IL 1RA Horse

Interleukin-1 Receptor Antagonist Horse Recombinant
Shipped with Ice Packs
In Stock

Description

Mechanism of Action

IL-1RA functions as a competitive inhibitor of IL-1α and IL-1β by binding to the IL-1 type I receptor (IL-1RI) without activating downstream signaling . Key interactions include:

  • Receptor Binding: Prevents IL-1 from recruiting the co-receptor IL-1RAP, blocking inflammation .

  • Decoy Receptor: IL-1RA binding to the inert IL-1RII may inadvertently potentiate IL-1 activity by reducing antagonist availability .

Gene Therapy for Osteoarthritis

A 2024 study evaluated self-complementary adeno-associated virus (scAAVIL-1ra) in 16 horses with induced carpal osteoarthritis :

ParameterImprovement ObservedSource
Lameness Score1.2-point reduction (0–5 scale)
Prostaglandin E2 (PGE2)30 pg/mL decline in synovial fluid
Synovial IL-1RA Levels0.5–9 μg/mL sustained over 70 days
HistopathologyReduced chondrocyte loss, fewer bone lesions

This therapy also induced T-cell infiltration in synovial membranes, a common viral transduction response .

Biomarker Correlation in Racehorses

A 2024 study of 312 Standardbred racehorses developed sensitive ELISAs for plasma IL-1β and IL-1RA :

  • Concentration Ranges: IL-1β (0–48 ng/mL), IL-1RA (0–112 ng/mL) .

  • Correlation: Strong positive correlation (Pearson’s r = 0.911) between IL-1β and IL-1RA at rest .

  • Outliers: Seven horses exhibited elevated levels (>6,000 pg/mL IL-1β; >2,000 pg/mL IL-1RA), suggesting active inflammation .

IRAP (Interleukin-1 Receptor Antagonist Protein)

IRAP therapy involves autologous blood processing to concentrate IL-1RA for intra-articular injection :

  • Protocol: 50 mL blood incubated with glass beads to amplify IL-1RA, followed by 3–5 injections at 7–10-day intervals .

  • Efficacy: Best results in joints with mild-to-moderate osteoarthritis unresponsive to corticosteroids .

Recombinant IL-1RA Production

Commercial recombinant equine IL-1RA (e.g., GoldBio’s 4313) is produced in E. coli for research use, showing in vitro and in vivo anti-inflammatory activity .

Product Specs

Introduction
Interleukin-1 receptor antagonist (IL-1RA) is a protein belonging to the interleukin 1 cytokine family. It acts by blocking the activity of interleukin-1 alpha (IL1A) and interleukin-1 beta (IL1B), thereby regulating various immune and inflammatory responses associated with interleukin-1. The gene encoding IL-1RA, along with five other structurally similar cytokine genes, forms a cluster spanning roughly 400 kb on chromosome 2. Polymorphisms in this gene have been linked to an elevated risk of osteoporosis-related fractures and gastric cancer. Four alternatively spliced transcript variants, which produce distinct isoforms of IL-1RA, have been identified.
Description
Recombinant Horse Interleukin-1 Receptor Antagonist is a single, non-glycosylated polypeptide chain comprising 152 amino acids, resulting in a molecular weight of 17.4 kDa. It is produced in E.coli cells. The purification process involves the use of proprietary chromatographic techniques to ensure high purity.
Physical Appearance
Sterile Filtered White lyophilized powder.
Formulation
The IL-1RA undergoes lyophilization from a 0.2 µm filtered solution concentrated in phosphate-buffered saline (PBS) at pH 7.4.
Solubility
To reconstitute the lyophilized IL-1RA, it is recommended to dissolve it in sterile water to a minimum concentration of 100 µg/ml. This solution can be further diluted using other aqueous solutions as needed.
Stability
Lyophilized IL-1RA demonstrates stability at room temperature for up to 3 weeks. However, for extended storage, it is recommended to store it in a desiccated state below -18°C. Once reconstituted, IL-1RA should be stored at 4°C for a period of 2-7 days. For long-term storage, it is advisable to add a carrier protein such as 0.1% HSA or BSA. It is important to avoid repeated freeze-thaw cycles to maintain stability.
Purity
The purity of the recombinant Horse Interleukin-1 Receptor Antagonist is greater than 95.0%, as determined by two methods: Reverse-Phase High-Performance Liquid Chromatography (RP-HPLC) and Sodium Dodecyl Sulfate-Polyacrylamide Gel Electrophoresis (SDS-PAGE).
Biological Activity
The biological activity of the recombinant Horse Interleukin-1 Receptor Antagonist is determined by its ability to inhibit IL-1α-dependent proliferation of murine D10.G4.1 helper T cells. The ED50, representing the concentration at which 50% inhibition is observed, is less than 3.0 µg/ml. This corresponds to a specific activity greater than 333 IU/mg in the presence of 50 pg/ml recombinant human IL-1α.
Synonyms
Interleukin-1 receptor antagonist protein, IL-1RN, IL-1ra, IRAP, IL1 inhibitor, IL1RN, IL1RA.
Source
Escherichia Coli.
Amino Acid Sequence
HPLGKRPCKM QAFRIWDVNQ KTFYMRNNQL VAGYLQESNT KLQEKIDVVP IEPDALFLGL HGRKLCLACV KSGDEIRFQL EAVNITDLSK NKEENKRFTF IRSNSGPTTS FESAACPGWF LCTAQEADRP VSLTNKPKES FMVTKFYLQE DQ.

