IL33 Antibody

Interleukin-33, Mouse Anti Human
Shipped with Ice Packs
In Stock

Product Specs

Introduction
Interleukin 33 (IL-33) is a 32kDa proinflammatory cytokine that may also regulate gene transcription in producer cells. It is structurally similar to IL-1, inducing helper T cells to produce type 2 cytokines. IL-33 functions by binding to the IL1RL-1 receptor (also known as ST2), which activates NF-kappa-B and MAP kinases. This activation leads to the production of cytokines like IL-4, IL-5, and IL-13 by Th2 cells in vitro. In vivo, IL-33 can cause significant pathological changes in mucosal organs. Notably, caspase-1 can cleave IL-33 into a 12kDa N-terminal fragment and an 18kDa C-terminal fragment.
Physical Appearance
Clear, colorless solution, sterile-filtered.
Formulation
Supplied as a 1 mg/ml solution in PBS, pH 7.4, containing 0.1% sodium azide.
Storage Procedures
Store at 4°C for up to 1 month. For long-term storage, store at -20°C. Avoid repeated freeze-thaw cycles.
Stability / Shelf Life
Stable for 12 months at -20°C. Stable for 1 month at 4°C.
Applications
This IL33 antibody has been validated for ELISA and Western blot applications, demonstrating high specificity and reactivity. However, optimal working dilutions should be determined empirically for each application. For Western blot analysis, a dilution range of 1:500 to 1:1000 is recommended, with 1:500 as a starting point.
Synonyms
Interleukin 33, DVS27, NF-HEV, NKHEV, C9orf26, Interleukin-1 family member 11, IL- 1F11, Nuclear factor from high endothelial venules, NFEHEV, DKFZp586H0523, RP11-575C20.2, IL-33.
Purification Method
IL33 antibody was purified from mouse ascitic fluids by protein-G affinity chromatography.
Type
Mouse Anti Human Monoclonal.
Clone
P4E9AT.
Immunogen
Anti-human IL33 mAb, is derived from hybridization of mouse F0 myeloma cells with spleen cells from BALB/c mice immunized with Recombinant human IL33 amino acids 112-270 purified from E. coli.
Ig Subclass
Mouse IgG2b heavy chain and κ light chain.

Q&A

How does IL-33 signaling work at the molecular level?

IL-33 signaling operates through two distinct pathways. The reduced form of IL-33 (IL-33red) signals through the membrane-bound suppression of tumorigenicity 2 (ST2) receptor expressed on various immune cells, including mast cells and type 2 innate lymphoid cells . Following oxidation and disulfide bond formation, oxidized IL-33 (IL-33ox) signals via the receptor for advanced glycation end products/epidermal growth factor receptor (RAGE/EGFR) complex . These different signaling pathways lead to varied cellular responses: ST2-dependent pathways typically drive inflammatory responses, while the RAGE/EGFR pathway can influence epithelial cell migration and repair . Understanding these dual signaling mechanisms is crucial for developing targeted antibody therapeutics.

What applications are IL-33 antibodies commonly used for in research settings?

IL-33 antibodies are routinely employed in multiple laboratory applications including Western Blot (WB), immunohistochemistry (IHC), immunofluorescence (IF), and enzyme-linked immunosorbent assay (ELISA) . The typical working dilutions vary by application, with Western Blot generally requiring 1:1000-1:4000 dilution . These antibodies are most commonly used to detect IL-33 expression in human and mouse samples, helping researchers investigate IL-33's role in various physiological and pathological processes. Antibodies with different specificities may target distinct epitopes on IL-33, making careful selection essential for specific research applications .

How can IL-33 antibodies be used to differentiate between IL-33red and IL-33ox signaling pathways?

Distinguishing between IL-33red and IL-33ox signaling requires antibodies with specific binding profiles. High-affinity IL-33-neutralizing monoclonal antibodies like Tozorakimab can inhibit both IL-33red and IL-33ox activities, while other antibodies may selectively target one pathway . Methodologically, researchers can employ competition assays to evaluate binding site overlap between antibodies and determine their epitope specificity . To assess antibody effects on IL-33red signaling, functional assays measuring ST2-dependent inflammation (such as IFNγ production in human peripheral blood mononuclear cells) can be utilized. For IL-33ox pathway inhibition, epithelial cell scratch repair assays with A549 cells provide informative data . Importantly, not all anti-IL-33 antibodies inhibit both pathways equally, necessitating comprehensive characterization of each antibody's pharmacological profile .

