Loss-of-function mutations in IL36RN are strongly linked to generalized pustular psoriasis (GPP), an autoinflammatory disorder characterized by recurrent sterile pustules, systemic inflammation, and fever .
Feature | IL36RN Mutation Carriers | Non-Carriers |
---|---|---|
Age of Onset | Earlier (pediatric cases common) | Later (adult-onset) |
Psoriasis Vulgaris (PV) | Less likely to have PV history | More likely to have PV |
Systemic Inflammation | Higher frequency of fever, leukocytosis | Lower systemic involvement |
Therapeutic Response | Higher recurrence risk post-acitretin withdrawal | Better sustained remission |
IL-36 Signaling Dysregulation:
Toll-Like Receptor (TLR) Interactions:
Parameter | IL36RN-Deficient Mice vs. Wild-Type Mice |
---|---|
Wound Closure Time | Delayed by 3 days post-injury |
Neutrophil/Macrophage Influx | Increased (3 days post-injury) |
Granulation Tissue Formation | Excessive, with elevated TGF-β and CXCL1 |
Diagnostic Criteria:
Pediatric onset GPP without PV history.
Recurrent sterile pustules with systemic inflammation (fever, leukocytosis).
Genetic Testing:
Ethnic Disparities: Higher IL36RN mutation prevalence in Asian populations warrants population-specific screening strategies .
Biomarker Development: Identifying IL-36γ or CXCL1 as serum markers for disease activity .
Personalized Therapies: IL-36R antagonists (e.g., spesolimab) require larger clinical trials to assess long-term efficacy .
IL36RN is one of nine IL-1 family genes (IL1A, IL1B, IL37, IL36A, IL36B, IL36G, IL36RN, IL38, IL1RN) located on human chromosome 2q13 clustered within 430 kb . This clustering suggests they originated from a common ancestral gene that underwent multiple gene duplications. The IL36RN gene specifically encodes IL-36Ra, which functions as a signaling inhibitor in the IL-36 pathway.
From an evolutionary perspective, the close proximity of these genes in the IL-1 cluster indicates a shared ancestry and functional relationship. The IL-1 cluster genes show conservation of protein structure, specifically a 12-strand beta-trefoil (12SBT) structure, with similar immunomodulatory functions . Understanding this genomic organization provides important context for interpreting mutations and their functional impacts.
Methodologically, researchers investigating the genomic organization should employ:
Comparative genomic analysis across species
Phylogenetic studies of IL-1 family genes
Functional analysis of conserved regions
IL-36Ra (encoded by IL36RN) functions as a competitive antagonist by binding to IL-36R without allowing productive engagement with IL-1RAcP (IL-1 receptor accessory protein) . This mechanism differs fundamentally from receptor blockade:
IL-36Ra binds to IL-36R with high affinity
This binding prevents IL-36 agonists (IL-36α, IL-36β, IL-36γ) from engaging the receptor
Most critically, IL-36Ra binding prohibits recruitment of IL-1RAcP, which is essential for signal transduction
Without IL-1RAcP recruitment, downstream signaling molecules (MyD88, IRAKs, TRAF6) cannot assemble properly
This prevents activation of NF-κB and MAPK pathways that would normally trigger inflammatory responses
To experimentally verify this mechanism, researchers should employ:
Binding affinity studies comparing IL-36Ra vs. IL-36 agonists
Co-immunoprecipitation experiments examining receptor complex formation
Reporter assays measuring NF-κB or MAPK activation
Structural studies of IL-36Ra/IL-36R interactions
Research indicates significant ethnic variation in IL36RN mutation patterns:
The c.115+6T>C mutation appears particularly prevalent in Chinese populations, occurring in multiple independent studies . When designing genetic screening protocols, researchers should:
Employ population-appropriate mutation panels
Consider whole exome sequencing for novel mutation discovery
Validate findings with functional assays
Use matched control populations for accurate interpretation of results
Effective genotype-phenotype correlation studies require:
Precise clinical categorization:
Comprehensive genetic analysis:
Full sequencing of IL36RN coding and splice regions
Analysis of copy number variations
Consideration of modifier genes
Statistical approaches:
Multivariate analysis controlling for confounding factors
Stratification by mutation type (nonsense, missense, splice-site)
Meta-analytic methods for combining data across cohorts
The evidence indicates IL36RN mutations occur approximately 3.82 times more frequently in GPP alone compared to GPP+PV (95% CI, 2.63–5.56), with similar effect sizes in European (OR = 4.03, 95% CI, 2.23–7.26) and Asian populations (OR = 3.69, 95% CI, 2.27–6.00) .
