IL 1 Beta Antibody

Interleukin-1b, Mouse Anti-Human
Shipped with Ice Packs
In Stock

Description

Structure and Function of IL-1β

IL-1β is synthesized as a 31 kDa inactive pro-form (pro-IL-1β) in monocytes, macrophages, and dendritic cells . Proteolytic cleavage by caspase-1, activated via inflammasomes, converts it into a 17 kDa active form . Unlike classical cytokines, IL-1β lacks a signal peptide and is secreted via non-canonical pathways alongside caspase-1 . It binds to receptors IL-1RI and IL-1RII, triggering downstream inflammatory cascades .

Mechanism of IL-1 Beta Antibodies

IL-1β antibodies block the cytokine’s interaction with its receptors or neutralize its active form. Key mechanisms include:

  • Direct neutralization: Antibodies like canakinumab bind IL-1β, preventing receptor activation .

  • Inflammasome modulation: Some antibodies indirectly reduce IL-1β maturation by targeting upstream pathways .

  • Pro-form targeting: Antibodies such as MAB6964 detect pro-IL-1β, enabling research into its regulation .

Autoimmune and Inflammatory Diseases

  • Rheumatoid arthritis: IL-1β antibodies reduce joint inflammation and cartilage degradation .

  • Cryopyrin-associated periodic syndromes (CAPS): Canakinumab (Ilaris®) is FDA-approved for CAPS, suppressing systemic inflammation .

Hematological Malignancies

  • Leukemia and myeloma: IL-1β blockade inhibits leukemia stem cell (LSC) proliferation and enhances tyrosine kinase inhibitor efficacy .

  • Graft-versus-host disease (GVHD): Neutralizing IL-1β ameliorates intestinal mucositis and GVHD severity in preclinical models .

Metabolic Disorders

  • Type 2 diabetes: Anti-IL-1β antibodies improve glycemic control by reducing pancreatic β-cell stress and systemic inflammation .

Key Research Findings

Study FocusModel/DesignOutcomeSource
Melanoma metastasisWT/DJ-1 KO miceAnti-IL-1β reduced lung nodules in KO mice but increased them in WT mice
Glycemic controlObese mice (13-week treatmentHbA1c reduced by 0.45%; proinsulin levels decreased by 56%
Engineered pro-CanakinumabMMP-9-activated antibodyEnhanced tumor suppression and reduced systemic toxicity

Clinical Trials and Outcomes

  • CANOPY-1 (NSCLC): Phase III trial of canakinumab + chemotherapy showed no survival benefit but supported further evaluation in adjuvant settings .

  • CANTOS trial: IL-1β inhibition reduced lung cancer incidence by 67% in high-risk cardiovascular patients .

Future Directions

  • Combination therapies: Pairing IL-1β antibodies with checkpoint inhibitors or chemotherapy .

  • Precision targeting: Antibody-drug conjugates for hematological malignancies .

Product Specs

Introduction
Interleukin-1 beta (IL-1β) is a cytokine produced by activated macrophages that plays a crucial role in inflammatory responses. IL-1β stimulates thymocyte proliferation by inducing IL-2 release, promotes B-cell maturation and proliferation, and enhances fibroblast growth factor activity. As an endogenous pyrogen, IL-1β contributes to fever and inflammation. It also stimulates the release of prostaglandin and collagenase from synovial cells.
Formulation
The antibody is supplied as a solution containing 1 mg/ml in phosphate-buffered saline (PBS) after reconstitution.
Shipping Conditions
The antibody is shipped in a lyophilized form at ambient temperature.
Storage Procedures
For long-term storage, keep the lyophilized antibody at 4°C in a dry environment. After reconstitution, the antibody can be stored at 4°C for up to one month. For longer storage, aliquot the antibody and store at -20°C.
Solubility
To reconstitute, add sterile water (H2O) to the vial. Mix gently by swirling and allow 30-60 seconds for the antibody to dissolve completely. Avoid creating foam.
Applications
This antibody is suitable for various applications, including direct enzyme-linked immunosorbent assay (ELISA), Western blotting, immunoprecipitation, and immunohistochemistry.
Note
This antibody was generated by immunizing BALB/c mice.
Titer
The antibody exhibits high titer as determined by direct ELISA. A 1:20,000 dilution yields an optical density (O.D.) greater than 1.0 when using alkaline phosphatase-conjugated rabbit anti-mouse immunoglobulin G (IgG) from Jackson Laboratories. Furthermore, a 1:2,000 dilution completely inhibits a thymocyte co-stimulation assay using suboptimal concentrations of concanavalin A.
Synonyms
Catabolin, Lymphocyte-activating factor (LAF), Endogenous Pyrogen (EP), Leukocyte Endogenous Mediator (LEM), Mononuclear Cell Factor (MCF), IL1F2, IL-1 beta.
Purification Method
Ion Exchange.
Type
Mouse Anti Human Monoclonal.
Clone
NYR-hIL1b.
Immunogen
r.Human IL-1b.
Ig Subclass
Mouse IgG2b.

