recombinant monoclonal antibody is generated through a multi-step process involving the acquisition of IL1R1 antibody genes, their introduction into suitable host cells, and the synthesis of IL1R1 antibodies using a cell-based expression and translation system. This approach not only enhances the purity and stability of the resulting IL1R1 recombinant monoclonal antibodies but also improves their affinity and specificity. The IL1R1 recombinant monoclonal antibody undergoes purification via affinity chromatography and is subjected to testing in both ELISA and FC assays. It is important to note that this antibody selectively recognizes the human IL1R1 protein.

IL1R1 is a receptor protein that plays a crucial role in mediating inflammatory responses and immune system activation. Its activation by IL-1 cytokines initiates a cascade of events that contribute to the body's defense against infections and the regulation of inflammation. Dysregulation of the IL-1 pathway can lead to chronic inflammatory diseases and autoimmunity.

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Description

The IL1R1 Recombinant Monoclonal Antibody is produced through a multi-step process that involves obtaining the IL1R1 antibody genes, introducing them into suitable host cells, and subsequently synthesizing the IL1R1 antibodies using a cell-based expression and translation system. This approach not only enhances the purity and stability of the resulting IL1R1 recombinant monoclonal antibodies but also improves their affinity and specificity. The IL1R1 Recombinant Monoclonal Antibody undergoes purification via affinity chromatography and is subjected to rigorous testing using both ELISA and FC assays. It is crucial to note that this antibody selectively recognizes the human IL1R1 protein.

IL1R1 is a receptor protein that plays a crucial role in mediating inflammatory responses and immune system activation. Its activation by IL-1 cytokines initiates a cascade of events that contribute to the body's defense against infections and the regulation of inflammation. Dysregulation of the IL-1 pathway can lead to the development of chronic inflammatory diseases and autoimmune disorders.

Form
Liquid
Lead Time
Typically, we can ship the products within 1-3 business days after receiving your order. Delivery times may vary depending on the method of purchase or location. Please consult your local distributors for specific delivery timeframes.
Synonyms
Antigen CD121a antibody; CD121 antigen-like family member A antibody; CD121a antibody; CD121a antigen antibody; D2S1473 antibody; IL 1 antibody; IL 1R 1 antibody; IL 1R alpha antibody; IL 1RT1 antibody; IL-1R-1 antibody; IL-1R-alpha antibody; IL-1RT-1 antibody; IL-1RT1 antibody; IL1 inhibitor antibody; IL1 Receptor I antibody; IL1R antibody; IL1R1 antibody; IL1R1_HUMAN antibody; IL1RA antibody; IL1RN antibody; IL1RT1 antibody; Interleukin 1 receptor alpha type 1 antibody; Interleukin 1 receptor antagonist protein (IRAP) antibody; Interleukin 1 receptor type I antibody; Interleukin receptor 1 antibody; Interleukin-1 receptor alpha antibody; Interleukin-1 receptor type 1 antibody; Interleukin-1 receptor type I antibody; p80 antibody
Target Names
Uniprot No.