Several cell models offer distinctive advantages for IL36RN research:
Keratinocytes:
Intestinal epithelial cells:
Other validated models:
When selecting models, researchers should consider:
Expression levels of IL-36 pathway components
Tissue relevance to disease of interest
Availability of genetic modification tools
Reproducibility of inflammatory responses
Multiple complementary approaches provide robust assessment of IL-36Ra function:
Protein activity assays:
Biochemical approaches:
Cellular response measurements:
Quantification of IL-36-induced cytokines (RT-qPCR, ELISA)
Phosphorylation of downstream signaling components (Western blot)
Neutrophil recruitment assays (transwell migration)
Advanced methodologies:
Timing is critical - the NF-κB response to IL-36 stimulation peaks between 3-9 hours, making this window optimal for experimental measurements .
Proteolytic processing represents a critical regulatory step for IL-36 cytokines:
N-terminal truncation mechanism:
Protease involvement:
Research approaches:
In vitro digestion with purified proteases
Mass spectrometry identification of cleavage sites
Activity comparison of different N-terminal variants
Co-culture systems with neutrophils and IL-36-producing cells
Research indicates that IL36RN mutations may disrupt not only IL-36Ra production but potentially alter its processing, affecting the balance between active and inactive forms of both the antagonist and agonists.
Understanding pathway interactions is crucial for comprehensive IL36RN research:
Cross-regulation with other IL-1 family members:
Documented synergistic stimuli:
Inflammasome connections:
Experimental approaches:
Combinatorial stimulation experiments
Conditional knockout models
Multiplex cytokine profiling
Phospho-proteomic analysis of signaling networks
These interactions suggest IL36RN mutations may have broader impacts beyond direct IL-36 pathway dysregulation, potentially explaining phenotypic heterogeneity in patients.
Single-cell approaches offer transformative opportunities for IL36RN research:
Cellular heterogeneity insights:
Identification of specific cellular subsets that express/respond to IL-36
Characterization of variable penetrance of IL36RN mutations at cellular level
Discovery of compensatory mechanisms in individual cells
Disease-relevant applications:
Profiling of skin lesions from GPP patients with/without IL36RN mutations
Comparison of transitional states between healthy and inflamed tissue
Monitoring of treatment responses at single-cell resolution
Methodological approaches:
scRNA-seq to identify transcriptional signatures
CyTOF for protein-level phenotyping
Spatial transcriptomics to preserve tissue context
Single-cell ATAC-seq for epigenetic regulation
Analytical considerations:
Trajectory analysis to map inflammatory progression
Integration with genetic data
Cell-cell interaction modeling
Pseudo-time analysis of IL-36 pathway activation
These technologies could resolve longstanding questions about cell-specific roles in IL36RN-related pathology that bulk approaches cannot address.
The relationship between IL36RN genotype and clinical phenotype shows distinctive patterns:
Clinical subtype associations:
Treatment implications:
IL36RN mutation carriers may respond differently to conventional treatments
Targeted IL-36 pathway inhibitors potentially more effective in mutation carriers
Genotyping could inform personalized therapeutic approaches
Research design considerations:
Retrospective analysis of treatment outcomes stratified by mutation status
Prospective clinical trials incorporating IL36RN genotyping
Development of mutation-specific biomarkers for treatment monitoring
A comprehensive approach should include:
Standardized clinical assessment protocols
Long-term follow-up to capture recurrence patterns
Pharmacogenetic analyses
Correlation with other genetic factors (HLA status, CARD14 mutations)
Developing representative disease models requires multiple complementary approaches:
Cellular models:
Primary keratinocytes from IL36RN-mutant patients
CRISPR-engineered cell lines with specific patient mutations
3D skin equivalents incorporating IL36RN-deficient keratinocytes
Co-culture systems with immune cells to model inflammatory cascade
Animal models:
Conventional IL36RN knockout mice
Humanized mouse models expressing patient-specific mutations
Conditional tissue-specific IL36RN deletion
Xenograft models using patient-derived cells
Ex vivo approaches:
Skin explant cultures from patient biopsies
Precision-cut tissue slices with ex vivo drug testing
Microfluidic organ-on-chip technologies
Validation criteria:
Recapitulation of histological features of pustular psoriasis
Similar transcriptional profiles to patient lesions
Appropriate inflammatory cell recruitment
Response to established therapies mirroring clinical observations
These models provide platforms for both mechanistic studies and preclinical therapeutic testing.
