Recombinant Mouse Interleukin-1 receptor antagonist protein (Il1rn)

Shipped with Ice Packs
In Stock

Description

Mechanism of Action

IL1RN functions as a competitive inhibitor of IL-1 signaling through two primary mechanisms:

  • Receptor Blockade: Binds to IL-1RI with high affinity, preventing IL-1α/β from activating downstream proinflammatory pathways .

  • Decoy Receptor Interaction: Attaches to IL-1RII, a non-signaling receptor, further sequestering IL-1 cytokines .

This dual action suppresses NF-κB activation, cytokine production (e.g., TNF-α, IL-8), and inflammatory cell recruitment .

Inflammatory Disease Models

  • Lung Injury: Mesenchymal stem cells (MSCs) secreting IL1RN reduced IL-1α mRNA levels by 7.3-fold in bleomycin-induced pulmonary fibrosis models, outperforming recombinant IL1RN delivery .

  • Arthritis: Demonstrates efficacy in murine rheumatoid arthritis models by decreasing synovial inflammation and cartilage degradation .

Cancer Studies

  • Oral Squamous Cell Carcinoma (OSCC): Low IL1RN expression correlates with tumor aggressiveness. Recombinant IL1RN inhibits IL-1-mediated angiogenesis (VEGF/IL-8 suppression) and reduces metastasis in xenograft models .

  • Melanoma and Breast Cancer: Suppresses tumor growth by blocking IL-1-induced cyclooxygenase-2 and glycolytic pathways .

Neurological and Dermatological Studies

  • Allergic Contact Dermatitis: Used to validate IL-1β-targeting therapies in preclinical trials .

  • Sickness Behavior: Central administration improves behavioral outcomes in aged mice with inflammation-induced cognitive deficits .

Clinical and Therapeutic Relevance

ConditionEffect of IL1RNReference
Rheumatoid ArthritisReduces joint swelling and erosions (Anakinra, a human IL1RN analog, is FDA-approved)
DIRA (Genetic Disorder)Biallelic IL1RN mutations cause systemic inflammation; recombinant IL1RN reverses symptoms
OsteoarthritisIntra-articular IL1RN mRNA injections reduce pain and cartilage degradation

Comparative Efficacy in Delivery Systems

Delivery MethodAdvantagesLimitations
Recombinant ProteinImmediate bioavailability; standardized dosingShort half-life; frequent administration
MSC-Secreted IL1RNSustained release; targets inflamed tissuesComplex production; regulatory hurdles
Gene Therapy (mRNA)Long-term expression; single-dose potentialImmunogenicity risks

Challenges and Future Directions

While recombinant IL1RN shows promise, its short plasma half-life (~4–6 hours) necessitates advanced delivery systems, such as nanoparticle encapsulation or gene editing of MSCs . Ongoing research focuses on optimizing pharmacokinetics and expanding applications to sepsis, diabetes, and neurodegenerative diseases .

Product Specs

Buffer
0.2 µm filtered PBS, pH 7.4, lyophilized
Form
Liquid or lyophilized powder
Lead Time
5-10 business days
Shelf Life
Shelf life depends on several factors including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized formulations have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquot to prevent repeated freeze-thaw cycles.
Tag Info
Tag-free
Synonyms
Il1rn; Il-1raInterleukin-1 receptor antagonist protein; IL-1RN; IL-1ra; IRAP; IL1 inhibitor
Datasheet & Coa
Please contact us to get it.
Expression Region
27-178aa
Mol. Weight
17.3 kDa
Protein Length
Full Length of Mature Protein
Purity
>97% as determined by SDS-PAGE.
Research Area
Immunology
Source
E.Coli
Species
Mus musculus (Mouse)
Target Names
Il1rn
Uniprot No.

