IL37 Human

Interleukin-37 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to IL-37

Interleukin-37 (IL-37), also known as IL-1 family member 7 (IL-1F7), is a human-specific anti-inflammatory cytokine discovered in 2000 through bioinformatics analysis . It belongs to the IL-1 family but exhibits distinct regulatory properties, primarily acting as a "brake" on excessive inflammation and immune responses . Unlike other IL-1 family members, IL-37 lacks a mouse homolog, necessitating transgenic models for preclinical studies . Its dual functionality—acting intracellularly and extracellularly—sets it apart as a critical modulator of innate and adaptive immunity .

Gene Structure and Isoforms

The IL-37 gene is located on chromosome 2q14 in humans and undergoes alternative splicing to generate five isoforms (IL-37a–e), which differ in exon composition and tissue expression .

IsoformExonsTissue ExpressionBiological Function
IL-37a3,4,5,6Brain, thymus, bone marrowAnti-inflammatory (functional)
IL-37b1,2,4,5,6Heart, kidney, blood, skinMost studied; inhibits cytokine genes
IL-37c1,2,5,6Kidney, lung, colonNon-functional (misfolded)
IL-37d1,4,5,6Bone marrow, testisAnti-inflammatory (limited data)
IL-37e1,5,6TestisTissue-specific role (unexplored)

IL-37b is the longest and most biologically active isoform, sharing structural homology with IL-1 family members but lacking a signal peptide .

Gene Expression and Synthesis

IL-37 is constitutively expressed in immune cells (monocytes, NK cells, B lymphocytes) and epithelial tissues (keratinocytes, intestinal epithelial cells) . Its expression is upregulated by pro-inflammatory stimuli, including:

  • TLR agonists (e.g., LPS)

  • Cytokines (IL-1β, TGF-β)

  • Pathogens (bacterial/viral components) .

Synthesis Pathway:

  1. Precursor Form: IL-37 is synthesized as an inactive precursor in the cytosol.

  2. Cleavage: Caspase-1 processes the precursor into mature IL-37 during inflammasome activation .

  3. Secretion: Both precursor and mature forms are secreted via non-classical pathways (exact mechanism undefined) .

Mechanisms of Action

IL-37 exerts anti-inflammatory effects through intracellular and extracellular pathways .

Intracellular Mechanism

  • Mature IL-37 binds phosphorylated Smad3, forming a complex that translocates to the nucleus.

  • Directly inhibits transcription of pro-inflammatory cytokines (IL-1β, TNF-α, IFN-γ) and chemokines .

Extracellular Mechanism

  • Forms a complex with IL-18Rα and IL-1R8 (Sigirr) on cell surfaces.

  • Blocks NF-κB and p38 MAPK signaling, reducing cytokine production (e.g., IL-6, CXCL8) .

  • IL-37 binding induces GSK3β activation, promoting Sigirr degradation via ubiquitination .

Clinical Implications and Disease Associations

IL-37’s role in human diseases is context-dependent, with therapeutic potential in inflammation-driven disorders.

DiseaseIL-37 LevelsEffectReferences
Autoimmune DiseasesElevatedLimits tissue damage in rheumatoid arthritis, lupus, and multiple sclerosis
Inflammatory Bowel Disease (IBD)ReducedTransgenic IL-37 mice show reduced colitis severity
CancerContext-dependent↑ IL-37: Tumor suppressive (breast, ovarian cancer)
↓ IL-37: Promotes progression (lung, colon, AML)
SepsisElevatedCorrelates with survival; inhibits cytokine storms

Cancer Dichotomy: IL-37 exhibits tumor-suppressive effects in solid tumors (e.g., renal cell carcinoma via IL-6/STAT3 inhibition) but may promote hematological malignancies like AML by modulating IL-6 signaling .

Key Research Trends

  1. Bibliometric Analysis:

    • Top journals: Cytokine, Nature Immunology

    • Leading countries: China (48.1% publications), USA (23.2%)

    • Pioneers: Charles Dinarello (most publications), Marcel Nold (high citations) .

  2. Emerging Hotspots:

    • Immunotherapy: IL-37-engineered cells for cancer treatment .

    • Gut Microbiome: Role in intestinal epithelial barrier function .

    • Ethnic Variability: Higher IL-37 levels observed in Chinese populations .