Meta-analysis data reveals important distribution patterns:
Patient Group | Total Patients | GPP Alone | GPP+PV | Adult GPP | Pediatric GPP |
---|---|---|---|---|---|
All studies combined | 683 | 332 | 351 | Not fully specified | Not fully specified |
XiuYan Li (2014) | 62 | 17 | 45 | 46 | 16 |
XiaoHua Wang (2017) | 41 | 17 | 24 | Not specified | Not specified |
ZhongTao Li (2018) | 43 | 24 | 19 | Not fully specified | 18 |
Wang (2015) | 83 | 29 | 54 | Not specified | Not specified |
Sugiura (2013) | 31 | 11 | 20 | 23 | 8 |
Key findings include:
Nearly equal distribution between GPP alone (332) and GPP+PV (351) in the total cohort
Significantly higher frequency of IL36RN mutations in GPP alone vs. GPP+PV
This data supports distinct genetic mechanisms in GPP subtypes and highlights the importance of standardized patient classification in research studies.
Molecular characterization of IL36RN mutations shows population-specific patterns:
Notable observations:
Some mutations (c.28C>T) show population-specific prevalence
Multiple studies used RFLP-PCR and Sanger sequencing, providing methodological consistency
A polymorphism of c.115+6T>C specifically led to IL36RN mutation in 393 cases
This mutation spectrum informs targeted genotyping approaches and highlights potential founder effects in different populations.
Investigating endogenous IL-36 regulation requires multi-faceted approaches:
Transcriptional regulation:
Characterization of promoter elements controlling IL36RN expression
Identification of transcription factors regulating IL36RN
Analysis of epigenetic modifications affecting IL36RN expression
Post-translational processing:
Pathway integration:
Novel methodologies:
CRISPR screens to identify regulatory genes
Proximity labeling to identify protein-protein interactions
Advanced imaging to track IL-36 processing and trafficking
Data shows that stimuli like IL-1β, TNF, PMA, and flagellin induce IL-36α and IL-36γ expression in epithelial cells, with IL-36γ being the most abundant . Understanding these regulatory mechanisms may reveal new therapeutic targets.
IL36RN research has significant implications for precision dermatology:
Diagnostic applications:
Development of targeted genotyping panels for GPP patients
Creation of risk prediction algorithms incorporating IL36RN status
Identification of biomarkers correlating with IL36RN function
Therapeutic stratification:
Selection of biologics based on IL36RN mutation status
Dose optimization guided by genetic profile
Timing of therapeutic intervention (earlier for high-risk genotypes)
Novel therapeutic approaches:
Development of recombinant IL-36Ra variants resistant to degradation
Small molecule stabilizers of mutant IL-36Ra
Gene therapy to correct IL36RN mutations
IL-36R-targeted monoclonal antibodies
Research infrastructure needs:
Integration of genotypic and phenotypic data in clinical registries
Biobanking initiatives for IL36RN-mutated patient samples
Collaborative networks for studying rare genotype combinations
The strong genetic association between IL36RN mutations and specific GPP subtypes provides a foundation for more targeted therapies in this difficult-to-treat condition.
IL-36RA is a secreted protein that antagonizes the proinflammatory signals of the IL-36 family members, which include IL-36α, IL-36β, and IL-36γ . The IL-36 family belongs to the larger IL-1 superfamily and consists of three agonists (IL-36α/β/γ), one antagonist (IL-36RA), one cognate receptor (IL-36R), and one accessory protein (IL-1RAcP) . The receptor activation follows a two-step mechanism where the agonist first binds to IL-36R, and the resulting binary complex recruits IL-1RAcP. This assembled ternary complex brings together intracellular TIR domains of receptors, which activate downstream NF-κB and MAPK signaling .
IL-36RA is normally expressed at low levels but is induced upon stimulation. It acts on various cells, including epithelial and immune cells . In the skin, IL-36RA contributes to host defense through inflammatory responses. However, when dysregulated, IL-36RA stimulates keratinocytes and immune cells to enhance the Th17/Th23 axis, leading to psoriatic-like skin disorders . Genetic mutations in IL-36RA are associated with generalized pustular psoriasis, a rare but life-threatening skin disease .
Deficiency of IL-36RA (DITRA) is a life-threatening autoinflammatory disease caused by autosomal-recessive mutations in the IL36RN gene . Patients with DITRA develop recurrent episodes of generalized pustular psoriasis (GPP) with systemic inflammation and fever . Biological treatments targeting inflammatory cytokines, such as TNF-alpha, IL-12/23, and IL-17, have shown varying degrees of efficacy in treating DITRA .
Anti-IL-36 antibodies have been shown to attenuate IMQ or IL-23 induced skin inflammation in mice, illustrating IL-36’s involvement in mouse models of psoriasis . Further research and clinical trials are warranted to explore the therapeutic potential of IL-36RA in treating inflammatory and autoimmune diseases.