Target Background

Function
This protein inhibits interleukin-1 activity by binding to the IL1R1 receptor, thereby preventing its interaction with the IL1RAP coreceptor and subsequent signaling. It exhibits no interleukin-1-like activity.
Gene References Into Functions
  • IL1Ra plays a critical role in maintaining the diversity and composition of the gut microbiota and may influence TLR4-mediated mucosal Th17 cell induction in autoimmune disease (PMID: 28645307).
  • Preclinical studies indicate that IL-1Ra may be a therapeutic target for bronchopulmonary dysplasia by addressing key pathophysiological features, including ex vivo airway hyperreactivity caused by maternal inflammation and postnatal hyperoxia (PMID: 27482635).
  • IL1RA release in response to genotoxic stress contributes to the immunosuppressive effects of apoptotic cells (PMID: 26873573).
  • Mesenchymal stem cell (MSC)-secreted IL1RA controls macrophage polarization toward an M2 phenotype and inhibits B cell differentiation in vivo (PMID: 26661518).
  • MSC allotransplantation reduces Con A liver injury by increasing IL-10 production through an IL1Ra-dependent macrophage switch (PMID: 26276675).
  • Small intestinal eosinophils regulate intestinal homeostasis by controlling Th17 cells via IL-1Ra production (PMID: 26951334).
  • In cystic fibrosis, NLRP3 contributes more to deleterious inflammatory responses than NLRC4, correlating with deficient NLRC4-dependent IL-1Ra production (PMID: 26972847).
  • IL-1Ra deficiency may promote early atherosclerotic plaque development (PMID: 26413013).
  • IL-1Ra is involved in regulating MMP-13 expression independently of the IL-1 signaling pathway (PMID: 26474296).
  • F-spondin and IL-1Ra have novel roles in type B CpG oligodeoxynucleotide-mediated macrophage proliferation and migration (PMID: 26042735).
  • Studies highlight the coordinated role of adaptive and innate immunity in autoimmune disease development (PMID: 26108163).
  • Certain stimuli enhance interleukin-1 receptor antagonist (IL-1RA) production (PMID: 25909959).
  • IL-1Ra may offer protection against Ebola virus infection (PMID: 26209680).
  • IL-1ra regulates baseline GABAergic transmission in the central amygdala and is crucial for ethanol's effects at these synapses (PMID: 25479427).
  • In obesity, normalizing IL-1Ra improves insulin sensitivity by reducing liver inflammation and enhancing hepatic insulin sensitivity, independently of weight changes (PMID: 25244011).
  • Loss of the IL-1ra gene is associated with spontaneous arthritis (PMID: 21414240).
  • Aspergillus fumigatus galactosaminogalactan inhibits cytokine production by inducing interleukin-1 receptor antagonist (PMID: 24603878).
  • IL-1Ra deficiency in A. actinomycetemcomitans-infected osteoblasts promotes inflammatory cytokine expression and alters genes involved in bone resorption (PMID: 24566623).
  • IL-1rn(-/-) mice develop spinal abnormalities resembling human intervertebral disc degeneration (PMID: 23396662).
  • Classical IL-6 signaling modulates the IL-1ra pathway, demonstrating IL-6's anti-inflammatory and protective effects (PMID: 22980031).
  • In ischemic heart and hypoxic cardiomyocytes, IL-1ra inhibits mitochondria-activated caspases (PMID: 23308180).
  • IL-1ra deficiency leads to spontaneous autoimmune arthritis in Balb/c mice, but not in DBA/1 mice (PMID: 22942082).