Q&A

What is the physiological relationship between IL-1β and IL-1RA in horses?

IL-1β and IL-1 receptor antagonist (IL-1RA) are interconnected inflammatory mediators that demonstrate a positive correlation in horses. Studies of Standardbred racehorses (n=312) show that both cytokines increase significantly following traumatic injury, with plasma concentrations ranging from 0-48 ng/mL for IL-1β and 0-112 ng/mL for IL-1RA. This relationship is important because IL-1RA blocks inflammatory signaling triggered by IL-1β, and an imbalance with overexpression of IL-1β relative to IL-1RA may drive inflammatory disease processes in equine joints. The correlation between these biomarkers (Pearson's r = 0.875) provides critical insight into disease states and potential therapeutic effectiveness.

How do researchers accurately measure IL-1β and IL-1RA levels in equine samples?

Researchers have developed specialized sandwich enzyme-linked immunosorbent assays (ELISAs) to overcome traditional sensitivity limitations in detecting equine IL-1β and IL-1RA. These advanced assays employ analyte-specific polyclonal antibodies (PAb) for capture and biotinylated conjugates for enhanced detection sensitivity. The validation process includes rigorous testing for linearity, specificity, precision, and accuracy, with recombinant proteins used to generate standard curves for quantification. During validation, researchers confirm absence of cross-reactivity with other proteins and verify that serial dilution of plasma samples produces proportional decreases in signal intensity. This methodological approach has enabled accurate measurement of these cytokines in experimental settings and clinical evaluations of horses.

What is the pathophysiological significance of IL-1β in equine osteoarthritis?

IL-1β serves as a primary inflammatory mediator in equine joint degeneration, initiating a cascade of catabolic events that contribute to osteoarthritis progression. This pro-inflammatory cytokine triggers the expression of matrix-degrading enzymes (including MMP-3 and MMP-13), stimulates production of prostaglandin E2, and contributes to chondrocyte apoptosis and matrix degradation. The significance of IL-1β in equine osteoarthritis is evidenced by research showing that targeted inhibition through IL-1RA gene therapy results in measurable clinical improvements, including reduced lameness (improvement of 1.2 on a 0-5 scale), decreased prostaglandin E2 levels (decline of approximately 30 pg/mL), and histological preservation of articular cartilage structure.

How are equine osteoarthritis models established for IL-1RA research?