Target Background

Function
IL1R1 acts as a receptor for IL1A, IL1B, and IL1RN. Upon binding to interleukin-1, it associates with the coreceptor IL1RAP to form the high-affinity interleukin-1 receptor complex. This complex mediates interleukin-1-dependent activation of NF-kappa-B, MAPK, and other signaling pathways. The signaling process involves the recruitment of adapter molecules, such as TOLLIP, MYD88, and IRAK1 or IRAK2, via the respective TIR domains of the receptor/coreceptor subunits. IL1R1 binds ligands with comparable affinity, and the binding of the antagonist IL1RN prevents association with IL1RAP, thus inhibiting the formation of a signaling complex. IL1R1 plays a role in IL1B-mediated costimulation of IFNG production from T-helper 1 (Th1) cells.
Gene References Into Functions
  1. Interleukin-1 receptor 1/MyD88 signaling has roles in the development and progression of pulmonary hypertension PMID: 27418552
  2. A study highlights the association of genetic polymorphisms of IL1R1 with knee osteoarthritis susceptibility in the Northwestern Chinese Han population. PMID: 27980229
  3. Serum ST2 levels are elevated in heart failure patients and correlate with disease severity. PMID: 28164491
  4. Since mumps virus SH coimmunoprecipitated with tumor necrosis factor receptor 1 (TNFR1), RIP1, and IRAK1, it is hypothesized that SH exerts its NF-kappaB activation inhibitory function by interacting with TNFR1, interleukin-1 receptor type 1 (IL-1R1), and TLR3 complexes in the plasma membrane of infected cells. PMID: 28659487
  5. Salt-inducible kinase inhibition decreases proinflammatory cytokines in human myeloid cells upon IL-1R stimulation. PMID: 26590148
  6. A study confirmed that genetic variants in IL-1R1 (rs3917267) have a significant association with HBV infection and HBV breakthrough infection in children, providing new insights into the pathogenesis of chronic HBV infection in children. PMID: 28027994
  7. The study identified critical regions within the TIR domain of IL-1 receptor type in both humans and mice. PMID: 26279140
  8. The IL1R1 SNP rs949963 is associated with the susceptibility to asthma in children from Central China and may increase the serum expression of IL1R1. PMID: 26975823
  9. A study investigated the association between four inflammatory cytokines (CD121a, interleukin [IL]-1beta, IL-8, and IL-11) and coronary heart disease. PMID: 26098632
  10. This study found that the mRNA expression of IL-1R1, TNFR1, and TNFR2 was significantly higher in schizophrenia. PMID: 25749018
  11. IL-1R1 expression appears to define a tissue regulatory T cell phenotype along with the expression of CD25, glucocorticoid-induced tumor necrosis factor receptor family-related gene, and CTLA-4. PMID: 26076982
  12. TNF-alpha (-308) GG, IL-10 (-819) TT, IL-10 (-1082) GG, and IL1R (+1970) CC genotypes are found to be predominant (p=0.01, p=0.02, p=0.0001, and p=0.001, respectively) in both tuberculoid and lepromatous leprosy patients. PMID: 25697140
  13. IL-1 receptor gene polymorphisms could be one of the factors influencing the expression of membrane-bound IL-1 receptors on immunocompetent cells. PMID: 24976267
  14. Severe forms of pancreatitis are associated with increased levels of IL-6, IL-10, and IL-1ra. PMID: 25240697
  15. SUMOylation is a novel mechanism in the regulation of beta-arrestin 2-mediated IL-1R/TRAF6 signaling. PMID: 25425640
  16. Genetic polymorphism is associated with atopic dermatitis in the Iranian pediatric population. PMID: 23253688
  17. Blood monocytes from renal cell carcinoma patients displayed a tumor-promoting transcriptional profile that supported functions like angiogenesis and invasion. Induction of this protumor phenotype required an interleukin-1 receptor (IL-1R)-dependent mechanism. PMID: 25453823
  18. The NLRP3 inflammasome/IL-1RI axis is dispensable for PM10-facilitated allergic sensitization. PMID: 24988285
  19. Thirty-eight single nucleotide polymorphisms (SNPs) were identified within genomic regions containing all exons and relevant exon-intron boundaries in IL1R1 and IL1R2 in Japanese aggressive periodontitis. PMID: 24818754
  20. Only the IL1RN tandem repeats polymorphism may be associated with Hashimoto's thyroiditis susceptibility; TSHR and IL1RN polymorphisms may represent prognostic factors for predicting the severity of the disease. PMID: 24328419
  21. Findings provide evidence of subgroups of women with breast cancer who report distinct trajectories of attentional function and of a genetic association between subgroup membership and an IL1R1 promoter polymorphism. PMID: 24315345
