IL36B Human

Interleukin-36 Beta Human Recombinant
Shipped with Ice Packs
In Stock

Description

Overview of IL36B Human

IL36B Human (Interleukin-36 beta) is a pro-inflammatory cytokine belonging to the interleukin-1 (IL-1) family, encoded by the IL36B gene located on chromosome 2 . It shares structural and functional similarities with IL-1 cytokines, including a 12-stranded β-trefoil fold . IL36B is actively secreted and binds to the IL-36 receptor (IL1RL2/IL-36R) to activate NF-κB and MAPK signaling pathways, driving inflammatory responses in epithelial barriers, immune cells, and neutrophils .

Biological Functions and Signaling Pathways

IL36B orchestrates innate and adaptive immune responses, particularly in epithelial and immune cells. Key functions include:

  • Pro-inflammatory signaling: Activates NF-κB and MAPK pathways via IL1RL2/IL-36R and IL1RAP coreceptor .

  • Neutrophil recruitment: Promotes CXCL1 and IL-6 production, driving neutrophil infiltration in lung and skin inflammation .

  • Synergy with other cytokines: Cooperates with IL-17A, TNF-α, and GM-CSF to amplify inflammation in psoriasis and colitis .

Functional Partners

PartnerRoleScore
IL1RL2Receptor for IL36B; forms signaling complex with IL1RAP 0.991
IL36RNAntagonist; inhibits IL36B signaling by blocking IL1RL2 0.989
IL1RAPCoreceptor; stabilizes IL1RL2-IL36B interaction 0.853
IL1F10Modulates IL36B-induced cytokine production (e.g., reduces IL8 in PBMCs) 0.868

Lung Inflammation

  • Neutrophilic response: Il36r knockout mice show reduced neutrophil influx and IL-1β/CXCL1 levels in CS-exposed or H1N1-infected lungs .

  • Therapeutic target: IL-36R blockade preserves IL-1-mediated innate immunity while mitigating tissue damage .

Skin and Autoimmune Diseases

  • Psoriasis: IL36B drives IL-17A/TNF-α production in keratinocytes, synergizing with IL-22 .

  • DITRA syndrome: Loss-of-function IL36RN mutations cause generalized pustular psoriasis; anti-IL-36R therapies show promise .

Inflammatory Bowel Disease (IBD)

  • Colonic inflammation: IL36B induces TNF-α and IL-17A in keratinocytes, sustaining a pro-inflammatory loop .

  • Fibrosis: IL-36R blockade reduces intestinal fibrosis in chronic colitis models .

Therapeutic Potential and Challenges

TargetMechanismApplication
IL-36R antagonistsBlock IL36B/IL1RL2 interactionPsoriasis, DITRA, IBD
IL-36B neutralizationReduce IL36B-driven neutrophil recruitmentCOPD, acute respiratory infections
IL1RAP inhibitionDisrupt IL-36/IL-1 signaling complexSystemic inflammatory diseases