Researchers establish equine osteoarthritis models primarily through surgical induction of osteochondral chip fragments. The validated protocol involves arthroscopic creation of fragments in the middle carpal joint while performing sham operations on contralateral joints as controls. This approach allows for paired comparison within each subject. The experimental timeline typically extends 70 days post-induction, with regular clinical evaluations including lameness assessment, imaging, and biomarker analysis throughout the study period. Terminal evaluations include gross examination, advanced imaging, and comprehensive histopathology. This model effectively replicates post-traumatic osteoarthritis pathophysiology and provides a controlled environment for testing IL-1RA therapeutic interventions while minimizing variables that might confound results interpretation.

What gene transfer techniques are most effective for delivering IL-1RA to equine joints?

Self-complementary adeno-associated viral vectors carrying the IL-1RA transgene cassette (scAAVIL-1ra) have demonstrated superior efficacy in equine joints. The technique involves direct intra-articular injection of the viral vector, which transduces synovial cells to produce therapeutic levels of IL-1RA protein within the joint environment. Research has evaluated dosages across a 100-fold range to determine optimal therapeutic concentrations. A dose of 5 × 10^12 viral genomes has been identified as providing sustained therapeutic levels (0.5-9 μg/mL in synovial fluid) without adverse effects. The vector design enables long-term transgene expression, with therapeutic levels maintained throughout 70-day experimental periods. This approach is advantageous compared to protein administration as it provides continuous local production of the anti-inflammatory mediator within the affected joint microenvironment.

What methods are used to assess the efficacy of IL-1RA gene therapy in horses?

Researchers employ a multimodal assessment approach combining clinical, biochemical, imaging, and histological evaluations. Clinically, standardized lameness scoring systems quantify functional improvement on a 0-5 scale. Biochemically, synovial fluid analysis measures key inflammatory mediators including IL-1RA concentrations (via ELISA) and prostaglandin E2 levels, while serum glycosaminoglycan (GAG) serves as a systemic biomarker of cartilage metabolism. Imaging assessments include radiography and advanced modalities to evaluate joint structural integrity. Histopathological evaluation employs standardized scoring systems examining articular cartilage (chondrocyte loss, chondrone formation) and subchondral bone (osteochondral splitting, lesion development). Additionally, immunohistochemical methods identify cellular infiltrates (e.g., CD3-positive lymphocytes) to characterize immune responses. Statistical analysis compares these metrics between treated and control groups, both within and between subjects, to comprehensively evaluate therapeutic efficacy.

What cellular and molecular mechanisms underlie the histological improvements observed with IL-1RA gene therapy?

The histological improvements following IL-1RA gene therapy stem from multiple cellular and molecular mechanisms. IL-1RA competitively inhibits IL-1β binding to its receptors, thereby blocking downstream NF-κB activation and subsequent pro-inflammatory gene transcription. This inhibition reduces expression of matrix-degrading enzymes (MMP-3, MMP-13) that would otherwise contribute to cartilage matrix breakdown. At the cellular level, IL-1RA therapy preserves chondrocyte viability and reduces apoptosis, evidenced by decreased chondrocyte loss and chondrone formation in histological specimens. The therapy also appears to maintain chondrocyte phenotypic stability, preventing dedifferentiation and inappropriate matrix synthesis. In subchondral bone, IL-1RA prevents osteochondral splitting and lesion development by modulating osteoclast activity and inflammatory bone remodeling. These mechanisms collectively preserve joint tissue homeostasis despite the presence of mechanical injury, demonstrating how targeted cytokine inhibition can interrupt the progression from acute injury to chronic degenerative disease.

What are the immunological implications of viral vector-mediated gene transfer in equine joints?