  22. No significant differences are detected in the Fcgamma-RIIa and IL1-Ra polymorphisms compared to controls in a study to search for associations between antineutrophil cytoplasm antibody-associated vasculitis and IL1R1 polymorphisms. PMID: 24356554
  23. Results demonstrate clear functional consequences of the rs917997 risk polymorphism; this polymorphism leads to a loss-of-function through decreased IL-18RAP, IL-18R1, and IL-1R1 protein expression, which impairs autocrine IL-18 and IL-1 signaling. PMID: 24842757
  24. The frequency of the IL-1RN rs315952 CT genotype was significantly lower among patients with systemic lupus erythematosus compared with healthy controls. PMID: 23722873
  25. Common IL-33 and IL1RL1 polymorphisms contribute to the risk of inflammatory bowel disease. PMID: 23634226
  26. The IL1R1 promoter SNP rs2192752 may contribute to the location of papillary thyroid carcinoma (PTC), and the C allele of rs2192752 may be a risk factor for the development of PTC in both lobes. PMID: 22594576
  27. These results demonstrate that IL-1alpha and IL-1R1 signaling is essential for microabscess formation, neutrophil recruiting chemokine expression, and acanthosis in psoriasis-like skin inflammation induced by imiquimod. PMID: 23407395
  28. The internalization of IL1RI after IL-1 binding is not a mechanism of feedback inhibition but of critical importance for the IL-1-induced gene expression. PMID: 23022958
  29. Analysis of the data suggested that IL-1R1 expression in the neonatal period provides an additional level of Myd88-dependent signaling during this period of heightened susceptibility to infection. PMID: 22435759
  30. The IL-1R1 1498T>C polymorphism is associated with early-stage endometriosis in Korean women. PMID: 22509941
  31. A genetic link between rs1921622 IL1RL1 polymorphism and disease severity in RSV bronchiolitis. PMID: 22574108
  32. This study demonstrated that carriers of the minor allele for a single nucleotide polymorphism in interleukin (IL)1-receptor 1 (IL1R1) (rs2110726) were less likely to report breast pain prior to surgery. PMID: 22515947
  33. The LCAP-related changes in sIL-1RI and IL-1Ra might impact the clinical outcome during treatment with an LCAP filter in patients with inflammatory bowel disease. PMID: 22267087
  34. No sufficient evidence was available to support any associations between IL1-Ra and IL6-147G/C polymorphisms and ischemic stroke. PMID: 22417897
  35. A role for TILRR in selective amplification of NF-kappaB responses through IL-1RI and suggest that the specificity is determined by changes in receptor conformation and adapter protein recruitment. PMID: 22262840
  36. The study presents the crystal structure of IL-1beta bound to its primary receptor IL-1RI and its receptor accessory protein IL-1RAcP. PMID: 22426547
  37. Data demonstrated that IL-1R polymorphism is associated with susceptibility to ankylosing spondylitis. PMID: 22285486
  38. IL1R1 regulates S100A8/A9-dependent keratinocyte resistance to bacterial invasion. PMID: 22031183
  39. Findings demonstrate that neutrophils express IL-1R1 and that IL-1R1 is induced following extravasation and exists in a mobile intracellular compartment. PMID: 22385245
  40. Data show that variants in SFTPD, CD46, and IL1R1 are associated with IPD in both EA and AA. PMID: 21858107
  41. IL-1RI expression in regulatory T (Treg) cells identifies a subpopulation at an early stage of differentiation. PMID: 21998454
  42. This study found for the first time an epigallocatechin gallate-induced downregulation of the IL-1RI expression, possibly caused by NF-kappaB inhibition. PMID: 21787753
  43. IL-1RI expression was not altered in endometrioid carcinoma cells in comparison with endometrial cells, clear-cells, serous, and mucinous ovarian cancer cells. PMID: 21083841
  44. Interleukin-1 receptor-mediated inflammation impairs the heat shock response of human mesothelial cells. PMID: 21435443
  45. No association was found between recurrent spontaneous abortion and gene polymorphisms in the interleukin-1 receptor 1 gene. PMID: 20956022
  46. IL1RL1 polymorphisms are associated with serum IL1RL1-a, blood eosinophils, and asthma in childhood. PMID: 21281963
  47. Serum levels of ECP, IL-5, -6, -8, and -10, G-CSF, MCP-1, IL-1 ra, and IP-10 were significantly elevated in acute compared with stable childhood asthma. PMID: 20523065
  48. This study indicated that statistically significant higher protein and mRNA levels of the il1r in the frontal cortex in bipolar disorder. PMID: 19488045
  49. Findings that human Tregs preferentially express receptors for TNF and IL-1 suggest a potential function in sensing and dampening local inflammation. PMID: 20066156
  50. An association between severe hand osteoarthritis and the IL1R1 gene. PMID: 20353565