  • CCR5 deficiency induces melanoma cell apoptosis via NF-kappaB inhibition and IL-1Ra upregulation (PMID: 22567084).
  • Hepatocyte-derived IL-1Ra contributes to hepatitis resolution (PMID: 22539301).
  • IL-1Ra suppresses local responses in LPS-injected eyes (PMID: 22267332).
  • IL-1ra may regulate immune responses and potentially alcohol consumption (PMID: 21309947).
  • IL-1/IL-1Ra signaling imbalance at IL-1R1 modulates cardiac remodeling severity after myocardial infarction (PMID: 22140485).
  • PGC-1alpha and AMP-activated protein kinase control the expression of inflammatory mediators in the liver, including interleukin-1 receptor antagonist induction (PMID: 22117073).
  • Leishmania major infection in IL-1RA(-/-) mice worsens disease outcome (PMID: 21525884).
  • Intestinal epithelia secrete sIL-1Ra in a TLR5-dependent manner, suggesting that TLR5 loss promotes inflammation (PMID: 20844479).
  • IL-1ra exhibits a protective effect in TLR9-associated liver injury (PMID: 20727895).
  • Different cellular sources of IL-1Ra respond to various systemic inflammatory stimuli (PMID: 20639493).
  • Psoriasis-like lesions in Il1rn-deficient mice depend on TNF, but not IL-6 or IL-17 (PMID: 20610641).
  • TLR4-mediated IL-1Ra production negatively regulates hyaluronan-induced lung inflammation (PMID: 20357263).
  • IL-1Ra deficiency in inflammatory cells induces aortic valve inflammation, with TNF-alpha involvement in aortic stenosis development (PMID: 20110570).
  • MSK1 regulates IL-1ra transcription upon TLR activation in macrophages (PMID: 19922413).
  • Studies examining the effects of MPC-11 myeloma and MPC-11 + IL-1 receptor antagonist treatment on mouse bone properties (PMID: 11792572).
  • The balance between IL-1 and IL-1Ra during the primary immune response influences T cell effector function (PMID: 11810022).
  • AP-1 regulates intracellular IL-1 receptor antagonist gene transcription (PMID: 11933077).
  • IL-1 receptor antagonist genetic alterations affect plasma cholesterol and foam cell lesion size (PMID: 11983917).
  • IL-1Ra is highly expressed in liver, spleen, and white adipose tissue (WAT), upregulated in obesity (PMID: 12716739).
  • IL-1ra deficiency promotes neointimal formation after arterial injury (PMID: 12874179).
  • CB1 and CB2 receptors modulate IL-1ra release from glial cells; IL-1ra is essential for the neuroprotective effects of cannabinoids (PMID: 12878687).
  • Sleep deprivation significantly increases interleukin-1 receptor antagonist (PMID: 14583241).
  • IL-1Ra suppresses lesion development during early atherogenesis and modulates plaque composition (PMID: 15059807).
  • T cell IL-1 receptor antagonist deficiency disrupts immune system homeostasis, with TNF-alpha playing a key role in T cell activation (PMID: 15578092).
  • Hepatocyte-derived IL-1ra controls inflammatory dendritic cell activation and migration in granulomatous liver disease (PMID: 15749858).
  • IL-1Ra regulates adipogenesis, food intake, and energy expenditure (PMID: 16306368).
  • IL-1Ra reduces insulin sensitivity in rats through decreased muscle glucose uptake (PMID: 16385385).
  • IL-1ra absence may suppress TGF-beta signaling, crucial for collagen deposition and neovascularization in wound healing (PMID: 16622029).
Database Links