Q&A

What are the primary applications for IL-1 beta antibodies in immunological research?

IL-1 beta antibodies are utilized across multiple research applications including Western blotting, ELISA, immunohistochemistry (IHC), immunoprecipitation (IP), flow cytometry, and immunofluorescence microscopy. Most commercially available IL-1 beta antibodies, such as ABIN964782, are validated for these specific applications, allowing researchers to detect the mature 17 kDa form of IL-1 beta in various experimental systems . The application versatility makes these antibodies essential tools for studying inflammation, cytokine signaling, and immune responses in different pathological contexts. When designing experiments, researchers should consider that application-specific optimizations may be required, particularly for techniques like flow cytometry where non-specific interactions with the Fc domain of rabbit IgG molecules can occur, necessitating appropriate controls .

How do polyclonal and monoclonal IL-1 beta antibodies differ in research applications?

Polyclonal IL-1 beta antibodies, such as the rabbit anti-mouse IL-1 beta (ABIN964782), recognize multiple epitopes on the target protein, offering high sensitivity but potential variability between lots . These antibodies are often preferred for applications requiring robust signal detection like Western blotting and immunohistochemistry. Monoclonal antibodies, such as MAB601, recognize a single epitope and provide higher specificity and consistency between experiments . This makes monoclonal antibodies particularly valuable for applications requiring precise quantification, such as sandwich ELISAs or neutralization assays where defined epitope targeting is critical. For comprehensive experimental design, researchers often employ both antibody types: monoclonals for capture in sandwich assays and polyclonals for detection to maximize both specificity and sensitivity .

How can researchers verify the specificity of IL-1 beta antibodies before experimental use?

Verification of IL-1 beta antibody specificity should follow a multi-step approach:

  • Positive and negative control samples: Use stimulated cells known to express IL-1 beta (e.g., LPS-treated THP-1 cells or PBMCs) alongside unstimulated controls .

  • Blocking peptide validation: Pre-incubate the antibody with recombinant IL-1 beta protein to confirm signal elimination in Western blots or immunostaining.

  • Cross-reactivity assessment: Test against related family members (e.g., IL-1α) or IL-1 beta from different species when cross-species reactivity is a concern .

  • Molecular weight verification: Confirm detection of appropriate bands (~17 kDa for mature IL-1 beta, ~31-36 kDa for pro-IL-1 beta) in Western blot applications .

  • Knockout/knockdown validation: When available, use IL-1 beta knockout cells or tissues as definitive negative controls.

These validation steps are particularly important when studying complex inflammatory conditions where multiple cytokines may be present, requiring confident differentiation between IL-1 family members .

What methodological approaches can distinguish between pro-IL-1 beta and mature IL-1 beta in biological samples?

Distinguishing between pro-IL-1 beta (~31-36 kDa) and mature IL-1 beta (~17 kDa) requires strategic antibody selection and experimental techniques:

  • Western blotting with reducing conditions: Using antibodies like MAB601 under reducing conditions allows separation by molecular weight, with pro-IL-1 beta appearing at approximately 36 kDa and mature IL-1 beta at 17 kDa . Sample preparation should include protease inhibitors to prevent artificial processing.