Viral vector administration in equine joints presents complex immunological considerations that impact both safety and efficacy. The observed perivascular infiltration of CD3-positive lymphocytes in synovial membranes indicates a T-cell mediated immune response to viral transduction. This response may limit transgene expression duration, affect repeated dosing potential, and possibly contribute to inflammation. Vector design features, such as the self-complementary AAV construct, influence immunogenicity, with modifications potentially reducing capsid antigenicity. Pre-existing neutralizing antibodies to viral vectors may exist in horses with prior environmental exposure, potentially reducing transduction efficiency. The local joint environment's relative immune privilege offers some protection against systemic immune responses, but intra-articular inflammation associated with osteoarthritis may enhance immune surveillance and response to vectors. Understanding these complex immunological interactions is crucial for optimizing therapeutic approaches, particularly when considering potential translation to clinical applications requiring repeated administrations over a horse's lifetime.

How can researchers address the variability in transgene expression observed across different studies?

Researchers should implement comprehensive standardization and validation protocols to address variability in transgene expression. First, vector production requires stringent quality control with consistent manufacturing processes and batch testing for potency and purity. Delivery techniques should be standardized with precise injection protocols, consistent joint volumes, and controlled post-injection management. Study designs should include internal controls with reporter gene constructs to quantify transduction efficiency independent of therapeutic effect. Advanced analytical methods, including digital droplet PCR for copy number assessment and single-cell RNA sequencing to identify transduced cell populations, can precisely characterize transgene integration and expression. Statistical approaches should employ mixed-effects models that account for individual horse variation, and dose-normalization can control for weight and joint size differences. Finally, researchers should develop standardized pharmacokinetic/pharmacodynamic models specifically for gene therapy approaches in equine joints to better predict and interpret variable responses across different experimental conditions.

What gene expression analyses provide the most insight into IL-1RA therapeutic mechanisms?

Comprehensive gene expression analysis for IL-1RA therapeutic evaluation should employ a multi-tiered approach starting with targeted PCR analysis of key inflammatory mediators, particularly IL-1β, TNF-α, MMP-3, and MMP-13 using validated equine-specific primers and probes. Quantification should utilize the ΔΔCt method with 18S as a reference gene to ensure accurate normalization. Beyond targeted analysis, RNA-sequencing provides unbiased examination of transcriptome-wide changes, potentially identifying unexpected pathway modulations. Researchers should complement these techniques with protein-level validation through ELISAs for key mediators (particularly IL-1RA and PGE2) and tissue-level expression analysis via immunohistochemistry to confirm cellular sources of relevant molecules. Temporal analysis capturing expression changes across multiple timepoints provides crucial insight into therapeutic dynamics, while single-cell approaches can identify cell-specific responses to treatment. Integration of these datasets through systems biology approaches allows for identification of master regulators and pathway intersections that mediate therapeutic effects, offering the most comprehensive understanding of IL-1RA's mechanism of action in equine osteoarthritis.

What optimization strategies might enhance IL-1RA gene therapy for equine osteoarthritis?

Future optimization of IL-1RA gene therapy should focus on several key areas. Vector engineering could incorporate tissue-specific promoters to restrict expression to synovial fibroblasts or utilize inducible systems responsive to inflammatory signals. Delivery system innovations might include hydrogel-based sustained release or nanoparticle formulations to enhance transduction efficiency. Combination therapies warrant exploration, particularly IL-1RA delivery alongside complementary anti-inflammatory or chondroprotective genes. Personalized dosing algorithms could be developed based on joint size, disease severity, and individual biomarker profiles. Advanced bioimaging approaches using reporter genes could enable non-invasive monitoring of transgene expression dynamics. Careful investigation of repeated dosing protocols is needed to determine optimal timing for maintaining therapeutic levels while minimizing immune responses. Finally, development of equine-specific vectors with modified capsids could improve tissue tropism and reduce immunogenicity. These multifaceted optimization strategies require systematic evaluation in relevant disease models before clinical translation.

How might the findings from equine IL-1RA studies translate to human osteoarthritis treatments?