What role does IL-33 play in breaking immune tolerance and autoantibody generation?

IL-33 contributes to autoimmunity by promoting B cell activation and autoantibody production through the induction of B cell activating factor (BAFF). Experimental evidence shows that even temporary, short-term increases in IL-33 can result in a primary IgM response to self-antigens, particularly DNA . This transient DNA-specific autoantibody response is dependent on BAFF induction, with radiation-resistant cells serving as the major source of BAFF rather than myeloid cells . Chronic exposure to IL-33 leads to dramatic increases in BAFF levels, elevated numbers of B and T follicular helper cells, germinal center formation, and ultimately class-switching from IgM to IgG autoantibodies . These findings suggest IL-33 may be a key factor in breaking immune tolerance and initiating autoimmune responses, potentially explaining its association with various autoimmune diseases.

How do different anti-IL-33 antibodies compare in their pharmacological profiles?

Anti-IL-33 antibodies exhibit diverse pharmacological profiles despite targeting the same cytokine. Research comparing several high-affinity IL-33-neutralizing monoclonal antibodies (including Tozorakimab from AstraZeneca, APE4909 from AnaptysBio, 9675P from Regeneron, Ab43 from Lilly, and IL-33158LS from Pfizer) revealed that while all had high sub/low-picomolar affinity for IL-33red and potently inhibited ST2-dependent inflammation, they differed significantly in their ability to inhibit IL-33ox-dependent functions . These differences likely arise from variations in epitope binding, which affects the antibody's capacity to block interaction with either ST2 or the RAGE/EGFR complex. When designing experiments or therapeutic approaches targeting IL-33, researchers should carefully select antibodies based on whether blocking the IL-33red pathway, IL-33ox pathway, or both is desired for the specific application .

What are the optimal conditions for storing and using IL-33 antibodies in laboratory settings?

For optimal preservation of IL-33 antibody activity, storage at -20°C is recommended, where the antibody remains stable for one year after shipment. Most commercial IL-33 antibodies are supplied in PBS with 0.02% sodium azide and 50% glycerol at pH 7.3 . Aliquoting is generally unnecessary for -20°C storage, particularly for smaller volumes (20μl) that often contain 0.1% BSA as a stabilizer . When using IL-33 antibodies in experiments, it's essential to titrate the reagent in each testing system to obtain optimal results, as the effective concentration can be sample-dependent . For Western blot applications, typical dilutions range from 1:1000 to 1:4000, but these may vary based on the specific antibody and experimental conditions . Always verify the antibody's reactivity with your species of interest, as some antibodies (like 66235-1-Ig) show confirmed reactivity with human samples but may also work with mouse samples based on cited literature .

How can researchers validate IL-33 antibody specificity and performance in their experimental systems?

Validating IL-33 antibody specificity requires a multi-faceted approach. First, researchers should perform positive control experiments with cells or tissues known to express IL-33, such as THP-1 cells, hTERT-RPE1 cells, or human lung tissue . Negative controls should include IL-33 knockout samples or cells where IL-33 expression has been silenced. Western blot analysis should confirm detection at the expected molecular weight (calculated 30 kDa, but often observed at 35 kDa for IL-33) . For functional validation, neutralization assays measuring the antibody's ability to block IL-33-induced responses in cell-based systems are essential. These can include measuring inhibition of cytokine production in peripheral blood mononuclear cells or epithelial cell migration in scratch assays . Competition assays with known IL-33 binders (such as soluble ST2) can further confirm specificity. Lastly, cross-reactivity testing against related IL-1 family members can ensure the antibody is truly specific for IL-33.

What controls should be included when using IL-33 antibodies in animal models of disease?

When designing experiments with IL-33 antibodies in animal models, several controls are critical. First, include isotype control antibodies to account for non-specific effects of antibody administration. Second, incorporate both wild-type and gene-deficient animals (IL-33-/-, ST2-/-, or double knockout) to distinguish IL-33-specific effects from potential compensatory mechanisms . Third, consider strain differences, as research has shown that C57BL/6 substrains may differ genetically and phenotypically, potentially affecting experimental outcomes . A sensitivity analysis should be performed to determine the statistical power of the experimental design in detecting meaningful differences between treatment groups . Additionally, when studying chronic conditions, consider the possibility that other alarmins like TSLP or IL-25 may compensate for the lack of IL-33 signaling . Finally, careful monitoring of BAFF levels can provide insight into IL-33-induced autoimmune responses, as BAFF plays a crucial role in IL-33-mediated autoantibody generation .