  • Domain-specific antibodies: Utilize antibodies targeting either the pro-domain (MAB6964 targeting Met1-Asp116) or mature domain exclusively . This approach enables selective detection of specific forms.

  • Subcellular fractionation: Since pro-IL-1 beta is primarily intracellular while mature IL-1 beta may be secreted, separating cellular and supernatant fractions before analysis can provide form-specific information.

  • Two-color immunofluorescence: Employ antibodies targeting different domains with distinct fluorophores to visualize both forms simultaneously in cellular contexts.

  • Mass spectrometry validation: For definitive identification, processed samples can be analyzed by mass spectrometry to distinguish between forms based on peptide sequences.

These methods have been crucial in studies investigating inflammasome activation and IL-1 beta processing, providing insights into the regulation of inflammatory responses .

Quantification of IL-1 beta antibody neutralization potency requires systematic bioassay approaches:

  • Proliferation-based bioassays: Using responsive cell lines like D10.G4.1 mouse helper T cells, researchers can measure proliferation induced by recombinant IL-1 beta and its inhibition by neutralizing antibodies . The neutralization dose 50 (ND50), typically 0.05-0.2 μg/mL for antibodies like MAB601, provides a standardized measure of potency .

  • Comparative dose-response analysis: Novel antibodies can be directly compared to established therapeutics like canakinumab using parallel dose-response curves. As demonstrated with the P2D7KK antibody, this approach revealed a neutralization potency more than 10 times higher than the marketed antibody canakinumab .

  • Receptor binding competition assays: Surface plasmon resonance or competitive ELISAs can determine the ability of antibodies to prevent IL-1 beta from binding to its receptors (IL-1R1/IL-1RAcP), providing mechanism-of-action insights.

  • In vivo neutralization models: Animal disease models associated with IL-1β pathology provide functional validation of neutralization efficacy in complex biological systems .

  • Affinity maturation assessment: Engineered antibodies can demonstrate >30-fold increased affinity compared to parent antibodies, as seen with novel therapeutic candidates, providing a quantitative measure of binding improvement .

These quantitative approaches are essential for developing next-generation IL-1 beta targeting therapeutics with improved efficacy profiles.

What are the optimal stimulation conditions for IL-1 beta production in different cell types for antibody validation?

Optimal stimulation conditions for IL-1 beta production vary by cell type and should be carefully selected for rigorous antibody validation:

Cell TypeStimulation ConditionsIncubation TimeExpected IL-1β DetectionReference
THP-1 (human monocytic)200 nM PMA (24h) + 10 μg/mL LPS (3-24h)24-48 hoursWestern blot: ~36 kDa band
PBMCs1% serum + 10 ng/mL E. coli LPS24 hoursWestern blot: 17 kDa from supernatants
Whole blood cultures1.0 μg/ml LPS18 hoursELISA: secreted IL-1β in plasma

When validating IL-1 beta antibodies, researchers should include both time-course and dose-response analyses to identify optimal detection windows. For quantitative techniques like ELISA or flow cytometry, protein transport inhibitors (e.g., Brefeldin A) may be necessary to prevent secretion and enable intracellular detection. Additionally, consideration of inflammasome activators (e.g., ATP, nigericin) as secondary signals may be required for optimal mature IL-1 beta production in certain experimental systems .

What controls are essential when using IL-1 beta antibodies in multiplex immunoassays?

When incorporating IL-1 beta antibodies into multiplex immunoassays, the following controls are essential:

  • Isotype controls: Include species-matched, non-targeting IgG controls (e.g., rabbit IgG for rabbit-derived IL-1 beta antibodies) to assess non-specific binding .

  • Pre-blocking validations: Pre-incubate IL-1 beta antibodies with recombinant IL-1 beta to confirm signal elimination in one channel while maintaining other analyte signals.

  • Single-analyte standards: Run recombinant IL-1 beta standards alone to confirm assay specificity before multiplex analysis.