Product Specs

Introduction
Human Interleukin-36 beta (IL-36b) is a cytokine belonging to the IL-1 family, which includes key mediators of inflammation and immunity. IL-36b shares structural similarities with other IL-1 family members, adopting a characteristic beta-trefoil configuration. It is secreted by various immune cells, such as monocytes and B cells, and its biological activity is mediated through the IL-1 receptor complex, specifically involving IL-1Rrp2 and IL-1RAcP. Upon binding to its receptor, IL-36b initiates signaling cascades that activate NF-kB and MAPK pathways, ultimately contributing to inflammatory responses.
Description
Recombinant human IL-36b is produced in E. coli and undergoes purification to obtain a single, non-glycosylated polypeptide chain. This protein comprises 157 amino acids with a molecular weight of 17.7 kDa. Purification is achieved using proprietary chromatographic techniques.
Physical Appearance
Sterile Filtered White lyophilized powder.
Formulation
The protein is lyophilized from a 0.2 µm filtered solution in phosphate-buffered saline (PBS) at a pH of 7.4.
Solubility
To reconstitute the lyophilized IL-36b, it is recommended to dissolve it in sterile 18 megaohm-cm (MΩ·cm) water to a concentration of at least 100 µg/ml. This solution can be further diluted in other aqueous solutions.
Stability
Lyophilized IL-36b remains stable at room temperature for up to three weeks. However, for long-term storage, it is recommended to store the lyophilized product desiccated at a temperature below -18°C. After reconstitution, the IL-36b solution should be stored at 4°C for no more than 7 days. For extended storage, freeze the reconstituted IL-36b below -18°C. Repeated freezing and thawing cycles should be avoided.
Purity
The purity of the IL-36b is determined to be greater than 95% as assessed by SDS-PAGE analysis.
Biological Activity
The biological activity of IL-36b is evaluated by its ability to bind to recombinant human IL-1 Rrp2 Fc Chimera using a functional ELISA assay.
Synonyms
Interleukin 36 beta, interleukin 1 family member 8 (eta), Interleukin-1 homolog 2, IL1F8 (Canonical product IL-1F8a), IL-1F8 (FIL1-eta), Interleukin-1 Superfamily e, IL1H2, MGC126880, MGC126882.
Source
Escherichia Coli.
Amino Acid Sequence
MNPQREAAPK SYAIRDSRQM VWVLSGNSLI AAPLSRSIKP VTLHLIACRD TEFSDKEKGN MVYLGIKGKD LCLFCAEIQG KPTLQLKEKN IMDLYVEKKA QKPFLFFHNK EGSTSVFQSV SYPGWFIATS TTSGQPIFLT KERGITNNTN FYLDSVE

Q&A

What is IL-36B and how does it relate to the IL-1 family?

IL-36B (IL-36 beta, formerly known as IL-1F8) is a member of the interleukin-1 (IL-1) superfamily of cytokines. It is one of three IL-36 agonists, alongside IL-36α and IL-36γ, that were previously designated as IL-1F6, IL-1F8, and IL-1F9 before the terminology was unified in 2010. The IL-36 family also includes an antagonist called IL-36Ra (formerly IL-1F5). The genes encoding IL-36 family proteins are located on human chromosome 2, with IL-36Ra specifically encoded by the gene IL-36RN. IL-36B shares approximately 52% homologous amino acid sequence with IL-1Ra, reflecting their evolutionary relationship within the broader IL-1 family .

What signaling pathways does IL-36B activate?

IL-36B, like other IL-36 agonists, binds to heterodimeric receptor complexes composed of IL-36R and IL-1 receptor accessory protein (IL-1RAcP). Upon binding, these form heterotrimer complexes that signal through intracellular functional domains. This interaction activates the adaptor protein myeloid differentiated protein 88 (MyD88), followed by downstream activation of mitogen-activated protein kinase (MAPK) and nuclear factor-kappa B (NF-κB) signaling pathways. This signaling cascade ultimately leads to the regulation of target genes involved in inflammatory responses .

In human keratinocytes, IL-36 stimulation induces IκBζ expression, which is required for the expression of genes involved in inflammatory signaling, neutrophil chemotaxis, and leukocyte activation. In human endothelial cells, IL-36γ stimulation promotes the generation of IL-8, CCL2, CCL20, and upregulates adhesion molecules like VCAM-1 and ICAM-1 .

What are the primary biological functions of IL-36B in immune responses?

IL-36B plays important roles in both innate and adaptive immune responses:

Innate immunity roles:

  • Activates human monocytes, significantly upregulating expression of IL-1α, IL-1β, and IL-6

  • Enhances dendritic cell (DC) activation, upregulating markers such as CD80, CD86, and MHCII

  • Induces production of IL-6 and IL-12 in murine DCs

  • Elevates levels of IL-12p70, IL-23, and IL-10 in murine MDDCs

  • Contributes to neutrophil recruitment and CXCL1 generation

Adaptive immunity roles:

  • Influences T cell responses

  • Involved in antigen presentation processes

  • Contributes to pro-inflammatory factor production

Through these mechanisms, IL-36B contributes significantly to inflammatory cascades relevant to various disease states.