Show More

Hide All

Database Links

HGNC: 5993

OMIM: 147810

KEGG: hsa:3554

STRING: 9606.ENSP00000233946

UniGene: Hs.701982

Protein Families
Interleukin-1 receptor family
Subcellular Location
Membrane; Single-pass type I membrane protein. Cell membrane. Secreted.
Tissue Specificity
Expressed in T-helper cell subsets. Preferentially expressed in T-helper 1 (Th1) cells.

Q&A

Fundamental Understanding of IL1R1 and Its Antibodies

The interleukin-1 receptor type 1 (IL1R1) represents a critical component in inflammatory signaling pathways, functioning as the primary receptor for IL-1 family cytokines. Before discussing the antibodies against this target, researchers must thoroughly understand the biological context and significance of IL1R1 in immune regulation and disease pathogenesis.

IL1R1 is a membrane-bound protein that can also exist in a soluble form after cleavage by matrix metalloproteases. Both forms are biologically active and play roles in regulating inflammatory responses through interactions with cytokines. The protein has a molecular mass of approximately 65.4 kilodaltons and is encoded by the IL1R1 gene in humans . This receptor is positioned at the "epicenter" of inflammatory signaling networks, making it a critical target for immunological research .

What is IL1R1 and what are its primary biological functions?

IL1R1 (interleukin 1 receptor type 1) functions as the primary receptor for IL1A, IL1B, and IL1RN (IL-1 receptor antagonist). After binding to interleukin-1, it associates with the coreceptor IL1RAP to form a high-affinity receptor complex that mediates interleukin-1-dependent activation of NF-kappa-B, MAPK, and other signaling pathways . This signaling cascade involves the recruitment of adapter molecules such as TOLLIP, MYD88, and IRAK1 or IRAK2 via their respective TIR domains . IL1R1 is critically involved in innate immunity, inflammatory responses, and has been implicated in numerous disease states including neurodegenerative conditions like Alzheimer's disease, Parkinson's disease, and multiple sclerosis .

How do IL1R1 expression patterns differ across tissue types and disease states?

IL1R1 expression exhibits tissue-specific patterns that can change dramatically during disease progression. In the central nervous system (CNS), small amounts of mRNA have been found in glial cells under basal conditions, though there is some controversy regarding its expression in microglia . Under pathological conditions, IL1R1 may be induced by IL-1β itself, elevating the mRNA levels of this receptor on glial cells . The regulation of IL1R1 expression appears tightly linked to the activated status of human glial cells, with significant upregulation observed in different models of neuroinflammation .

Technical Considerations for IL1R1 Recombinant Monoclonal Antibodies

Recombinant monoclonal antibodies against IL1R1 offer significant advantages over traditional monoclonal antibodies, including improved batch-to-batch consistency, reduced background signal, and enhanced specificity. These technical characteristics make them powerful tools for investigating IL1R1 biology in research contexts.

When selecting an IL1R1 recombinant monoclonal antibody, researchers should consider the host species, clonality, validated applications, and cross-reactivity profile. For instance, rabbit-derived recombinant monoclonal antibodies like EPR22198-36 have been validated for Western blot applications with mouse samples . The technical specifications of these antibodies directly impact experimental design and interpretation of results.

How do recombinant monoclonal antibodies against IL1R1 differ from traditional monoclonals?

Recombinant monoclonal antibodies against IL1R1 are produced through recombinant DNA technology, where antibody genes are cloned and expressed in cell culture systems. This differs from traditional hybridoma-produced monoclonal antibodies and offers several advantages. Recombinant antibodies demonstrate superior batch-to-batch consistency due to their defined genetic sequence, eliminating the variability inherent in hybridoma cultures . Additionally, they can be engineered for enhanced specificity, affinity, and reduced immunogenicity. For IL1R1 research, recombinant antibodies like EPR22198-36 provide more consistent detection across experimental replicates, which is particularly important when studying subtle changes in IL1R1 expression during disease progression or therapeutic interventions.

What experimental validation should researchers expect for high-quality IL1R1 recombinant antibodies?

High-quality IL1R1 recombinant monoclonal antibodies should undergo rigorous validation across multiple applications and experimental systems. At minimum, researchers should expect Western blot validation demonstrating specific detection of IL1R1 at the expected molecular weight (approximately 65.4 kDa) . For immunohistochemistry applications, antibodies should show specific staining patterns consistent with known IL1R1 expression. Cross-reactivity testing across species (human, mouse, rat) is essential for comparative studies. Additionally, knockout/knockdown validation provides the gold standard for specificity, where the antibody signal should be absent or significantly reduced in samples where IL1R1 expression has been eliminated. Publications citing the antibody provide further evidence of reliability in real research contexts.

How should researchers optimize Western blot protocols when using IL1R1 recombinant antibodies?

Western blot optimization for IL1R1 detection requires careful consideration of several parameters. Sample preparation is critical—researchers should use appropriate lysis buffers containing protease inhibitors to prevent IL1R1 degradation. Given that IL1R1 can exist in both membrane-bound (full-length) and soluble (cleaved) forms, membrane protein extraction protocols may be necessary to capture the complete expression profile . For gel electrophoresis, gradient gels (4-12%) often provide better resolution for the 65.4 kDa IL1R1 protein. During transfer, standard PVDF membranes are typically suitable, but transfer time and voltage may need optimization for complete transfer of this mid-sized protein. Blocking with 5% BSA rather than milk is often recommended for phospho-specific detection. Primary antibody concentration should be titrated (typically starting at 1:1000 dilution for recombinant antibodies), and overnight incubation at 4°C may improve signal-to-noise ratio compared to shorter incubations at room temperature.

Applications of IL1R1 Recombinant Monoclonal Antibodies in Research

IL1R1 recombinant monoclonal antibodies serve as versatile tools across a spectrum of research applications, enabling detailed investigation of IL1R1 biology in both normal and pathological contexts. These applications range from standard protein detection methods to complex functional studies examining the role of IL1R1 in disease mechanisms.