How can researchers reconcile conflicting data on IL-33's role in disease models?

Resolving discrepancies in IL-33 research requires thorough methodological analysis. First, consider mouse strain differences—results obtained in C57BL/6 mice may differ from those in BALB/c mice, and even C57BL/6 substrains can exhibit phenotypic variations . Genetic drift is another potential source of discrepancy, particularly in small mouse colonies; backcrossing to the inbred control strain every 5-10 generations is recommended to maintain genetic similarity . Sample size and statistical power are critical—many studies lack sufficient numbers to reliably detect small to moderate group differences, as revealed by sensitivity analyses . Additionally, compensatory mechanisms involving other cytokines (like TSLP or IL-25) may mask IL-33 effects in some models . Different antibody epitope specificities can also lead to contradictory findings, as not all anti-IL-33 antibodies inhibit both IL-33red and IL-33ox pathways equally . When interpreting conflicting data, researchers should comprehensively evaluate these factors and consider integrating multiple readouts (cytokine profiles, histology, functional assays) beyond single measurements like ear thickness in dermatitis models .

What considerations are important when analyzing IL-33 expression patterns in different tissues?

When analyzing IL-33 expression across tissues, several methodological considerations are essential. First, recognize that IL-33 is constitutively expressed in epithelial barrier tissues and human blood vessels as a nuclear alarmin, but expression patterns can change dramatically during inflammation or injury . For immunohistochemistry or immunofluorescence applications, appropriate antigen retrieval methods are crucial, as nuclear localization of IL-33 may require specific protocols. Consider dual staining with cell-type-specific markers to identify IL-33-expressing populations accurately. For Western blot analysis, be aware that the observed molecular weight (35 kDa) may differ from the calculated weight (30 kDa) . Different isoforms or post-translational modifications of IL-33 may be present in various tissues, affecting antibody recognition. IL-33 is rapidly released following tissue damage, so sample collection methods and timing can significantly impact results—quick tissue processing helps preserve the native state of IL-33. Finally, when comparing expression levels across studies, consider that different antibodies may have varying sensitivities or recognize different epitopes of IL-33, potentially leading to inconsistent results.

How should researchers interpret the dual roles of IL-33 in inflammation and tissue repair?

The interpretation of IL-33's seemingly contradictory roles in promoting both inflammation and tissue repair requires understanding its distinct signaling pathways. IL-33 exists in both reduced (IL-33red) and oxidized (IL-33ox) forms, which signal through different receptor complexes—ST2 for IL-33red and RAGE/EGFR for IL-33ox . The ST2-dependent pathway typically drives inflammatory responses via immune cell activation, while the RAGE/EGFR pathway influences epithelial cell migration and repair . Importantly, these pathways may be activated sequentially during the course of tissue injury and healing, with the inflammatory response preceding repair mechanisms. When analyzing experimental data, researchers should consider which form of IL-33 predominates under their specific conditions and which downstream pathways are engaged. Antibodies that selectively block one pathway while sparing the other can help dissect these distinct functions . Additionally, the timing of IL-33 release and signaling is critical—acute, transient signaling may lead to different outcomes than chronic IL-33 exposure, which has been linked to autoimmunity through BAFF induction and class-switched autoantibody production .

What are the emerging therapeutic applications of IL-33 antibodies beyond allergic inflammation?

While IL-33 antibodies were initially developed targeting allergic and type 2 inflammatory conditions, emerging research suggests broader therapeutic potential. IL-33's role in autoimmunity presents an opportunity for antibody therapeutics that target the IL-33-BAFF axis to potentially treat autoimmune diseases . The discovery that IL-33 contributes to breaking immune tolerance by promoting class-switching from IgM to IgG autoantibody responses suggests applications in conditions like systemic lupus erythematosus or rheumatoid arthritis . Additionally, the dual signaling pathways of IL-33 (via ST2 and RAGE/EGFR) open possibilities for tissue-specific therapeutic approaches . For instance, antibodies that selectively inhibit the inflammatory ST2 pathway while preserving the tissue-repair functions of the RAGE/EGFR pathway could offer advantages in certain inflammatory conditions. Conversely, in contexts where epithelial remodeling is detrimental (such as tumor microenvironments), targeting both pathways might be beneficial. Future therapeutic development will likely focus on creating antibodies with selective inhibitory profiles tailored to specific disease pathologies.