What are the optimal methods for detecting and quantifying IL-36B in human samples?

For accurate detection and quantification of IL-36B in human samples, ELISA (Enzyme-Linked Immunosorbent Assay) is the gold standard method. When selecting an ELISA kit, researchers should consider:

Sample types compatibility:

  • Human serum

  • Plasma

  • Cell culture supernatants

Performance metrics to evaluate:

  • Sensitivity (e.g., 18.75 pg/mL)

  • Detection range (e.g., 31.25-2000 pg/mL)

  • Specificity for IL-36B without cross-reactivity to other IL-36 family members

For human IL-36B research, sandwich ELISA formats typically offer superior sensitivity and specificity compared to competitive ELISAs. Additional detection methods include Western blotting, flow cytometry for cellular expression, and quantitative PCR for mRNA expression levels .

When working with recombinant IL-36B proteins for standards or experimental stimulations, proper reconstitution is critical. Typically, lyophilized IL-36B should be reconstituted at concentrations around 100 μg/mL in sterile PBS, with or without carrier proteins (such as 0.1% human or bovine serum albumin) depending on the experimental requirements .

How should researchers prepare and store recombinant IL-36B for experimental use?

Proper handling of recombinant IL-36B is essential for maintaining its biological activity:

Reconstitution protocols:

  • For carrier-containing formulations: Reconstitute at 100 μg/mL in sterile PBS containing at least 0.1% human or bovine serum albumin

  • For carrier-free formulations: Reconstitute at 100 μg/mL in sterile PBS without additives

Storage recommendations:

  • Store lyophilized protein at -20°C to -80°C

  • After reconstitution, aliquot and store at -20°C to -80°C

  • Avoid repeated freeze-thaw cycles as they significantly reduce protein activity

  • For short-term use (1-2 weeks), reconstituted protein can be stored at 2-8°C

Working solution preparation:

  • Dilute stock solutions to appropriate working concentrations immediately before use

  • Use low-binding tubes and pipette tips to minimize protein loss through adhesion

When designing experiments, it's important to note that E. coli-derived human IL-36B protein (Met1-Glu157) is commonly used for research applications, though mammalian cell-expressed versions may offer different post-translational modifications that could affect biological activity in certain experimental contexts .

What experimental models are most appropriate for studying IL-36B function?

Selecting appropriate experimental models for IL-36B research depends on the specific aspects being investigated:

In vitro models:

  • Human keratinocyte cell lines (HaCaT, NHEK) for skin-related studies

  • Human monocyte/macrophage cell lines (THP-1, U937) for immune response studies

  • Human endothelial cells (HUVEC) for vascular inflammation studies

  • Primary dendritic cells for antigen presentation research

  • T cell cultures for adaptive immunity investigations

In vivo models:

  • IL-36α knockout (–/–) mice have shown reduced neutrophil recruitment and CXCL1 generation

  • Similar knockout models for IL-36B provide insights into its specific functions

  • Tissue-specific conditional knockout or overexpression models help assess organ-specific effects

Disease-specific models:

  • Imiquimod-induced psoriasis models

  • DSS-induced colitis for inflammatory bowel disease studies

  • Collagen-induced arthritis for rheumatoid arthritis investigations

When working with these models, it's important to recognize the species-specific differences in IL-36 family expression and signaling, which may affect the translational relevance of findings between murine models and human disease .

What is the relationship between IL-36B and specific inflammatory diseases?

