IL10RB Human refers to the interleukin-10 receptor beta subunit, a transmembrane protein encoded by the IL10RB gene. It is a critical component of the interleukin-10 (IL-10) receptor complex and plays a pivotal role in modulating immune responses and cytokine signaling. As an accessory chain, IL10RB is essential for activating downstream pathways in response to IL-10, IL-22, IL-26, IL-28, and interferon lambda (IFNλ) .
Gene Location: IL10RB is located on chromosome 21q22.11, forming a cluster with other class II cytokine receptor genes (IFNAR1, IFNAR2, IFNGR2) .
Protein Structure: The 325-amino-acid protein belongs to the cytokine receptor family. It contains an extracellular domain with two fibronectin type-III repeats and a cytoplasmic tail critical for signal transduction .
Functional Partners: Forms heterodimers with IL10RA (IL-10 receptor) and IFNLR1 (IFNλ receptor) to mediate cytokine signaling .
IL10RB, in complex with IL10RA, binds IL-10 to suppress pro-inflammatory cytokines (e.g., TNF-α, IL-6) and limit tissue damage. This anti-inflammatory axis is critical in autoimmune diseases and chronic inflammation .
IL10RB/IFNLR1 heterodimers mediate IFNλ signaling, inducing antiviral genes (e.g., MX1, OAS1) in epithelial cells. This pathway is vital for mucosal defense but may contribute to barrier disruption in viral infections .
Inflammatory Bowel Disease (IBD): IL10RB mutations linked to early-onset, severe IBD due to impaired IL-10 signaling .
Systemic Lupus Erythematosus (SLE): Specific IL10RB haplotypes (e.g., CAA) correlate with SLE susceptibility and clinical manifestations .
Hepatitis B: Genetic variants may influence susceptibility to viral persistence .
Higher Expression: Elevated IL10RB levels in blood correlate with severe COVID-19 outcomes, including respiratory failure and end-organ damage .
Viral Load: In vitro overexpression increases SARS-CoV-2 replication, suggesting a dual role in immune evasion and tissue injury .
Cohort | IL10RB Expression Level | Outcome Association | Reference |
---|---|---|---|
Mount Sinai Biobank | High | End-organ damage, mortality | |
MVP (EUR/AFR) | Increased GReX | Respiratory failure, tracheostomy |
Mucosal Immunity: IL10RB’s restricted expression in epithelial cells ensures localized IFNλ responses, limiting systemic inflammation .
Cancer: Dysregulated IL10RB may modulate tumor microenvironments, though direct evidence remains limited .
Reported Variants: 41 public variants, including missense, nonsense, and splice-site mutations .
Disease Links: Rare pathogenic variants associated with early-onset IBD and immune dysregulation .
Interleukin-10 receptor subunit beta, IL-10 receptor subunit beta, IL-10R subunit beta, IL-10RB, Cytokine receptor class-II member 4, Cytokine receptor family 2 member 4, CRF2-4, Interleukin-10 receptor subunit 2, IL-10R subunit 2, IL-10R2, CDw210b, IL10RB.
Sf9, Insect cells.
MVPPPENVRM NSVNFKNILQ WESPAFAKGN LTFTAQYLSY RIFQDKCMNT TLTECDFSSL SKYGDHTLRV RAEFADEHSD WVNITFCPVD DTIIGPPGMQ VEVLADSLHM RFLAPKIENE YETWTMKNVY NSWTYNVQYW KNGTDEKFQI TPQYDFEVLR NLEPWTTYCV QVRGFLPDRN KAGEWSEPVC EQTTHDETVP SLEPKSCDKT HTCPPCPAPE LLGGPSVFLF PPKPKDTLMI SRTPEVTCVV VDVSHEDPEV KFNWYVDGVE VHNAKTKPRE EQYNSTYRVV SVLTVLHQDW LNGKEYKCKV SNKALPAPIE KTISKAKGQP REPQVYTLPP SRDELTKNQV SLTCLVKGFY PSDIAVEWES NGQPENNYKT TPPVLDSDGS FFLYSKLTVD KSRWQQGNVF SCSVMHEALH NHYTQKSLSL SPGKHHHHHH.