KEGG: mmu:16181

STRING: 10090.ENSMUSP00000110126

UniGene: Mm.882

Protein Families
IL-1 family
Subcellular Location
[Isoform 1]: Secreted.; [Isoform 2]: Cytoplasm.

Q&A

What is mouse Interleukin-1 receptor antagonist protein (Il1rn) and how does it function in inflammatory responses?

Il1rn belongs to the IL-1 family of cytokines and functions as a competitive inhibitor of IL-1α and IL-1β by binding to the same receptor without inducing downstream signaling. The protein serves as a natural regulator of IL-1-mediated inflammatory responses in various tissues and physiological systems. Il1rn binds specifically to the IL-1R type I, effectively blocking the pro-inflammatory actions of IL-1 . This competitive inhibition represents a physiological mechanism for controlling excessive inflammation.

In experimental settings, recombinant Il1rn has demonstrated protective effects against TNF-induced lethal shock in mice, highlighting its significant role in modulating inflammatory cascades . The protein's function illustrates the complex regulatory mechanisms that balance pro-inflammatory and anti-inflammatory processes in mammalian immune systems.

What are the strain-specific effects of Il1rn deficiency in laboratory mice?

Il1rn deficiency manifests differently depending on the genetic background of the mice. Research has identified at least three distinct inflammatory phenotypes that develop spontaneously in Il1rn-deficient mice:

  • Arterial inflammation - Observed in outbred 129×MF1 Il1rn−/− mice and BALB/c mice, characterized by transmural inflammation at sites of turbulence in elastic arteries

  • Psoriasiform cutaneous inflammation - Predominantly seen in Il1rn−/− BALB/c mice

  • Inflammatory arthritis - Confirmed in BALB/c strains

Additionally, deficiency of Il1rn affects bone mineral density (BMD) in a strain-dependent manner, decreasing BMD in Balb/c mice while increasing it in DBA/1−/− mice compared to respective wild type counterparts . These strain-specific effects highlight the importance of genetic background in determining the phenotypic consequences of cytokine imbalance.

How is Il1rn expression regulated in mesenchymal stem cells (MSCs)?

Il1rn expression in MSCs is highly regulated and restricted to specific subpopulations. Fractionation studies have demonstrated that MSCs are the principal source of Il1rn in murine bone marrow, with expression limited to unique subpopulations . Approximately 24% of immunodepleted murine MSCs express Il1rn, as determined by immunostaining techniques .

In human MSCs, a distinct subpopulation (approximately 5%) expresses Il1rn protein, suggesting that this expression pattern is conserved across species, although at different frequencies . The temporal regulation of Il1rn expression in MSCs shows a significant delay between IL-1 exposure and protein expression, with studies indicating a 72-hour lag before significant increases in Il1rn secretion following IL-1α stimulation .

What are the optimal methods for detecting and quantifying Il1rn expression in experimental systems?

For comprehensive analysis of Il1rn expression, researchers should employ multiple complementary techniques:

Transcriptional Analysis:

  • Serial Analysis of Gene Expression (SAGE) has successfully identified Il1rn transcripts in MSC populations

  • Real-time PCR can quantify Il1rn mRNA levels in tissues following experimental interventions

Protein Detection and Quantification:

  • ELISA assays of conditioned media provide accurate quantification of secreted Il1rn

  • Flow cytometry with appropriate antibodies can identify and quantify Il1rn-expressing cell subpopulations

  • Immunostaining techniques are effective for visualizing Il1rn expression patterns within heterogeneous cell populations

Functional Assays:

  • The IL-1α-dependent helper T lymphocyte cell line D10.G4.1 proliferation assay provides a biological readout of Il1rn activity, with inhibition of proliferation indicating functional Il1rn

When planning Il1rn detection experiments, researchers should consider including appropriate controls, including neutralizing anti-Il1rn antibodies to confirm specificity of observed effects.

What experimental approaches are most effective for studying Il1rn function in inflammatory disease models?

Multiple experimental approaches have proven effective for investigating Il1rn function:

Genetic Models:

  • Il1rn−/− knockout mice on different genetic backgrounds to study strain-specific effects

  • Double knockout models (e.g., Il1r1−/−Il1rn−/−) to determine receptor dependency of observed phenotypes

Administration Methods for Recombinant Il1rn:

  • Adenoviral vector delivery - Provides localized, relatively long-term expression

  • Osmotic minipumps - Enables consistent systemic administration throughout experimental duration

  • Direct injection - Appropriate for acute studies and dose-response experiments

Readout Parameters:

  • Cytokine profiling (TNF-α, IL-1α, IL-6) in tissues and biological fluids

  • Immune cell trafficking analyses in affected tissues

  • Histopathological assessments of inflammatory lesions

  • Functional measurements specific to the tissue under investigation

The bleomycin (BLM)-induced lung injury model has been particularly informative, allowing comparative assessment of different Il1rn delivery methods against MSC-mediated protection .