  • Cross-reactivity assessment: Evaluate potential cross-reactivity with other cytokines included in the multiplex panel, particularly IL-1 family members.

  • Sample dilution series: Perform serial dilutions of positive control samples to confirm linearity of IL-1 beta detection in the multiplex format.

  • Spike-recovery validation: Add known quantities of recombinant IL-1 beta to negative samples to verify accurate quantification in complex matrices.

For immunoprecipitation experiments specifically, pre-clearing samples with non-specific rabbit IgG is recommended to reduce background interference when using rabbit-derived IL-1 beta antibodies . These controls ensure reliable differentiation between true positive signals and artifacts in complex cytokine analysis systems.

How should researchers design experiments to compare the affinity and specificity of different IL-1 beta antibodies?

Designing experiments to compare IL-1 beta antibodies requires systematic approaches addressing multiple parameters:

  • Surface plasmon resonance (SPR) analysis:

    • Measure association (kon) and dissociation (koff) rates

    • Calculate equilibrium dissociation constants (KD)

    • Compare antibodies under identical conditions using the same recombinant IL-1 beta preparation

  • Comparative Western blot analysis:

    • Use identical sample preparation, loading, and transfer conditions

    • Test antibodies at normalized concentrations (e.g., molar equivalents)

    • Include gradient dilutions to determine detection limits for each antibody

    • Evaluate band specificity patterns across multiple cell types/tissues

  • Competitive binding assays:

    • Assess displacement of labeled IL-1 beta by unlabeled antibodies

    • Determine IC50 values for comparative binding strength

    • Map epitope binding regions through competition assays

  • Cross-reactivity profiling:

    • Test against IL-1 family members (IL-1α, IL-1Ra)

    • Evaluate species cross-reactivity with recombinant IL-1 beta from human, mouse, rat, and non-human primates

    • Document cross-reactivity percentages for standardized comparison

  • Functional neutralization comparison:

    • Determine neutralization dose-response curves

    • Calculate ND50 values in standardized bioassays

    • Compare neutralization efficiency in different cellular contexts

These systematic comparisons facilitated the development of engineered antibodies like P2D7KK, which demonstrated >30-fold increased affinity compared to parent antibodies and superior neutralization potency compared to marketed therapeutics .

Why might Western blots using IL-1 beta antibodies show unexpected band patterns?

Unexpected band patterns in IL-1 beta Western blots can result from several factors that researchers should systematically troubleshoot:

  • IL-1 beta processing states: The detection of multiple bands may represent different processing states of IL-1 beta, including the ~31-36 kDa pro-form and the ~17 kDa mature form . This is particularly common in stimulated immune cells like THP-1 or PBMCs, where both forms may be present simultaneously.

  • Non-specific binding: Background bands may represent cross-reactivity with other IL-1 family members or unrelated proteins. Validation with blocking peptides or IL-1 beta-deficient samples can help identify true IL-1 beta-specific signals.

  • Sample preparation issues: Inadequate denaturing conditions or protein degradation during sample preparation can generate artifactual bands. Ensure complete sample denaturation and use fresh protease inhibitors during lysis.

  • Post-translational modifications: Glycosylation, phosphorylation, or ubiquitination can alter the apparent molecular weight of IL-1 beta in certain cell types or disease states, resulting in band shifts.

  • Antibody specificity limitations: Some antibodies may recognize specific epitopes exposed only under certain conditions. For instance, the antibody described in source specifically detects the mature 17 kDa form, while others may detect both precursor and mature forms.

For optimal Western blot results, researchers should use positive controls from LPS-stimulated cells alongside unstimulated controls, and consider detecting the IL-1 beta precursor from cell lysates and mature IL-1 beta from culture supernatants separately .

How can researchers address non-specific binding issues with IL-1 beta antibodies in flow cytometry?

Non-specific binding in flow cytometry with IL-1 beta antibodies can be addressed through several methodological approaches:

  • Fc receptor blocking: Pre-incubate cells with species-appropriate Fc receptor blocking reagents to prevent non-specific binding through Fc interactions, which is particularly important when using rabbit IgG-derived IL-1 beta antibodies .