IL-36B has been implicated in several inflammatory conditions, with varying degrees of involvement:

DiseaseIL-36B RoleKey Research FindingsPotential Biomarker Value
PsoriasisMajor contributorElevated expression in lesional skin; Drives keratinocyte proliferation and neutrophil recruitmentHigh - correlates with disease severity
Inflammatory Bowel DiseaseContributing factorIncreased expression in colonic mucosa of IBD patients; Promotes intestinal inflammationModerate - may distinguish disease subtypes
Rheumatoid ArthritisPotential contributorDetected in synovial tissue; May enhance synovial fibroblast activationUnder investigation - preliminary connection
Systemic Lupus ErythematosusBeing investigatedPossible role in cutaneous manifestationsLimited evidence to date

The involvement of IL-36B in inflammatory diseases suggests potential therapeutic strategies targeting this cytokine or its signaling pathway. Understanding disease-specific mechanisms will help determine whether IL-36B represents a viable therapeutic target in different inflammatory conditions .

How does IL-36B interact with other members of the IL-36 family in regulating inflammatory responses?

The interplay between IL-36B and other IL-36 family members creates a complex regulatory network:

Agonist cooperation and redundancy:

  • IL-36α, IL-36β, and IL-36γ share the same receptor complex and induce similar but not identical downstream effects

  • Evidence suggests partial functional redundancy, though tissue-specific expression patterns may create distinct local effects

  • Co-expression of multiple IL-36 agonists often occurs in inflamed tissues, suggesting synergistic actions

Antagonist regulation:

Cross-regulation with other IL-1 family members:

  • IL-36 signaling can induce expression of other IL-1 family cytokines

  • IL-1β and IL-36 often operate in feed-forward inflammatory loops

  • IL-18 and IL-36 pathways show evidence of cross-talk in certain inflammatory contexts

Understanding these complex interactions is crucial for developing targeted therapeutic approaches that modulate specific aspects of the inflammatory response without completely suppressing protective immunity .

What are the challenges in developing IL-36B as a therapeutic target for inflammatory diseases?

Developing IL-36B-targeted therapies presents several significant challenges:

Biological complexity challenges:

  • Functional redundancy among IL-36 agonists may limit efficacy of IL-36B-specific inhibition

  • The wider IL-1 family network might compensate for IL-36B blockade

  • Cell type-specific and context-dependent effects complicate therapeutic planning

Target validation challenges:

  • Limited validation in human subjects compared to animal models

  • Varied expression and roles across different inflammatory diseases

  • Incomplete understanding of the relative importance compared to established therapeutic targets

Technical development challenges:

  • Designing specific inhibitors that don't cross-react with related IL-1 family members

  • Achieving appropriate tissue distribution, particularly for skin or mucosal diseases

  • Balancing efficacy with potential immunosuppressive side effects

Clinical translation challenges:

  • Identifying appropriate patient subgroups most likely to benefit

  • Developing reliable biomarkers to monitor therapeutic response

  • Determining optimal timing of intervention in disease course

Despite these challenges, the specific involvement of IL-36B in inflammatory pathways makes it an attractive target for future therapeutic development, particularly for diseases with limited treatment options or where existing therapies targeting broader inflammatory pathways have shown toxicity concerns .

What cutting-edge approaches are emerging for manipulating IL-36B in research and therapy?

Several innovative approaches are being developed to study and target IL-36B:

Advanced genetic tools:

  • CRISPR/Cas9-mediated gene editing to create precise human cell models

  • Inducible expression systems to study temporal aspects of IL-36B signaling

  • Single-cell RNA sequencing to identify IL-36B-responsive cell populations with high resolution

Novel therapeutic modalities:

  • Monoclonal antibodies specifically targeting IL-36B

  • Small molecule inhibitors of IL-36 receptor complex formation

  • Engineered IL-36Ra variants with enhanced antagonist properties

  • Antisense oligonucleotides and siRNA approaches to downregulate IL-36B expression

Innovative delivery strategies:

  • Topical formulations for skin diseases like psoriasis

  • Mucosa-targeted delivery systems for IBD

  • Nanoparticle-based delivery to specific immune cell populations

Combination approaches:

  • Dual targeting of IL-36B alongside established therapeutic targets

  • Sequential therapy protocols based on disease phase

  • Personalized approaches guided by IL-36 family expression patterns

These emerging approaches provide researchers with new tools to dissect IL-36B biology while simultaneously advancing potential therapeutic strategies toward clinical application .