Challenges

  • Lack of Mouse Models: Transgenic mice are primary tools for in vivo studies .

  • Isoform-Specific Functions: IL-37a, c, d, e require further characterization .

Product Specs

Introduction
Human interleukin 1 family member 7 (IL1F7), also known as IL-37, is a cytokine that acts as a natural suppressor of innate immunity. It exists in multiple isoforms with varying lengths and expression patterns. The longest isoform, IL1F7b, can bind to the interleukin-18 receptor (IL18R) with low affinity but does not activate it. Instead, IL1F7b interacts with the IL-18 binding protein (IL18BP), forming a complex that prevents IL-18 from binding to its receptor and initiating an inflammatory response.
Description
This product consists of the recombinant form of human Interleukin-37, produced in E. coli. It is a non-glycosylated polypeptide chain encompassing 167 amino acids (Lys27-Asp192) with a molecular weight of 18.6 kDa. The protein has been purified using specialized chromatographic techniques to ensure its high quality.
Physical Appearance
The product is provided as a sterile, white powder that has been lyophilized (freeze-dried) to maintain its stability.
Formulation

The lyophilization of IL37 was carried out from a 0.2 µM filtered solution containing 20mM PB (phosphate buffer), 150mM NaCl (sodium chloride), and 2mM DTT (dithiothreitol) at a pH of 7.4.

Solubility
For optimal use, it is recommended to briefly centrifuge the vial before reconstituting the lyophilized IL37 in phosphate-buffered saline (PBS) to a minimum concentration of 100 µg/ml. This solution can then be further diluted in other aqueous solutions as needed.
Stability
Lyophilized IL37 remains stable for up to 3 weeks at room temperature, but for long-term storage, it is best kept desiccated at a temperature below -18°C. Once reconstituted, the IL-37 solution should be stored at 4°C for 2-7 days. For extended storage, it can be kept at -18°C, but repeated freeze-thaw cycles should be avoided.
Purity

The purity of the IL37 is greater than 95.0%, as determined by SDS-PAGE analysis.

Biological Activity
The protein's biological activity, specifically its binding ability, has been assessed using a functional ELISA. Immobilized IL1F7 at a concentration of 1 µg/ml (100 µl/well) can bind to recombinant human IL-18 Receptor/Fc Chimera within a linear range of 0.015-1 µg/ml.
Synonyms
Interleukin-37, FIL1 zeta, IL-1X, Interleukin-1 family member 7, IL-1F7, Interleukin-1 homolog 4, IL-1H, IL-1H4, Interleukin-1 zeta, IL-1 zeta, Interleukin-1-related protein, IL-1RP1, Interleukin-23, IL-37, IL37, FIL1Z, IL1F7, IL1H4, IL1RP1, FIL1, FIL1(ZETA).
Source
Escherichia Coli.
Amino Acid Sequence
MKNLNPKKFSIHDQDHKVLVLDSGNLIAVPDKNYIRPEIFFALASSLSSASAEK
GSPILLGVSKGEFCLYCDKDKGQSHPSLQLKKEKLMKLAAQKESARRPFIFYR
AQVGSWNMLESAAHPGWFICTSCNCNEPVGVTDKFENRKHIEFSFQPVCKAE
MSPSEVSD.

Q&A

What is IL-37 and what are its primary functions in human immunity?

IL-37 is a member of the IL-1 family (also previously known as IL-1F7) discovered by computational cloning in 2000. It functions primarily as an immunosuppressive cytokine that inhibits innate immunity and inflammatory responses. IL-37 suppresses the production of pro-inflammatory cytokines (including IL-6, IL-1α, and TNF-α) in innate immune cells and inhibits the maturation of dendritic cells . Unlike most IL-1 family members that promote inflammation, IL-37 serves as a fundamental negative regulator of inflammation, making it a critical modulator in various inflammatory and autoimmune conditions.

Table 1: Publication Growth of IL-37 Research (2001-2021)

PeriodNumber of PublicationsNotable Research Focus
2001-201222Initial discovery and characterization
2013-2016~150Receptor identification and signaling
2017-2021>350Disease associations and mechanisms

How does IL-37 differ from other members of the IL-1 family in structure and function?