  • Optimized fixation protocols: Compare different fixation methods (paraformaldehyde, methanol, commercial fixatives) to identify conditions that preserve IL-1 beta epitopes while maintaining cellular integrity. Overfixation can create artifactual binding sites.

  • Validated permeabilization: Test different permeabilization reagents (saponin, Triton X-100, commercial buffers) to ensure optimal intracellular antibody access while minimizing non-specific binding.

  • Titration optimization: Perform antibody titration experiments to identify the optimal concentration that maximizes specific signal while minimizing background. The optimal concentration may differ significantly from Western blot applications.

  • Stringent washing: Implement additional washing steps with buffers containing mild detergents to reduce non-specific interactions.

  • Secondary antibody controls: When using indirect detection methods, include controls with secondary antibody alone to identify background contributed by the detection system.

  • Isotype-matched negative controls: Include species and isotype-matched control antibodies at identical concentrations to establish appropriate gates and quantify non-specific binding .

For flow cytometry applications specifically, researchers should be aware that the F(c) domain of rabbit IgG molecules may interact with cells non-specifically, and appropriate controls should be included to account for this potential artifact .

Why might IL-1 beta neutralization assays show inconsistent results between in vitro and in vivo experiments?

IL-1 beta neutralization assays often show discrepancies between in vitro and in vivo results due to several biological and methodological factors:

  • Antibody pharmacokinetics and biodistribution: In vivo, antibodies are subject to distribution limitations, metabolism, and clearance not present in vitro. Antibody half-life and tissue penetration significantly impact in vivo efficacy but are not factors in controlled in vitro systems.

  • Complex cytokine networks: In vivo environments contain compensatory cytokine pathways that may overcome IL-1 beta neutralization. For example, TNF-α or IL-6 may provide redundant inflammatory signaling not present in simplified in vitro models.

  • Cellular sources and targets: While in vitro systems often use purified cell populations, in vivo models involve complex cellular interactions. The P2D7KK antibody demonstrated efficacy in mouse models of IL-1β pathology, but required consideration of these complex interactions .

  • Antibody efficacy across species: Inconsistencies may arise from different neutralization potencies against human versus animal IL-1 beta. Some antibodies like P2D7KK show advantageous cross-reactivity across experimental animals, facilitating consistent preclinical development .

  • Dosing and timing considerations: Optimal dosing regimens determined in vitro often require adjustment in vivo to account for pharmacokinetics and tissue-specific requirements.

  • Multiple IL-1 beta forms: In vivo, both membrane-associated and soluble IL-1 beta forms may be present, whereas in vitro assays typically focus on soluble forms only.

To address these discrepancies, researchers should implement dose-response studies in vivo, validate target engagement through biomarker analysis, and perform pharmacokinetic/pharmacodynamic modeling to optimize translation between systems .

How are IL-1 beta antibodies being utilized in studies of endothelial dysfunction and metabolic disease?

IL-1 beta antibodies are revealing critical mechanisms in endothelial dysfunction and metabolic disorders through innovative experimental approaches:

  • Endothelial-mesenchymal transition (EndMT) studies: IL-1 beta antibodies have enabled researchers to visualize how IL-1 beta exposure causes human aortic endothelial cells (HAECs) to transition from cobblestone morphology to spindle-shaped mesenchymal phenotypes. This transition involves decreased CD31 (endothelial marker) and increased FSP1 (fibroblast marker) expression, particularly pronounced under high glucose conditions .

  • Mechanistic investigations in diabetic vascular complications: Immunofluorescence studies using IL-1 beta antibodies have demonstrated synergistic effects between high glucose (30 mM) and IL-1 beta (10 ng/ml) in promoting endothelial dysfunction, revealing potential intervention targets for diabetic vascular complications .

  • Alpha-1 antitrypsin (A1AT) interaction studies: IL-1 beta antibodies in ELISA applications have allowed quantification of how endogenous A1AT modulates IL-1 beta release in whole blood cultures, providing insights into inflammatory regulation in metabolic conditions .