IL10RB, also known as IL-10 R beta or IL-10 R2, functions as the signaling subunit of the IL-10 receptor heterotetramer complex. The human IL10RB gene encodes a 325 amino acid type I transmembrane protein with a 20 aa signal sequence, a 200 aa extracellular region, a 29 aa transmembrane segment, and a 76 aa cytoplasmic domain. The extracellular region contains two 100 aa subdomains resembling immunoglobulin constant regions, responsible for its alternative designation as CRF2-4 (cytokine receptor family class II/2 member 4). While IL10RB does not bind IL-10 independently, it is essential for signal transduction after IL-10 binds to the IL-10 R alpha subunit, activating the JAK1, TYK2, and STAT3 signaling cascade .
Unlike IL10RA (IL-10 R alpha), which is the ligand-binding subunit specific to IL-10, IL10RB is shared by receptors for multiple type-II cytokines including IL22, IL26, and interferon lambda (INFλ). IL10RA expression varies across cell types and can be upregulated upon cellular activation, whereas IL10RB is constitutively expressed in most cell types . Upon IL-10 binding to IL10RA, IL10RB undergoes a conformational change allowing it to also engage with IL-10, forming a complete signaling complex. This structural reorganization is necessary for activating the downstream signaling pathway involving JAK1, TYK2, and STAT3 .
Several complementary methodologies can be employed to detect IL10RB expression:
Flow cytometry: PE-conjugated monoclonal antibodies (such as Clone #90220) enable detection of IL10RB on cell surfaces. When analyzing human PBMC monocytes, comparison with isotype control antibodies (e.g., Catalog # IC002P) is essential for distinguishing specific staining .
Quantitative PCR: For measuring IL10RB gene expression at the mRNA level, with appropriate housekeeping genes for normalization.
Western blotting: For detecting IL10RB protein in cell lysates, requiring optimization of lysis conditions to preserve membrane proteins.
Immunohistochemistry/immunofluorescence: For visualizing IL10RB distribution in tissue contexts.
For reliable results, researchers should implement multiple detection methods and include appropriate controls. When using flow cytometry, proper compensation and matched isotype controls are critical for accurate interpretation.
Several strategies can be employed to manipulate IL10RB signaling:
Neutralizing antibodies: Anti-IL10RB antibodies can block receptor function in cellular assays.
RNA interference: siRNA or shRNA targeting IL10RB for transient knockdown.
CRISPR-Cas9 gene editing: For generating stable knockout cell lines or animal models.
Conditional knockout models: Using Cre-loxP systems for tissue-specific deletion of IL10RB, as demonstrated in studies with CD11c-Cre and LysM-Cre systems .
Receptor stimulation: Recombinant IL-10 administration to activate the receptor complex.
Pathway inhibitors: Small molecules targeting downstream components like JAK1, TYK2, or STAT3.
When designing such experiments, researchers should validate the efficacy of their intervention through functional assays and consider potential compensatory mechanisms that might affect interpretation.
Working with primary human cells requires specific considerations:
Cell isolation: Minimize processing time and mechanical stress during PBMC isolation.
Culture conditions: Prior to experimental use, rest cells for 2-4 hours in serum-containing media to recover from isolation stress.
Flow cytometry: For membrane proteins like IL10RB, include appropriate blocking steps with human serum or Fc receptor blocking reagents to prevent non-specific binding.
Stimulation conditions: Consider using TLR ligands which can alter receptor expression and responsiveness.
Cryopreservation: If storage is necessary, use controlled-rate freezing and media containing 10% DMSO.
Donor variability: Include sufficient biological replicates to account for genetic and environmental heterogeneity.