Unlike most IL-1 family cytokines that are primarily pro-inflammatory, IL-37 exhibits distinct anti-inflammatory properties. Structurally, IL-37 maintains the β-trefoil fold characteristic of the IL-1 family, but its unique receptor requirements and signaling pathways set it apart functionally. IL-37 specifically requires IL-18Rα and IL-1R8 (SIGIRR) receptors to execute its anti-inflammatory program . Furthermore, IL-37 demonstrates both intracellular and extracellular mechanisms of action, with extracellular forms requiring the IL-1 family decoy receptor IL-1R8 . This dual functionality gives IL-37 a more complex regulatory role compared to other IL-1 family members.

What are the primary signaling pathways through which human IL-37 exerts its immunosuppressive effects?

IL-37 exerts its immunosuppressive effects through multiple signaling pathways. The primary mechanism involves binding to IL-18Rα and IL-1R8 (SIGIRR) receptors, which initiates anti-inflammatory signaling cascades . This receptor engagement is critical for IL-37's ability to suppress NF-κB activation, as evidenced by the high centrality of "NF-κB" in keyword co-occurrence analysis . Intracellularly, IL-37 can translocate to the nucleus and influence gene expression. The extracellular forms of IL-37 inhibit innate inflammation but require the IL-1 family decoy receptor IL-1R8, highlighting the complexity of IL-37 signaling networks in modulating immune responses through multiple pathways.

How is IL-37 expression regulated across different human tissues and disease states?

IL-37 expression varies significantly across tissues and is dynamically regulated in different disease states. Bibliometric analysis shows that "expression" is one of the most important terms associated with IL-37 research, with 165 co-occurrences . Increased levels of circulating IL-37 have been documented in patients with rheumatoid arthritis, multiple sclerosis, and systemic lupus erythematosus . In rheumatoid arthritis patients specifically, there is elevated expression of synovial IL-1R8, which is essential for IL-37's anti-inflammatory effects. The regulation of IL-37 appears to be particularly responsive to inflammatory stimuli, suggesting a feedback mechanism where IL-37 increases during inflammation to help limit immune responses and prevent excessive tissue damage.

What are the most reliable methods for quantifying IL-37 expression in human clinical samples?

When quantifying IL-37 in human clinical samples, researchers should consider multiple complementary approaches to ensure reliable results. For protein detection, enzyme-linked immunosorbent assay (ELISA) remains the gold standard, though western blotting provides information about protein processing forms. For transcriptional analysis, quantitative PCR (qPCR) offers sensitive detection of IL-37 mRNA. Immunohistochemistry and flow cytometry are valuable for assessing cellular sources of IL-37 within tissues or blood samples.

Table 2: Recommended Methods for IL-37 Quantification

MethodApplicationStrengthsLimitations
ELISASerum/plasma levelsQuantitative, high-throughputCannot distinguish isoforms
qPCRmRNA expressionHighly sensitive, isoform-specificDoesn't confirm protein expression
Flow CytometryCellular sourcesSingle-cell resolutionRequires cell isolation
IHC/IFTissue localizationPreserves anatomical contextSemi-quantitative
Mass SpectrometryProtein modificationsIdentifies PTMs, high specificityComplex sample preparation

The bibliometric analysis indicates that "differential expression" (cluster #2) has been a significant research focus in IL-37 studies , highlighting the importance of comparative expression analysis between healthy and diseased states.

How should researchers design experiments to accurately assess IL-37's immunosuppressive functions?

Designing experiments to assess IL-37's immunosuppressive functions requires careful consideration of multiple factors. In vitro approaches should include:

  • Dose-response experiments with recombinant IL-37 on relevant human immune cells (monocytes, macrophages, dendritic cells)

  • Time-course studies to determine optimal pre-treatment or co-treatment schedules with inflammatory stimuli

  • Comparison of effects on multiple inflammatory mediators (cytokines, chemokines, surface markers)

  • Receptor blocking experiments to confirm IL-18Rα and IL-1R8 dependency

  • Knockdown/knockout studies using siRNA or CRISPR-Cas9

For in vivo studies, transgenic mouse models expressing human IL-37 have proven valuable, as evidenced by the high citation of research showing protection from colitis in IL-37-expressing mice . When designing these experiments, researchers must include appropriate controls and consider how the timing of IL-37 administration relative to inflammatory stimuli affects outcomes.