How can researchers overcome challenges in detecting low levels of IL-36B in biological samples?

Detecting low-abundance IL-36B presents specific challenges that can be addressed through methodological optimization:

Sample preparation enhancements:

  • Implement concentration techniques such as sample precipitation or ultrafiltration

  • Optimize extraction buffers to include protease inhibitors preventing degradation

  • Consider immunoprecipitation to enrich IL-36B before detection

Assay sensitivity improvements:

  • Select ultra-sensitive ELISA kits with lower detection limits (down to 18.75 pg/mL)

  • Implement signal amplification methods like biotin-streptavidin systems

  • Extend incubation times while maintaining temperature control

  • Reduce background through optimized blocking and washing steps

Alternative detection platforms:

  • Consider digital ELISA technologies (e.g., Simoa) for single-molecule detection capabilities

  • Explore mass spectrometry-based approaches for absolute quantification

  • Implement proximity extension assays for improved sensitivity in complex matrices

Data analysis approaches:

  • Utilize standard curve fitting with wider dynamic ranges

  • Apply appropriate statistical methods for values near detection limits

  • Consider ratio-based analyses comparing IL-36B to related family members

When dealing with particularly challenging samples, combining complementary detection methods often provides the most robust results and confidence in data interpretation .

What are the key considerations for designing IL-36B stimulation experiments?

Effective experimental design for IL-36B stimulation studies requires careful attention to several parameters:

Recombinant protein selection:

  • Consider the source (bacterial vs. mammalian expression) based on experimental needs

  • Verify activity through pilot experiments before full studies

  • Determine whether carrier-free formulations are needed to avoid BSA interference

Dosage and kinetics optimization:

  • Perform dose-response curves (typically 1-100 ng/mL range)

  • Establish appropriate time course (acute vs. chronic stimulation)

  • Consider sequential or pulsed stimulation paradigms for certain applications

Cell preparation factors:

  • Cell density and passage number significantly impact responsiveness

  • Serum starvation before stimulation may enhance sensitivity

  • Account for endogenous IL-36B production in certain cell types

Controls and validation:

  • Include IL-36Ra as competitive inhibitor control

  • Use neutralizing antibodies to confirm specificity

  • Consider IL-36 receptor knockdown validation studies

  • Include other IL-36 family members for comparative analysis

A systematic approach to optimization, beginning with established protocols and methodically adjusting parameters based on pilot results, generally yields the most reliable and reproducible experimental outcomes .

How can contradictory findings in IL-36B research be reconciled?

Contradictory findings in IL-36B research can arise from numerous sources, requiring systematic resolution approaches:

Methodological differences analysis:

  • Compare detection methods and their limitations

  • Assess cell types and their specific IL-36 receptor expression profiles

  • Evaluate stimulation conditions (concentration, duration, context)

  • Examine sample processing variations between studies

Biological context considerations:

  • Species differences (human vs. murine systems)

  • Disease model variations affecting background inflammation

  • Genetic background affecting IL-36 pathway components

  • Presence of endogenous antagonists like IL-36Ra

Statistical and reporting factors:

  • Sample size and power differences between studies

  • Threshold differences for reporting significant changes

  • Publication bias favoring positive findings

Resolution strategies:

  • Direct replication studies maintaining constant core methodology

  • Meta-analysis of available data with subgroup analysis

  • Collaborative cross-laboratory validation projects

  • Use of multiple complementary techniques to assess the same outcome

By systematically analyzing sources of variation and implementing rigorous validation approaches, researchers can build consensus around initially contradictory findings and advance the field toward more unified understanding of IL-36B biology .