Equine osteoarthritis research offers substantial translational value for human applications due to several significant similarities. Horses and humans share comparable joint biomechanics, cartilage thickness, and naturally occurring disease processes. The gene therapy approaches demonstrated in equine models, particularly using scAAVIL-1ra, provide proof-of-concept for human applications with similar vector constructs. The dose-finding studies in horses (identifying 5 × 10^12 viral genomes as optimal) provide scaling information for human trials, though with appropriate allometric adjustments. Safety profiles, including the observed lymphocytic infiltration patterns, inform monitoring protocols for human studies. The demonstrated duration of transgene expression (70+ days) suggests similar persistence might be achievable in human joints. Importantly, the functional outcome measures used in equine studies (lameness, imaging, biomarkers) have direct parallels in human clinical assessments. These translational insights could accelerate development of human gene therapy approaches while identifying potential challenges regarding immune responses, variability in expression, and optimal delivery protocols.

What novel biomarkers might complement IL-1β/IL-1RA measurements in evaluating therapeutic responses?

Emerging biomarker approaches to complement IL-1β/IL-1RA measurements should integrate multiple biological domains. Cartilage metabolism markers including CTX-II (type II collagen C-telopeptide) and COMP (cartilage oligomeric matrix protein) could provide direct evidence of reduced cartilage breakdown. Synovial fluid metabolomics profiles might identify novel small molecule signatures of treatment response. MicroRNA panels, particularly those regulating inflammation and matrix remodeling, could offer earlier response indicators than protein mediators. Advanced proteomics approaches may identify unexpected protein changes beyond canonical inflammatory pathways. Machine learning algorithms integrating multiple biomarker types could develop composite scores with greater predictive value than individual markers. Imaging biomarkers using contrast-enhanced MRI or molecular imaging could provide spatial information about treatment effects. Additionally, genetic polymorphism analysis might identify markers predicting response to IL-1RA therapy. These complementary biomarker approaches would provide a comprehensive assessment of therapeutic efficacy across multiple biological processes affected in osteoarthritis, potentially identifying responder/non-responder phenotypes and offering earlier indications of therapeutic success.

Product Science Overview

Introduction

Interleukin-1 Receptor Antagonist (IL-1Ra) is a naturally occurring anti-inflammatory protein that plays a crucial role in regulating the immune response by inhibiting the activities of interleukin-1 (IL-1). IL-1 is a pro-inflammatory cytokine involved in various inflammatory and autoimmune conditions. The recombinant form of IL-1Ra, specifically designed for equine use, has shown significant therapeutic potential in veterinary medicine.

Discovery and Development

IL-1Ra was first discovered in 1984 in the urine and serum of patients with leukemia and in the supernatant of cultured monocytes . It was named for its ability to competitively bind to the IL-1 receptor, thereby antagonizing the function of IL-1. The recombinant form of IL-1Ra has been developed to enhance its stability and bioactivity, making it suitable for therapeutic applications.

Mechanism of Action

IL-1Ra functions by binding to the IL-1 receptor on the cell membrane, preventing IL-1 from binding to its receptor and initiating pro-inflammatory signaling pathways . This competitive inhibition helps to reduce inflammation and modulate the immune response. In horses, IL-1Ra has been shown to be effective in treating conditions such as synovitis (inflammation of the joint lining) and mild to moderate osteoarthritis (OA) .

Production and Characterization

Recombinant IL-1Ra can be produced using various expression systems. One study reported the expression and characterization of recombinant IL-1Ra in Aspergillus oryzae, a filamentous fungus . This system offers advantages such as cost-effective production and enhanced stability of the recombinant protein. The recombinant IL-1Ra produced in Aspergillus oryzae was found to be N-glycosylated and exhibited good bioactivity and stability.

Therapeutic Applications in Equine Medicine

The therapeutic potential of IL-1Ra in equine medicine has been extensively studied. IRAP® (Interleukin-1 Receptor Antagonist Protein) therapy, originally developed in Europe, has been widely used in Germany and is now marketed in the United States by Arthrex VetSystems . This therapy is recommended for the treatment of synovitis and mild to moderate osteoarthritis in horses. The use of IL-1Ra in horses helps to reduce inflammation, alleviate pain, and improve joint function.

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.