What cell and animal models are most appropriate for investigating IL-37 mechanisms in different disease contexts?

The selection of appropriate models for IL-37 research depends on the specific disease context and research question. For cellular models, primary human monocytes, macrophages, and dendritic cells are preferred as they express the necessary receptors (IL-18Rα and IL-1R8) . The keyword analysis showing "dendritic cell" with high co-occurrence confirms their importance in IL-37 research .

For disease-specific studies, researchers should consider:

  • Rheumatoid arthritis: Fibroblast-like synoviocytes (IL-37 induces apoptosis in these cells)

  • Inflammatory bowel disease: Intestinal epithelial cells and colitis models

  • Cardiovascular disease: Vascular endothelial cells and atherosclerosis models

What analytical approaches should be employed when studying genetic variations in IL-37 across human populations?

When investigating IL-37 genetic variations across populations, researchers should implement a comprehensive analytical framework:

  • Study design considerations:

    • Account for population stratification using principal component analysis

    • Calculate adequate sample sizes based on expected effect sizes and allele frequencies

    • Include diverse populations (the research shows IL-37 studies span 50 countries)

  • Genotyping approaches:

    • Targeted sequencing of IL-37 and receptor genes

    • Genome-wide association studies for broader genetic context

    • Haplotype analysis to identify linked polymorphisms

  • Functional validation:

    • Reporter assays to assess promoter variants

    • Expression quantitative trait loci (eQTL) analysis

    • In vitro testing of variant IL-37 proteins

  • Data analysis:

    • Control for multiple testing using Bonferroni or false discovery rate methods

    • Conduct gene-environment interaction analysis

    • Perform meta-analysis across cohorts

The focus on "pathogenesis" and "disease" in keyword co-occurrence suggests that genetic variation in IL-37 may significantly contribute to disease susceptibility or progression, warranting rigorous analytical approaches.

How does IL-37 function within the broader cytokine network to maintain immune homeostasis?

IL-37 operates within a complex cytokine network to maintain immune homeostasis through multiple mechanisms:

  • Counter-regulation of pro-inflammatory cytokines: IL-37 suppresses the production of key inflammatory mediators including IL-6, IL-1α, TNF-α, and other cytokines

  • Receptor cross-talk: The requirement for IL-18Rα suggests competitive inhibition with IL-18 signaling pathways, representing an important checkpoint in inflammation

  • Cell-specific regulatory effects: IL-37 modulates dendritic cell maturation and function, affecting downstream T cell responses and adaptive immunity

  • Feedback inhibition loops: Inflammatory stimuli that activate NF-κB pathways induce IL-37 expression, which then suppresses further NF-κB activation, creating a negative feedback loop

The co-citation analysis showing "related cytokine" as cluster #5 highlights the academic interest in understanding IL-37's position within broader cytokine networks. Researchers investigating this area should employ systems biology approaches that can capture the dynamic interplay between multiple cytokines simultaneously, rather than studying IL-37 in isolation.

What is the current understanding of IL-37's role in the pathogenesis of autoimmune diseases?

IL-37 plays complex roles in autoimmune disease pathogenesis, often serving as an endogenous brake on inflammation:

Table 3: IL-37 in Major Autoimmune Conditions

DiseaseIL-37 Expression PatternFunctional SignificanceReference
Rheumatoid ArthritisElevated in circulation, increased synovial IL-1R8Induces apoptosis in fibroblast-like synoviocytes, counter-regulatory mechanism
Multiple SclerosisIncreased circulating levelsMay limit neuroinflammation
Systemic Lupus ErythematosusElevated in serumCounter-regulatory mechanism

The elevated IL-37 levels in these conditions suggest activation of endogenous anti-inflammatory mechanisms attempting to control disease progression. This has led researchers to investigate IL-37 supplementation as a potential therapeutic strategy. The keyword co-occurrence showing "rheumatoid arthritis" as a significant term confirms the importance of this autoimmune condition in IL-37 research.

Methodologically, researchers studying IL-37 in autoimmune diseases should examine both systemic and local expression patterns, while also investigating how genetic variations in IL-37 or its receptors might influence disease susceptibility or severity.

How do post-translational modifications influence the biological activity of human IL-37?