What genomic and epigenetic factors regulate IL-36B expression?

Understanding the genomic and epigenetic regulation of IL-36B represents an important frontier in research:

Genomic regulatory elements:

  • Promoter region characterization for transcription factor binding sites

  • Enhancer mapping through chromatin immunoprecipitation techniques

  • Identification of single nucleotide polymorphisms affecting expression levels

  • Investigation of long-range genomic interactions influencing IL-36B regulation

Epigenetic mechanisms:

  • DNA methylation patterns at the IL-36B locus under different inflammatory conditions

  • Histone modification profiles associated with active vs. silent IL-36B expression

  • Chromatin accessibility analysis through ATAC-seq or DNase-seq

  • Non-coding RNA involvement in post-transcriptional regulation

Cell-specific regulatory landscapes:

  • Comparison of regulatory mechanisms across relevant cell types (keratinocytes, immune cells)

  • Investigation of tissue-specific enhancers controlling contextual expression

  • Analysis of transcription factor networks in different cellular environments

Future studies employing chromosome conformation capture techniques, CRISPR-based epigenome editing, and single-cell multi-omics approaches will likely provide deeper insights into the complex regulatory mechanisms controlling IL-36B expression in health and disease .

How might IL-36B research contribute to personalized medicine approaches?

IL-36B research has significant potential to advance personalized medicine strategies:

Patient stratification opportunities:

  • IL-36B expression patterns as predictive biomarkers for treatment response

  • Genetic variants in IL-36 pathway components identifying susceptible subpopulations

  • Ratio analyses of IL-36 agonists to antagonists predicting disease severity

  • Combination biomarker panels including IL-36B improving diagnostic precision

Therapeutic targeting possibilities:

  • Selective IL-36B inhibition for patients with predominant IL-36B-driven pathology

  • Broader IL-36 family targeting for complex presentations

  • Complementary targeting approaches based on individual cytokine profiles

  • Localized vs. systemic intervention strategies based on disease manifestation

Monitoring applications:

  • Serial IL-36B measurements tracking treatment efficacy

  • Early warning system for disease flares based on IL-36B elevation

  • Tissue-specific sampling strategies for personalized assessment

  • Integration with other inflammatory markers for comprehensive monitoring

As research advances, the integration of IL-36B data with broader patient characteristics and response patterns will likely enable increasingly sophisticated personalized treatment algorithms for inflammatory diseases where IL-36B plays a significant role .

What are the unexplored roles of IL-36B in non-inflammatory physiological processes?

Beyond inflammation, several potential roles for IL-36B warrant further investigation:

Tissue homeostasis and repair:

  • Potential involvement in epithelial barrier maintenance

  • Roles in wound healing and tissue regeneration

  • Contributions to normal immune surveillance functions

  • Involvement in microbiome interaction at barrier surfaces

Developmental biology:

  • Expression patterns during embryonic and postnatal development

  • Potential roles in tissue patterning and cellular differentiation

  • Involvement in immune system maturation processes

  • Developmental switches in IL-36 family expression

Metabolic regulation:

  • Emerging connections between IL-36 family members and metabolic pathways

  • Potential roles in adipose tissue biology

  • Interactions with nutritional signals and metabolic stress

  • Cross-talk with hormonal regulation systems

Neurological functions:

  • Expression in neural tissues and potential signaling roles

  • Contributions to neuro-immune communication

  • Involvement in pain perception pathways

  • Potential roles in cognitive function

Exploring these non-inflammatory aspects of IL-36B biology may reveal unexpected functions and potential applications beyond current disease-focused research, potentially opening entirely new therapeutic avenues .

What are the most promising near-term applications of IL-36B research?