The search results indicate that available IL1R1 antibodies have been validated for numerous applications including Western blotting (WB), enzyme-linked immunosorbent assay (ELISA), immunocytochemistry (ICC), immunohistochemistry (IHC), flow cytometry (FCM), and immunoprecipitation (IP) . This versatility allows researchers to select the most appropriate methodology based on their specific research questions and experimental systems.

How can IL1R1 recombinant antibodies be effectively used in neuroinflammation research?

In neuroinflammation research, IL1R1 recombinant monoclonal antibodies serve as critical tools for investigating the role of IL-1 signaling in CNS pathology. These antibodies can be used to track IL1R1 expression changes in different neural cell populations (neurons, astrocytes, microglia) under inflammatory conditions using immunofluorescence or flow cytometry . For tissue analysis, immunohistochemistry using validated IL1R1 antibodies can reveal spatial distribution patterns of receptor expression in brain regions affected by neuroinflammatory conditions. Western blotting can quantify changes in total IL1R1 protein levels as well as the ratio between membrane-bound and soluble forms, which may shift during disease progression . Importantly, these antibodies can help resolve controversies regarding cell-specific expression, such as the debated presence of IL1R1 on microglia in different activation states . When combined with functional readouts like cytokine production or NF-κB activation, antibody-based detection of IL1R1 helps establish mechanistic links between receptor expression and inflammatory outcomes in neural tissues.

What considerations are important when using IL1R1 antibodies for flow cytometry?

When using IL1R1 recombinant monoclonal antibodies for flow cytometry, several technical considerations are critical for obtaining reliable results. First, antibody format is crucial—directly conjugated antibodies (PE, FITC, APC) eliminate the need for secondary detection steps, reducing background and simplifying protocols . Titration experiments should determine optimal antibody concentration, as both insufficient and excessive antibody can compromise sensitivity. For detecting IL1R1 on cell surfaces, gentle cell dissociation methods are essential to preserve membrane integrity and receptor epitopes. Appropriate controls must include isotype-matched antibodies to establish background levels and blocking experiments to confirm specificity. For intracellular detection of IL1R1, permeabilization protocols require careful optimization to maintain antibody accessibility while preserving epitope structure. When analyzing subpopulations, a comprehensive panel of lineage markers should accompany IL1R1 staining to identify specific cell types expressing the receptor. Finally, gating strategies should account for potential autofluorescence, particularly in cells like activated macrophages or microglia that may exhibit increased background signal during inflammatory responses.

How can researchers use IL1R1 recombinant antibodies to investigate receptor-ligand interactions?

Investigating IL1R1 receptor-ligand interactions requires sophisticated experimental approaches where high-quality recombinant antibodies play a central role. Co-immunoprecipitation (Co-IP) using IL1R1 antibodies can capture receptor complexes with IL1RAP and other signaling components following ligand stimulation . For these experiments, antibodies must recognize native conformations of IL1R1 and avoid binding sites that might interfere with ligand interaction. Proximity ligation assays (PLA) using IL1R1 antibodies paired with antibodies against putative binding partners can visualize protein-protein interactions in situ with subcellular resolution. Surface plasmon resonance (SPR) or bio-layer interferometry (BLI) studies can employ immobilized IL1R1 antibodies to capture receptors for quantitative binding studies with IL1A, IL1B, or IL1RA. Competitive binding assays can determine whether antibodies interfere with specific ligand binding, potentially identifying antibodies with receptor-modulating functions. For functional studies, researchers can use antibodies to track receptor internalization and trafficking following ligand binding using confocal microscopy or intracellular flow cytometry, providing insights into signaling dynamics.

Troubleshooting Common Issues with IL1R1 Antibodies

Despite their utility, working with IL1R1 recombinant monoclonal antibodies can present technical challenges that require systematic troubleshooting approaches. Understanding common issues and their solutions ensures more reliable and reproducible experimental outcomes.

The complexity of IL1R1 biology—including multiple isoforms, post-translational modifications, and conformational states—can complicate antibody-based detection. Additionally, the potential presence of soluble receptor forms can influence assay performance and interpretation . Recognizing these challenges is essential for developing robust experimental protocols.

What are the most common causes of false negative results when detecting IL1R1?

False negative results when detecting IL1R1 can stem from several sources. Inadequate sample preparation is a primary concern—IL1R1 is a membrane protein that may require specialized extraction methods to fully solubilize and maintain in its native conformation . Standard RIPA buffers may be insufficient, and gentler detergents like NP-40 or digitonin might better preserve epitope integrity. Another common issue involves epitope masking due to post-translational modifications, protein-protein interactions, or conformational changes in IL1R1. This is particularly relevant given that IL1R1 undergoes significant conformational changes upon ligand binding . Additionally, expression levels in some cell types or basal conditions may fall below detection thresholds, necessitating signal amplification methods or more sensitive detection systems. Finally, some antibody clones may recognize species-specific epitopes, leading to false negatives when used across species. For example, an antibody validated for human IL1R1 may fail to detect mouse IL1R1 despite sequence homology if the epitope region contains species-specific variations .