  • Intracellular signaling pathway investigation: Combined use of phospho-specific antibodies with IL-1 beta detection has revealed how IL-1 beta activates inflammatory signaling cascades in metabolically stressed tissues.

  • Therapeutic intervention assessment: Neutralizing IL-1 beta antibodies have been used to evaluate potential interventions for metabolic inflammation, with engineered antibodies showing promise in relevant disease models .

These applications have expanded our understanding of how IL-1 beta contributes to the pathogenesis of metabolic disorders and identified potential therapeutic strategies targeting this cytokine pathway .

What are the considerations for using IL-1 beta antibodies in studies of inflammasome activation?

Using IL-1 beta antibodies to study inflammasome activation requires specific methodological considerations:

  • Form-specific detection strategies: Select antibodies capable of distinguishing between pro-IL-1 beta (~31-36 kDa) and mature IL-1 beta (~17 kDa) to accurately assess inflammasome-mediated processing . Antibodies targeting the propeptide region (Met1-Asp116) are particularly valuable for tracking processing events .

  • Two-signal experimental design: Implement protocols that provide both Signal 1 (e.g., LPS for pro-IL-1 beta synthesis) and Signal 2 (e.g., ATP, nigericin for inflammasome activation) to accurately model the two-step process of IL-1 beta production and maturation.

  • Subcellular fractionation approaches: Separate cytosolic, membrane, and supernatant fractions to track IL-1 beta processing and secretion, as mature IL-1 beta released through gasdermin D pores or other unconventional secretion pathways may localize differently than pro-IL-1 beta.

  • Co-detection with inflammasome components: Combine IL-1 beta detection with visualization of inflammasome assembly (e.g., ASC specks, NLRP3 oligomerization) to correlate processing events with inflammasome formation.

  • Live-cell imaging considerations: For real-time studies, ensure antibody fragments or non-disruptive labeling approaches are used to avoid interfering with the dynamic process of inflammasome assembly.

  • Species-specific optimization: When studying inflammasome activation in mouse or rat models, select antibodies with validated cross-reactivity to ensure consistent detection across experimental systems .

These considerations have been valuable in studies examining the complex regulation of inflammasome-mediated IL-1 beta processing and secretion in inflammatory diseases and infection models .

How should researchers prepare samples for optimal IL-1 beta detection in different experimental systems?

Sample preparation protocols must be tailored to the specific biological system and detection method:

  • Cell culture systems:

    • For Western blotting: Harvest 2 × 10^6 endotoxin-stimulated peripheral blood mononuclear cells (PBMCs) stimulated with 1% serum plus 10 ng/mL E.coli LPS for 24 hours .

    • For immunofluorescence: Treat THP-1 cells with 200 nM PMA for 24 hours followed by 10 μg/mL LPS for 3-24 hours to induce robust IL-1 beta expression .

    • Separate supernatants (containing secreted mature IL-1 beta) from cell lysates (containing primarily pro-IL-1 beta) for comprehensive analysis.

  • Tissue samples:

    • For immunohistochemistry: Both paraffin fixation and cryofixation are suitable for IL-1 beta detection, though epitope retrieval may be necessary for formalin-fixed samples .

    • Include protease inhibitors during tissue homogenization to prevent artificial processing of pro-IL-1 beta.

    • Consider regional heterogeneity in IL-1 beta expression when sampling tissues.

  • Blood samples:

    • For ELISA: Use whole blood cultures stimulated with 1.0 μg/ml LPS for 18 hours to assess IL-1 beta release capacity .

    • For bioactivity assays: Collect serum or plasma with appropriate anticoagulants that don't interfere with downstream functional assays.

  • Storage considerations:

    • Minimize freeze-thaw cycles as they can affect IL-1 beta detection.

    • For long-term storage, maintain samples at -80°C rather than -20°C to preserve bioactivity.

These optimized preparation protocols ensure reliable detection of IL-1 beta across experimental systems and are critical for comparing results between different studies or disease models .