Post-translational modifications (PTMs) critically influence IL-37's biological activity through several mechanisms:

  • Proteolytic processing: Like many IL-1 family members, IL-37 requires specific proteolytic cleavage for full bioactivity. The processing of IL-37 by caspase-1 has been shown to enhance its anti-inflammatory properties.

  • Phosphorylation: Potential phosphorylation sites may regulate IL-37's interaction with signaling components or influence its intracellular trafficking.

  • Glycosylation: Glycosylation could affect IL-37's stability, receptor binding affinity, and circulation half-life.

  • Nuclear localization: Modifications affecting nuclear translocation may determine IL-37's ability to influence gene transcription directly.

The co-citation of papers discussing "extracellular forms of IL-37" suggests that processing of IL-37 is important for its function. Researchers investigating IL-37 PTMs should employ mass spectrometry-based approaches to identify modification sites, followed by site-directed mutagenesis to assess functional consequences. Additionally, researchers should consider how inflammatory environments might alter the PTM profile of IL-37, potentially affecting its anti-inflammatory properties in disease states.

What are the key challenges and strategies in developing IL-37-based therapeutic interventions?

Developing IL-37-based therapeutic interventions presents several challenges that researchers must address:

  • Delivery and formulation challenges:

    • Recombinant IL-37 has limited stability in circulation

    • Targeted delivery to specific tissues requires novel formulation approaches

    • Optimal dosing regimens remain undetermined

  • Mechanistic considerations:

    • Dual intracellular and extracellular functions complicate therapeutic strategies

    • Receptor requirements (IL-18Rα and IL-1R8) vary across tissues

    • Timing of intervention is critical given the context-dependent effects

  • Safety concerns:

    • Potential immunogenicity of recombinant IL-37

    • Risk of compromising immune responses to pathogens

    • Differential effects across various autoimmune conditions

Strategies to overcome these challenges include developing stabilized IL-37 variants, receptor-specific agonists, gene therapy approaches, and small molecule enhancers of endogenous IL-37 expression. The keyword analysis showing "inhibitor" as a term with high centrality suggests research interest in how IL-37 inhibits inflammatory processes, which could inform therapeutic strategies targeting similar pathways.

How should researchers reconcile contradictory findings regarding IL-37 expression patterns in various diseases?

Contradictory findings regarding IL-37 expression across diseases represent a significant challenge requiring systematic resolution approaches:

  • Methodological assessment:

    • Evaluate differences in sample collection, processing, and storage

    • Compare detection methods (ELISA kits, antibodies, PCR primers)

    • Assess statistical approaches and sample sizes

  • Contextual factors:

    • Disease heterogeneity and subtype classification

    • Treatment status of patients and medication effects

    • Disease duration and activity measures

    • Tissue-specific versus systemic expression patterns

  • Analytical strategies:

    • Meta-analysis with random effects models to account for heterogeneity

    • Subgroup analysis based on methodological factors

    • Bayesian approaches incorporating prior knowledge

The cluster analysis identifying "differential expression" (cluster #2) as a significant research focus confirms that expression variability is a recognized aspect of IL-37 biology. Longitudinal studies examining IL-37 expression throughout disease progression could help clarify apparent contradictions observed in cross-sectional data.

What factors explain the variability in IL-37 responses observed across different human populations?

The variability in IL-37 responses across populations stems from multiple interacting factors:

  • Genetic determinants:

    • Polymorphisms in IL-37 coding regions affecting protein function

    • Variations in promoter regions influencing expression levels

    • Receptor (IL-18Rα and IL-1R8) genetic diversity

    • Background genetic modifiers affecting signaling pathways

  • Environmental influences:

    • Different pathogen exposure histories shaping immune responses

    • Dietary factors affecting inflammatory status

    • Environmental pollutants modulating cytokine networks

    • Microbiome composition variations

  • Methodological considerations:

    • Standardization of assays across populations

    • Accounting for reference ranges in different ethnic groups

    • Controlling for comorbidities and medication use

The bibliometric analysis showing research contributions from 50 different countries/regions suggests recognition of population differences in IL-37 biology. Researchers should conduct cross-population studies with standardized protocols and consider genome-wide association approaches to identify genetic factors influencing IL-37 responses.

What methodological approaches should researchers employ when IL-37 data contradicts established immunological paradigms?