How can researchers distinguish between membrane-bound and soluble forms of IL1R1?

Distinguishing between membrane-bound (full-length) and soluble (cleaved) forms of IL1R1 requires strategic experimental design. Western blotting represents the most straightforward approach, as the two forms differ in molecular weight—membrane-bound IL1R1 appears at approximately 65.4 kDa, while the soluble form typically appears at a lower molecular weight due to cleavage of the transmembrane and cytoplasmic domains . Using antibodies targeting different epitopes can enhance discrimination; antibodies recognizing extracellular domains will detect both forms, while those targeting cytoplasmic regions will detect only the membrane-bound form. For in situ analysis, immunofluorescence with confocal microscopy can differentiate membrane-localized signal (appearing as cell surface staining) from diffuse extracellular signal (potentially representing soluble forms). Flow cytometry comparing permeabilized versus non-permeabilized cells can also help distinguish surface-bound from intracellular receptor pools. Finally, selective isolation techniques like cell surface biotinylation followed by streptavidin pull-down can physically separate membrane-bound receptors for subsequent detection with IL1R1 antibodies.

How should researchers address cross-reactivity concerns with IL1R1 antibodies?

Addressing cross-reactivity concerns with IL1R1 antibodies requires rigorous validation strategies. The first step involves comprehensive bioinformatic analysis to identify proteins with sequence homology to IL1R1, particularly other IL-1 receptor family members like IL1R2 that share structural features . Experimental validation should include parallel testing on samples with differential expression of IL1R1 and potential cross-reactive proteins. Knockout/knockdown controls provide the gold standard—testing the antibody on IL1R1-deficient samples should yield no signal if the antibody is truly specific. Competition assays with recombinant IL1R1 protein can further confirm specificity; pre-incubation with the target protein should abolish specific binding. For applications like immunohistochemistry, where spatial distribution provides additional context, comparing staining patterns with orthogonal methods (like in situ hybridization) can help verify that observed signals correspond to expected IL1R1 expression patterns. Finally, epitope mapping can identify the specific region recognized by the antibody, allowing researchers to predict potential cross-reactivities based on sequence conservation across related proteins.

Advanced Research Applications and Emerging Techniques

As research technologies evolve, innovative applications of IL1R1 recombinant monoclonal antibodies continue to emerge, enabling deeper understanding of IL1R1 biology and its roles in health and disease. These advanced approaches leverage the specificity and versatility of recombinant antibodies to address complex research questions.

The development of sophisticated imaging techniques, multi-omics approaches, and therapeutic applications represents the frontier of IL1R1 research. In these contexts, the quality and characterization of IL1R1 recombinant monoclonal antibodies become even more critical for generating reliable and translatable findings.

How can IL1R1 recombinant antibodies be integrated into single-cell analysis workflows?

Integrating IL1R1 recombinant monoclonal antibodies into single-cell analysis workflows represents a powerful approach for understanding cellular heterogeneity in IL-1 signaling responses. For single-cell protein analysis, antibody-based methods like CITE-seq (Cellular Indexing of Transcriptomes and Epitopes by Sequencing) can incorporate IL1R1 antibodies conjugated to oligonucleotide barcodes, allowing simultaneous measurement of IL1R1 protein expression alongside transcriptomic profiling at single-cell resolution. Flow cytometry or mass cytometry (CyTOF) panels can include IL1R1 antibodies within multiparameter panels to correlate IL1R1 expression with cell lineage markers and activation states across thousands of individual cells. For spatial analysis, techniques like multiplexed ion beam imaging (MIBI) or co-detection by indexing (CODEX) can incorporate IL1R1 antibodies into highly multiplexed panels for visualizing receptor expression in tissue contexts with preservation of spatial relationships. These approaches are particularly valuable for heterogeneous tissues like the CNS, where IL1R1 expression may vary dramatically across different neural cell types and disease states . The key technical consideration for these applications is antibody specificity, as false positives at the single-cell level can lead to misinterpretation of cellular subpopulations.

What are the prospects for using IL1R1 antibodies in therapeutic development?