How might next-generation IL-33 antibodies improve upon current research tools?

Next-generation IL-33 antibodies will likely incorporate several advancements over current research tools. First, improved epitope mapping and rational design could create antibodies with more selective inhibition profiles for either IL-33red or IL-33ox pathways, enabling more precise experimental manipulation . Development of bispecific antibodies that simultaneously target IL-33 and complementary pathways (like TSLP or IL-25) could address compensatory mechanisms observed in some disease models . Additionally, antibodies with enhanced tissue penetration properties would improve efficacy in targeting IL-33 at specific anatomical sites. Novel formats like single-domain antibodies or nanobodies might offer advantages in certain applications due to their smaller size and unique binding properties. Conjugated antibodies carrying reporter molecules or therapeutic payloads could enhance visualization of IL-33 expression or deliver targeted interventions. Finally, humanized versions of research antibodies would facilitate translation from preclinical models to clinical applications, bridging the gap between basic research and therapeutic development.

What methodological approaches might help resolve the current contradictions in IL-33 research findings?

Addressing contradictions in IL-33 research requires innovative methodological approaches. First, standardized experimental systems with careful genetic background control and sufficient statistical power are needed to minimize variability . Multi-laboratory validation studies using identical protocols could identify sources of discrepancy. Single-cell analysis techniques would provide higher resolution data on IL-33-responsive cell populations, potentially revealing heterogeneity masked in bulk analyses. Temporal studies tracking the kinetics of IL-33 release, oxidation state changes, and signaling activation could clarify the dynamic nature of IL-33 biology. Combining in vivo models with ex vivo organoid systems might bridge the gap between simplified in vitro assays and complex animal models. Development of biosensors that can distinguish between IL-33red and IL-33ox in real-time would transform our understanding of IL-33 biology in situ. Finally, comprehensive genetic approaches generating double- or triple-deficient mutants lacking IL-33 along with other alarmins like TSLP or IL-25 would address potential compensatory mechanisms . These methodological innovations would help reconcile conflicting findings and establish a more unified understanding of IL-33 biology.

Product Science Overview

Introduction to Interleukin-33 (IL-33)

Interleukin-33 (IL-33) is a cytokine belonging to the IL-1 family, which plays a crucial role in the immune system by acting as an alarm signal (alarmin) released upon cellular damage or stress. It is encoded by the IL-33 gene located on chromosome 9 in humans and chromosome 19 in mice . IL-33 is primarily expressed in the nuclei of endothelial and epithelial cells and is involved in various biological processes, including inflammation, immune response, and tissue homeostasis .

Biological Functions of IL-33

IL-33 exerts its effects by binding to the ST2 receptor (suppression of tumorigenicity 2), which is expressed on various immune cells such as T-helper 2 (Th2) cells, mast cells, eosinophils, and innate lymphoid cells . The IL-33/ST2 axis is pivotal in mediating immune responses, particularly in allergic and inflammatory diseases. IL-33 has been implicated in the pathogenesis of conditions such as asthma, chronic obstructive pulmonary disease (COPD), and age-related macular degeneration (AMD) .

Mouse Anti-Human IL-33 Antibodies

Mouse anti-human IL-33 antibodies are monoclonal antibodies developed to specifically target and neutralize human IL-33. These antibodies are produced in hybridomas and purified through affinity chromatography . They are widely used in research and clinical settings to study the role of IL-33 in various diseases and to develop potential therapeutic interventions.

Applications in Research and Medicine
  1. Biomarker Assays: IL-33 is a valuable biomarker for several inflammatory diseases. Sensitive and specific assays, such as enzyme-linked immunosorbent assays (ELISA) and immuno-polymerase chain reaction (iPCR) assays, have been developed to detect IL-33 levels in various human matrices .
  2. Therapeutic Target: Given its role in inflammatory diseases, IL-33 is considered a potential target for biological therapies. Neutralizing antibodies against IL-33 are being explored for their therapeutic potential in conditions like COPD and asthma .
  3. Neurological Research: Recent studies have shown that exogenous IL-33 can enhance lymphatic drainage in the brain and improve motor and memory skills in traumatic brain injury (TBI) models .

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.