When IL-37 data contradicts established immunological paradigms, researchers should follow these methodological principles:

  • Validation through multiple approaches:

    • Confirm findings using independent techniques

    • Replicate experiments in different model systems

    • Collaborate with independent laboratories for verification

  • Critical examination of assumptions:

    • Review the foundational evidence of the challenged paradigm

    • Identify contextual factors that might explain apparent contradictions

    • Consider cell-type or tissue-specific effects

  • Transparent reporting:

    • Document all methodological details thoroughly

    • Pre-register experimental protocols when possible

    • Report all results, including those that appear contradictory

  • Conceptual integration:

    • Propose mechanistic models that reconcile contradictory findings

    • Develop testable hypotheses that could resolve the contradiction

    • Consider paradigm refinement rather than replacement

The discovery that IL-37 is "a fundamental inhibitor of innate immunity" itself represented a paradigm shift in understanding IL-1 family cytokines, which were typically considered pro-inflammatory. This historical context provides a model for how researchers might approach new findings that challenge current understanding of IL-37 biology.

What statistical and bioinformatic approaches are most appropriate for analyzing complex IL-37 clinical datasets?

Analyzing complex IL-37 clinical datasets requires sophisticated statistical and bioinformatic approaches:

  • For observational clinical studies:

    • Multivariate regression models controlling for relevant confounders

    • Propensity score matching for case-control comparisons

    • Mediation analysis to assess causal pathways involving IL-37

  • For longitudinal data:

    • Mixed-effects models accounting for repeated measures

    • Time-series analysis for temporal patterns

    • Joint modeling for longitudinal biomarkers and clinical outcomes

  • For biomarker evaluation:

    • Receiver operating characteristic (ROC) curve analysis

    • Net reclassification improvement (NRI) assessment

    • Decision curve analysis for clinical utility

  • For integrative analyses:

    • Network analysis of cytokine interactions

    • Machine learning for pattern recognition in complex datasets

    • Multi-omics integration approaches

The keyword co-occurrence of "disease activity" suggests that correlating IL-37 levels with clinical indices is a common approach. Researchers should ensure appropriate sample size calculations during study design and consider Bayesian approaches when prior knowledge about IL-37 can inform analysis.

What emerging research trends are shaping the future of IL-37 investigation?

Bibliometric analysis reveals several emerging trends shaping the future of IL-37 research:

  • Expansion to diverse disease contexts:

    • The involvement of IL-37 in disorders beyond established autoimmune diseases

    • Cardiovascular diseases emerging as the largest cluster (#0) in co-citation analysis

    • Exploration of IL-37's role in metabolic disorders

  • Mechanistic refinement:

    • Additional immunomodulatory mechanisms beyond established anti-inflammatory effects

    • Understanding the dual intracellular and extracellular functions

    • Receptor-independent activities

  • Therapeutic applications:

    • "Suppression" continuing as an active research front in keyword burst analysis

    • Development of IL-37-based therapeutic interventions

    • Biomarker applications for disease monitoring

  • Tissue-specific biology:

    • "Epidermal barrier" (cluster #8) representing newer research directions

    • Tissue-resident immune cell responses to IL-37

    • Organ-specific functions and expression patterns

Researchers should position new studies to build upon these emerging trends while introducing innovative methodological approaches to address the complex biology of IL-37.

Which unexplored aspects of IL-37 biology warrant further investigation?

Despite significant research progress, several aspects of IL-37 biology remain underexplored:

  • Evolutionary biology and comparative immunology:

    • Why humans have IL-37 while mice naturally lack this gene

    • Evolutionary pressures that shaped IL-37 function

    • Comparative analysis across species with IL-37 homologs

  • Regulatory networks:

    • Transcriptional and post-transcriptional regulation of IL-37

    • microRNA networks controlling IL-37 expression

    • Epigenetic modifications of the IL-37 locus in different contexts

  • Cellular sources and targets:

    • Non-immune cell sources of IL-37

    • Cell-type specific responses to IL-37 signaling

    • The role of "mast cells" (cluster #6) in IL-37 biology

  • Developmental aspects:

    • IL-37 expression during different developmental stages

    • Role in tissue repair and regeneration

    • Influence on immune system development

The separate clustering of "IL-1F5" (SIGIRR, cluster #9) from other clusters suggests that the relationship between IL-37 and its receptor components still has unexplored facets that warrant further investigation.