IL1R1 antibodies hold significant potential in therapeutic development, particularly for conditions involving dysregulated IL-1 signaling. The research literature indicates that persistent activation of IL-1/IL-1R1 signaling is linked to numerous CNS pathologies, including Alzheimer's disease, Parkinson's disease, and multiple sclerosis . Antagonistic antibodies that block IL-1 binding to IL1R1 could serve as therapeutic agents by inhibiting downstream inflammatory signaling. The development of such antibodies requires detailed epitope mapping to ensure they interfere with ligand binding sites. Unlike small molecule inhibitors, antibodies targeting IL1R1 can achieve high specificity, potentially reducing off-target effects. For therapeutic applications, antibody engineering approaches—including humanization, Fc modifications for altered half-life or effector functions, and bispecific formats—may enhance efficacy and pharmacokinetic properties. Importantly, the distinction between membrane-bound and soluble IL1R1 forms presents both a challenge and opportunity; antibodies could be designed to selectively target one form over the other, potentially allowing more precise modulation of IL-1 signaling . Preclinical research using IL1R1 antibodies in disease models can help establish proof-of-concept for therapeutic targeting of this pathway.

How can IL1R1 antibodies contribute to understanding the spatial organization of IL-1 signaling complexes?

Advanced imaging approaches using IL1R1 recombinant monoclonal antibodies can reveal the spatial organization of IL-1 signaling complexes with unprecedented detail. Super-resolution microscopy techniques like Structured Illumination Microscopy (SIM), Stimulated Emission Depletion (STED), or Single-Molecule Localization Microscopy (SMLM) overcome the diffraction limit of conventional microscopy, allowing visualization of nanoscale distribution patterns of IL1R1 on cell membranes. These approaches can reveal receptor clustering, colocalization with signaling partners like IL1RAP, and changes in spatial organization following ligand binding . Multi-color imaging using IL1R1 antibodies alongside antibodies against other signaling components can map the complete signaling hub architecture. Live-cell imaging with fluorescently labeled non-blocking IL1R1 antibody fragments can track receptor dynamics in real-time, providing insights into trafficking, internalization, and signal termination. For tissue-level analysis, techniques like expansion microscopy combined with IL1R1 immunostaining can maintain spatial context while achieving sub-diffraction resolution. These sophisticated spatial analyses are particularly valuable for understanding how IL1R1 signaling is organized at the cell membrane and how this organization may be disrupted in disease states, potentially revealing new therapeutic targets.

Comparative Analysis and Selection Guidelines

The diversity of commercially available IL1R1 recombinant monoclonal antibodies necessitates careful selection based on intended applications, experimental systems, and specific research questions. A systematic approach to antibody comparison and selection maximizes the likelihood of experimental success.

According to the search results, there are at least 718 IL1R1 antibodies available across 31 suppliers, with varying properties, applications, and validation levels . This abundance of options highlights the need for clear selection criteria based on experimental requirements and quality indicators.

What criteria should guide the selection of IL1R1 recombinant antibodies for specific applications?

Selection of IL1R1 recombinant monoclonal antibodies should follow a systematic evaluation process based on application-specific criteria. For Western blotting applications, epitope accessibility in denatured conditions, demonstrated specificity at the expected molecular weight (approximately 65.4 kDa), and detection sensitivity at physiological expression levels are primary considerations . Immunohistochemistry applications require antibodies validated for the specific fixation method (formalin, paraformaldehyde, etc.) and tissue processing approach (paraffin-embedded, frozen sections) being employed. Flow cytometry applications necessitate antibodies that recognize native epitopes on cell surfaces, preferably with direct fluorophore conjugation for simplicity . For all applications, species cross-reactivity must align with experimental models—while some antibodies detect human, mouse, and rat IL1R1, others may be species-specific . Publication history provides valuable validation, with antibodies cited in peer-reviewed studies demonstrating real-world utility. Finally, technical support availability should be considered, particularly for challenging applications or troubleshooting scenarios. Researchers should request validation data specific to their application and experimental system before committing to a particular antibody clone.

How do different clones of IL1R1 recombinant antibodies compare in detecting various isoforms and post-translational modifications?

Different clones of IL1R1 recombinant monoclonal antibodies vary significantly in their ability to detect specific isoforms and post-translational modifications due to epitope differences. Antibodies targeting epitopes in regions common to all isoforms will detect multiple variants, while those recognizing isoform-specific regions can distinguish between variants like isoform 1 (canonical signaling) and isoform 2 (alternative signaling) . Epitope location also determines sensitivity to post-translational modifications—antibodies recognizing glycosylation sites may show reduced binding to glycosylated forms, while phospho-specific antibodies detect only phosphorylated receptor states. Domain-specific antibodies can provide functional insights; those targeting the extracellular domain detect both membrane-bound and soluble forms, while cytoplasmic domain-specific antibodies detect only full-length receptors . When comparing clones, researchers should consider epitope mapping data alongside application-specific validation. Western blot patterns across multiple cell lines or tissues can reveal differences in isoform detection, appearing as bands at different molecular weights. For comprehensive analysis of IL1R1 biology, researchers may need to employ multiple antibody clones recognizing different epitopes to capture the full spectrum of receptor variants and modifications present in their experimental system.