How should researchers control for species-specific differences when working with recombinant mouse Il1rn?

Species-specific differences are critical considerations in experimental design with mouse Il1rn:

  • TNF-Mediated Systems: Significant differences exist between murine TNF and human TNF in systems such as lethal shock and IL-6 induction. When using Il1rn as an intervention, researchers must account for these species-specific interactions .

  • Cross-Species Applications: When testing murine Il1rn in human cell systems or vice versa, validation experiments should confirm cross-reactivity and equivalent biological activity.

  • Expression Systems: The source of recombinant Il1rn (bacterial, mammalian, or insect cell expression systems) may affect glycosylation patterns and biological activity. Researchers should validate recombinant protein functionality regardless of source.

  • Control Groups: Proper experimental design should include:

    • Species-matched recombinant proteins

    • Vehicle controls matched to the recombinant protein buffer

    • Denatured protein controls to distinguish between specific biological activity and nonspecific protein effects

  • Neutralization Controls: Including experiments with neutralizing antibodies against Il1rn confirms that observed effects are specifically due to Il1rn activity rather than contaminants or buffer components .

How do you resolve the apparent contradiction between protective effects of exogenous Il1rn and the complex phenotypes of Il1rn knockout mice?

This apparent contradiction highlights the context-dependent role of Il1rn in inflammatory regulation:

Protective Effects of Exogenous Il1rn:

  • Recombinant Il1rn protects mice against lethal TNF injection, demonstrating acute anti-inflammatory potential

  • MSC-derived Il1rn inhibits bleomycin-induced inflammation and fibrosis in lungs, indicating therapeutic potential in tissue injury

Complex Phenotypes in Knockout Models:

  • Il1rn deficiency leads to strain-specific inflammatory diseases affecting multiple organ systems

  • The severity and nature of inflammation varies dramatically between strains (arteritis, psoriasiform inflammation, arthritis)

Resolution Framework:

  • Temporal considerations: Acute administration of recombinant Il1rn differs fundamentally from lifelong absence in knockout models

  • Tissue-specific effects: Il1rn function may differ across tissues and inflammatory contexts

  • Genetic compensation: Chronic absence may trigger compensatory mechanisms absent in acute intervention models

  • Dose-dependent effects: Physiological levels in normal tissues versus therapeutic doses of recombinant protein

Researchers should interpret data with these complexities in mind, recognizing that Il1rn's effects depend on genetic context, tissue environment, timing of intervention, and the specific inflammatory trigger being studied.

What explains the differential efficacy of Il1rn versus mesenchymal stem cells in inflammatory disease models?

Studies comparing MSC administration to recombinant Il1rn delivery have yielded important insights into their differential efficacy:

Comparative Efficacy Observations:

  • MSC administration more effectively inhibits BLM-induced increases in TNF-α, IL-1α, and IL1RN mRNA in lung compared to recombinant Il1rn delivered via adenoviral infection

  • MSCs are more effective than recombinant Il1rn in preventing lymphocyte and neutrophil trafficking into injured lung tissue

  • Systemic administration of recombinant Il1rn only modestly inhibits BLM-induced increases in IL-1α mRNA levels, while MSC administration significantly reduces these levels

Explanatory Factors:

  • Multifactorial Effects: MSCs likely exert protection through multiple mechanisms beyond Il1rn production

  • Cellular Integration: MSCs can home to sites of injury and respond dynamically to local environmental cues

  • Sustained Production: MSCs may provide more consistent local production of Il1rn compared to exogenous administration

  • Subpopulation Specialization: Specific MSC subpopulations express Il1rn, suggesting specialized immunomodulatory functions

  • Paracrine Effects: MSCs secrete multiple bioactive factors that work synergistically with Il1rn

These differences underscore the potential advantages of cell-based therapies over recombinant proteins in complex inflammatory diseases, while also highlighting the need for deeper understanding of MSC subpopulation biology.