What are the best practices for antibody validation when studying both human and animal models of IL-1 beta-mediated inflammation?

Best practices for IL-1 beta antibody validation across species require a comprehensive approach:

  • Cross-reactivity documentation:

    • Systematically test antibodies against recombinant IL-1 beta proteins from all species of interest (human, mouse, rat, non-human primates).

    • Quantify cross-reactivity percentages for each species rather than relying on binary assessments .

    • Document detection sensitivities across species to account for potential differences in affinity.

  • Epitope conservation analysis:

    • Compare antibody epitope regions across species using sequence alignment.

    • Select antibodies targeting highly conserved regions when cross-species applications are planned.

    • The novel antibody P2D7KK demonstrates advantages in cross-reactivity that facilitates preclinical development across species .

  • Species-specific positive controls:

    • Generate positive controls from each species by stimulating appropriate primary cells (e.g., mouse bone marrow-derived macrophages, human PBMCs).

    • Include species-matched recombinant proteins as standards in quantitative assays.

  • Validation in multiple techniques:

    • Confirm cross-reactivity in multiple applications (e.g., Western blot, ELISA, IHC) as species cross-reactivity may differ between techniques.

    • Document application-specific dilutions for each species, as optimal concentrations may vary.

  • Functional validation:

    • For neutralizing antibodies, confirm bioactivity neutralization across species using appropriate bioassays.

    • Document ND50 values for each species to account for potential potency differences .

These validation practices are particularly important when developing therapeutic antibodies, where consistent cross-reactivity facilitates translational research from animal models to human applications .

By following these comprehensive validation approaches, researchers can ensure reliable and comparable data when studying IL-1 beta across different species models of inflammation.

Product Science Overview

Interleukin-1β (IL-1β)

Interleukin-1β (IL-1β) is a pro-inflammatory cytokine that plays a crucial role in the body’s immune response. It is produced primarily by activated monocytes and macrophages. IL-1β is involved in various cellular activities, including cell proliferation, differentiation, and apoptosis. It is a key mediator in the inflammatory response and is implicated in a variety of diseases, including autoimmune disorders, infectious diseases, and cancer.

IL-1β is synthesized as a 31 kDa precursor protein. To become active, this precursor must be cleaved by caspase-1 between Asp116 and Ala117, resulting in a 17 kDa mature form . The detection of this mature form is often used as an indicator of caspase-1 activity.

Mouse Anti-Human IL-1β Antibodies

Mouse anti-human IL-1β antibodies are monoclonal antibodies developed in mice that specifically target human IL-1β. These antibodies are widely used in research and clinical applications to study the role of IL-1β in various diseases and to develop therapeutic strategies.

Applications

Mouse anti-human IL-1β antibodies can be used in several scientific applications, including:

  • Western Blotting (WB): To detect IL-1β protein levels in cell or tissue extracts.
  • Enzyme-Linked Immunosorbent Assay (ELISA): To quantify IL-1β levels in biological samples.
  • Immunohistochemistry (IHC): To visualize IL-1β expression in tissue sections.
  • Immunocytochemistry (ICC): To detect IL-1β in cultured cells.
  • Flow Cytometry: To analyze IL-1β expression on the cell surface or within cells .
Specificity and Cross-Reactivity

Mouse anti-human IL-1β antibodies are designed to specifically bind to human IL-1β. They do not cross-react with IL-1β from other species, such as mouse, rat, rabbit, or macaques . This specificity is crucial for accurate detection and quantification of human IL-1β in various experimental settings.

Types of Mouse Anti-Human IL-1β Antibodies

There are different types of mouse anti-human IL-1β antibodies available, including:

  • Monoclonal Antibodies: These are produced by a single clone of B cells and are highly specific to a single epitope on the IL-1β molecule.
  • Polyclonal Antibodies: These are produced by multiple clones of B cells and can recognize multiple epitopes on the IL-1β molecule.
  • Recombinant Monoclonal Antibodies: These are genetically engineered antibodies that combine the specificity of monoclonal antibodies with the consistency and reproducibility of recombinant technology .

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.