Based on current knowledge, several promising near-term applications of IL-36B research are emerging:

Diagnostic applications:

  • Biomarker development for early disease detection

  • Differential diagnosis tools for inflammatory skin conditions

  • Disease activity monitoring in established conditions

  • Predictive markers for treatment response

Therapeutic applications:

  • Targeted inhibitors for IL-36B-mediated diseases

  • Combination therapies addressing multiple inflammatory pathways

  • Topical interventions for localized inflammatory conditions

  • Preventative approaches for high-risk individuals

Research tool applications:

  • Improved detection methods for basic and clinical research

  • Standardized stimulation protocols for immunology studies

  • Reporter systems for pathway activation analysis

  • Humanized mouse models for translational research

These applications represent the most direct translation of current IL-36B knowledge into practical tools that could impact patient care and research capabilities within the next 3-5 years .

What collaboration models would accelerate IL-36B research?

Accelerating progress in IL-36B research would benefit from specific collaborative approaches:

Interdisciplinary research consortia:

  • Integration of immunologists, dermatologists, gastroenterologists, and rheumatologists

  • Bioengineers and chemists for therapeutic development

  • Computational biologists for systems-level analysis

  • Clinical researchers for translational applications

Resource sharing initiatives:

  • Biobanks with well-characterized samples from relevant diseases

  • Data repositories combining clinical and molecular information

  • Protocol standardization for improved cross-study comparison

  • Reagent validation and sharing programs

Coordinated clinical investigation:

  • Multi-center observational studies with standardized biospecimen collection

  • Collaborative clinical trials testing IL-36-targeted interventions

  • Patient registries with longitudinal sampling and outcomes tracking

  • International networks capturing population-level diversity

Industry-academic partnerships:

  • Joint development of therapeutic candidates

  • Collaborative biomarker discovery and validation

  • Shared research facilities with complementary expertise

  • Pre-competitive collaborations on fundamental biology

Product Science Overview

Structure and Activation

IL-36β is synthesized as an inactive precursor and requires proteolytic processing for activation. This processing typically involves the removal of the N-terminal propeptide, which is necessary for the cytokine to become biologically active . The active form of IL-36β binds to the IL-36 receptor (IL-36R), which then recruits the IL-1 receptor accessory protein (IL-1RAcP) to form a signaling complex. This complex activates intracellular signaling pathways, including NF-κB and MAPK, leading to the production of pro-inflammatory cytokines .

Expression and Function

IL-36β is primarily expressed in epithelial cells, including keratinocytes in the skin, as well as in various immune cells such as dendritic cells and macrophages . It is upregulated in response to inflammatory stimuli and plays a crucial role in the immune response by promoting the activation and polarization of T cells and dendritic cells .

In the skin, IL-36β contributes to the inflammatory response and is involved in the pathogenesis of several skin disorders, including psoriasis. It enhances the Th17/Th23 axis, which is known to be dysregulated in psoriatic lesions . IL-36β is also expressed in other tissues, such as the lungs, intestines, and joints, where it may play roles in various inflammatory conditions .

Clinical Implications

The dysregulation of IL-36β has been implicated in several inflammatory diseases. For instance, mutations in the IL-36Ra gene, which encodes the antagonist of IL-36β, are associated with generalized pustular psoriasis, a severe and potentially life-threatening skin condition . Additionally, elevated levels of IL-36β have been observed in psoriatic plaques compared to healthy skin .

Therapeutic strategies targeting IL-36β are being explored for the treatment of inflammatory diseases. Anti-IL-36 antibodies have shown promise in preclinical models of psoriasis, where they attenuate skin inflammation . These findings suggest that IL-36β could be a potential therapeutic target for managing inflammatory conditions.

Recombinant IL-36β

Recombinant human IL-36β is produced using various expression systems, including E. coli. The recombinant protein is typically purified to high levels of purity and is used in research to study its biological functions and potential therapeutic applications . It is available in both carrier-free and carrier-containing formulations, depending on the intended use .

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.