How can Il1rn-expressing MSC subpopulations be harnessed for therapeutic applications?

The identification of Il1rn-expressing MSC subpopulations offers promising therapeutic avenues:

Isolation and Enrichment Strategies:

  • Immunodepletion techniques have successfully enriched for Il1rn-expressing MSCs, increasing secreted Il1rn on a per cell basis

  • Flow cytometry-based sorting could potentially isolate the approximately 24% of murine MSCs or 5% of human MSCs that express Il1rn

Therapeutic Potential:

  • In bleomycin-induced lung injury models, MSCs demonstrate superior efficacy to recombinant Il1rn in reducing inflammation and preventing fibrosis

  • The cellular vector approach may provide more sustained and regulated Il1rn delivery compared to recombinant protein administration

Translational Considerations:

  • The conservation of Il1rn-expressing MSC subpopulations across species supports potential translation to human applications

  • Il1rn-expressing MSCs may represent a specialized stromal cell subtype with natural roles in modulating bone turnover and inflammation

  • Targeting these cells to specific tissue sites could enhance their therapeutic efficacy

Practical Applications:

  • Chronic inflammatory diseases, particularly those affecting the lung, represent promising targets

  • Combined approaches using engineered MSCs with enhanced Il1rn expression could potentially improve efficacy

  • Patient-derived MSCs could be screened for high Il1rn-expressing subpopulations prior to expansion and reinfusion

What genetic interactions determine the phenotypic outcomes of Il1rn deficiency?

Genetic interaction studies provide critical insights into the mechanisms underlying Il1rn-related pathologies:

Receptor Dependency:

  • Epistasis studies between Il1rn and Il1r1 demonstrate that the inflammatory phenotypes in Il1rn-deficient mice depend on IL-1 receptor signaling

  • In a study of Il1rn−/−Il1r1+/+ and Il1r1−/−Il1rn−/− littermates followed to 200 days, aortic root lesions developed in 10/12 mice with functioning IL-1 receptors but in 0/20 mice lacking IL-1 receptors

Strain-Specific Effects:

  • The genetic background significantly influences disease manifestations in Il1rn-deficient mice

  • QTL (Quantitative Trait Locus) analysis can identify modifier genes that influence phenotypic outcomes

  • Bone mineral density effects of Il1rn deficiency demonstrate opposing patterns in different mouse strains

Genomic Analysis Approaches:

  • Genenetwork analysis can help reconstruct genetic networks based on genome expression data

  • Correlation analyses between Il1rn and other genes in relevant datasets (e.g., spleen and bone) provide insights into strain-specific effects

Understanding these genetic interactions is essential for predicting therapeutic responses and developing targeted interventions for IL-1-mediated inflammatory diseases.

How does Il1rn interface with other cytokine systems in complex inflammatory networks?

Il1rn functions within an intricate network of cytokine interactions that determines inflammatory outcomes:

TNF-α Interactions:

Disease-Specific Sensitization:

  • The increased susceptibility of tumor-bearing mice to human TNF is not mediated by IL-1, as demonstrated by Il1rn intervention studies

  • Similarly, sensitization to TNF by RU38486 or D(+)-galactosamine appears independent of IL-1 pathways

Temporal Dynamics:

  • There exists a significant delay between IL-1 exposure and Il1rn expression (approximately 72 hours), creating a temporal window where inflammatory signals dominate

  • High endogenous Il1rn levels in tissues reflect ongoing inflammatory responses rather than resolution

Bone Homeostasis Network:

  • Both TNF-α and IL-1 function as potent bone-resorbing factors, effects that can be modulated by Il1rn

  • Il1rn-expressing MSC subpopulations may represent specialized stromal cells that regulate bone turnover through cytokine modulation

Understanding these complex interactions is essential for designing effective interventions that target multiple nodes in inflammatory networks rather than single mediators.

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.