IL1R1 Antibodies in Disease Research Models

IL1R1 recombinant monoclonal antibodies have become indispensable tools in studying the role of IL-1 signaling across diverse disease models. These applications span neurodegenerative disorders, autoimmune conditions, and inflammatory diseases, reflecting the broad pathological relevance of IL1R1.

Research indicates that IL1R1 signaling is implicated in numerous disease states, including Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, multiple sclerosis, schizophrenia, and prion diseases . The availability of high-quality, well-characterized antibodies has facilitated mechanistic studies in these disease contexts.

How are IL1R1 antibodies used to investigate neuroinflammatory mechanisms in neurodegenerative disease models?

In neurodegenerative disease models, IL1R1 recombinant monoclonal antibodies serve as critical tools for interrogating neuroinflammatory mechanisms. In Alzheimer's disease models, these antibodies help track IL1R1 expression changes in response to amyloid-β-peptide (Aβ) plaques, revealing how receptor upregulation correlates with disease progression and microglial activation states . Immunohistochemistry using IL1R1 antibodies can map receptor distribution in proximity to pathological features, identifying cell populations that may drive or respond to neuroinflammation. In Parkinson's disease models, IL1R1 antibodies help investigate how α-synuclein aggregates trigger inflammatory responses, with particular focus on microglial and astrocytic IL1R1 expression patterns . For mechanistic studies, co-immunoprecipitation using IL1R1 antibodies can identify disease-specific changes in receptor-associated signaling complexes. Time-course analyses across disease stages using quantitative approaches like Western blotting can establish temporal relationships between IL1R1 expression, inflammatory markers, and neurodegeneration. Importantly, these antibodies help resolve controversies regarding cell-specific IL1R1 expression in the CNS, particularly in microglia, where conflicting reports exist regarding receptor presence under basal and inflammatory conditions .

What insights have IL1R1 antibodies provided about receptor expression in clinical samples from inflammatory diseases?

IL1R1 recombinant monoclonal antibodies have yielded valuable insights from clinical samples across various inflammatory diseases. In rheumatoid arthritis tissues, immunohistochemistry with IL1R1 antibodies has revealed increased receptor expression in synovial fibroblasts and infiltrating immune cells, correlating with disease severity and treatment response. Analysis of multiple sclerosis brain tissue using IL1R1 antibodies has demonstrated altered receptor distribution at lesion sites, with particular upregulation in reactive astrocytes at the lesion edge . In type 2 diabetes samples, IL1R1 antibody staining has identified pancreatic islet cells expressing the receptor, supporting the role of IL-1 signaling in β-cell dysfunction. Comparative studies between healthy and diseased tissues using consistent IL1R1 antibody-based detection methods have established disease-specific expression patterns. Beyond qualitative observations, quantitative assessment of soluble IL1R1 in patient serum using antibody-based ELISAs has explored its potential as a biomarker for disease activity . The ratio between membrane-bound and soluble IL1R1 forms appears altered in several inflammatory conditions, potentially reflecting dysregulated receptor processing. These clinical findings, enabled by specific antibodies, have directly informed therapeutic strategies targeting the IL-1 pathway in inflammatory diseases.

How can IL1R1 antibodies help evaluate the efficacy of therapies targeting the IL-1 signaling pathway?

IL1R1 recombinant monoclonal antibodies provide essential tools for evaluating therapies targeting the IL-1 signaling pathway across multiple dimensions. For receptor occupancy studies, non-competing IL1R1 antibodies can quantify what percentage of receptors remain available following administration of IL-1 blocking agents like anakinra (IL-1Ra). This approach helps establish optimal dosing regimens by correlating receptor occupancy with clinical or physiological outcomes. When evaluating downstream effects of pathway inhibition, IL1R1 antibodies used in phospho-flow cytometry or Western blotting can measure changes in signaling cascade activation (like MAPK or NF-κB phosphorylation) following therapeutic intervention . For receptor expression modulation, quantitative approaches using calibrated IL1R1 antibodies can determine whether therapies alter receptor levels through transcriptional regulation or protein turnover mechanisms. In target engagement studies, competitive binding between therapeutics and labeled IL1R1 antibodies can confirm molecular interaction with the intended target. Long-term treatment effects can be monitored through periodic sampling and IL1R1 immunostaining to assess whether therapies produce sustained changes in receptor expression patterns. These antibody-based approaches collectively provide mechanistic understanding of therapeutic efficacy beyond simple clinical endpoints, potentially identifying responder/non-responder biomarkers and informing next-generation therapeutic design.

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.