How might emerging technologies advance our understanding of IL-37's functions and therapeutic potential?

Emerging technologies promise to revolutionize our understanding of IL-37 biology:

  • Single-cell technologies:

    • Single-cell RNA sequencing to identify specific cellular sources and responders

    • Single-cell proteomics to characterize IL-37 production at protein level

    • Spatial transcriptomics to map IL-37 expression in tissue contexts

  • Advanced genetic engineering:

    • CRISPR-Cas9 base editing for precise modification of IL-37 and receptor genes

    • Inducible expression systems for temporal control of IL-37

    • Humanized mouse models with physiological IL-37 expression

  • Structural and interaction biology:

    • Cryo-EM of IL-37-receptor complexes

    • Protein-protein interaction mapping using proximity labeling

    • Hydrogen-deuterium exchange mass spectrometry for conformational dynamics

  • Computational approaches:

    • Machine learning for pattern recognition in IL-37-related datasets

    • Systems biology modeling of IL-37 in cytokine networks

    • Virtual screening for small molecule modulators of IL-37 activity

These technologies will enable researchers to address complex questions about IL-37 function with unprecedented resolution and precision, potentially accelerating therapeutic applications.

What interdisciplinary approaches could yield breakthrough insights into IL-37 biology?

Interdisciplinary approaches hold particular promise for advancing IL-37 research:

  • Immunology + Systems Biology:

    • Network modeling of IL-37 within broader cytokine systems

    • Identification of emergent properties in inflammatory regulation

    • Multi-scale modeling from molecular to organism level

  • Immunology + Cardiovascular Research:

    • The cluster analysis identifying "cardiovascular disease" as the largest cluster

    • Investigating IL-37's role in atherosclerosis and heart failure

    • Effects on vascular inflammation and endothelial function

  • Immunology + Neuroscience:

    • Neuroimmune interactions mediated by IL-37

    • Role in neuroinflammatory and neurodegenerative conditions

    • Effects on microglial function and neuronal health

  • Immunology + Microbiome Research:

    • IL-37's role in host-microbiota interactions

    • Effects on intestinal barrier function and mucosal immunity

    • Microbial regulation of IL-37 expression

  • Immunology + Computational Drug Design:

    • Structure-based design of IL-37 mimetics or enhancers

    • AI-driven discovery of small molecule modulators

    • Modeling of pharmacokinetics and tissue distribution

These interdisciplinary approaches can address complex questions about how IL-37 functions in different physiological contexts and accelerate the translation of basic research findings into therapeutic applications.

Product Science Overview

Introduction

Interleukin-37, formerly known as Interleukin-1 Family Member 7, is a member of the Interleukin-1 family of cytokines. It is a newly discovered cytokine that plays a crucial role in suppressing innate inflammation and acquired immune responses. Interleukin-37 has five isoforms, designated as Interleukin-37a through Interleukin-37e, with Interleukin-37b being the most studied and longest isoform .

Biological Function

Interleukin-37 functions as a natural inhibitor of inflammatory and immune responses. It has been shown to suppress the production of pro-inflammatory cytokines such as Interleukin-1 alpha, Interleukin-1 beta, and Tumor Necrosis Factor alpha. Overexpression of Interleukin-37 in epithelial cells or macrophages almost completely suppresses the production of these cytokines, whereas silencing of endogenous Interleukin-37 increases their abundance in human blood cells .

Therapeutic Potential

Due to its potent anti-inflammatory properties, Interleukin-37 holds significant potential for treating a wide array of human inflammatory and autoimmune disorders. Studies have shown that administration of recombinant Interleukin-37 can ameliorate experimental psoriasis, alleviate rheumatoid arthritis, and reduce bleomycin-induced lung injury and fibrosis. It has also been found to decrease renal ischemia-reperfusion injury and inhibit the growth of cancer cells .

Recombinant Production

Recombinant Interleukin-37 can be produced using various expression systems, including bacterial, yeast, insect, and mammalian cells. Recently, plants have emerged as a novel expression platform for the production of human Interleukin-37. Transgenic plants have been developed to produce different forms of Interleukin-37b, including the unprocessed full-length precursor form and the mature form. The expression of these forms is driven by a strong constitutive promoter, and the resulting proteins are biologically active .

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.