These optimizations help ensure reproducible results when studying IL10RB in primary human cells.
Deleterious mutations in IL10RB are strongly associated with very early-onset inflammatory bowel disease (VEO-IBD), typically presenting within the first months of life . These mutations disrupt IL-10 signaling, which is critical for maintaining intestinal immune homeostasis. The causative relationship is supported by animal models, as IL10 and IL10R-deficient mice develop spontaneous intestinal inflammation . Since IL10RB is shared by other cytokine receptors (IL22, IL26, and INFλ), mutations may affect multiple immune pathways, contributing to the complex pathogenesis of IBD.
When analyzing patient samples for IL10RB mutations, researchers should sequence the complete coding region and conduct functional assays to determine the impact of novel variants on signaling capacity. The severity and early onset of disease in patients with IL10RB mutations highlight the non-redundant role of this pathway in intestinal immune regulation.
IL10RB signaling affects multiple cell types in mucosal tissues to maintain immune homeostasis:
Dendritic cells: Loss of IL10R signaling in DCs leads to enhanced secretion of proinflammatory cytokines like IL6 and TNF upon stimulation .
Macrophages: IL10R-deficient macrophages show increased production of inflammatory mediators in response to microbial stimuli .
T cells: IL10RB signaling in regulatory T cells is important for their suppressive function.
Epithelial cells: IL10RB expression on intestinal epithelial cells suggests a direct role in maintaining barrier function.
The ubiquitous expression of IL10RB creates a network of IL-10 responsive cells that collectively maintain intestinal homeostasis. Research designs should consider this multi-cellular involvement when investigating IL10RB-dependent processes in mucosal immunity.
To study IL10RB-microbiota interactions:
Germ-free animal models: Compare IL10RB-deficient animals in germ-free versus conventional housing to determine microbiota dependence of phenotypes.
Controlled colonization experiments: Introduce defined bacterial communities to determine species-specific effects on IL10RB signaling.
16S rRNA sequencing: Characterize microbiota composition in models with altered IL10RB signaling.
Metatranscriptomics: Assess changes in microbial gene expression in response to altered host IL10RB signaling.
Ex vivo co-culture systems: Test how bacterial species affect IL10RB-expressing cells isolated from different intestinal compartments.
Intestinal organoids: Examine epithelial IL10RB responses to microbial products in a controlled system.
These approaches can reveal how microbial recognition through pattern recognition receptors induces IL10 production, creating a negative feedback loop to prevent excessive inflammation. Careful control of experimental variables is essential due to the complex nature of host-microbiota interactions.
The structural dynamics of IL10RB are critical for its signaling function:
Upon IL-10 binding to IL10RA, IL10RB undergoes a conformational change that enables it to engage with IL-10, completing the receptor complex .
The extracellular immunoglobulin-like domains are crucial for these protein-protein interactions.
Researchers investigating structure-function relationships should consider:
Site-directed mutagenesis of key residues in different domains
Structural biology techniques like X-ray crystallography or cryo-EM
Proximity-based assays to monitor real-time conformational changes
Molecular dynamics simulations to predict mutation effects
Understanding these structural mechanisms provides insights into how genetic variants might affect signaling and offers potential targets for therapeutic intervention.
Despite ubiquitous IL10RB expression, signaling outcomes vary considerably between cell types due to:
Differential expression of IL10RA and other receptor components
Varying levels of JAK/STAT pathway components
Cell-specific chromatin accessibility at IL-10-responsive genes
Cross-talk with other signaling pathways
For example, in dendritic cells, IL10R signaling inhibits maturation and cytokine production, whereas in macrophages, it can promote anti-inflammatory phenotypes . In T cells, effects depend on the specific subset and activation state. These variations necessitate cell-type specific analyses when studying IL10RB signaling, ideally using purified populations or single-cell approaches to avoid masking important heterogeneity.
To study signal integration involving IL10RB:
Simultaneous cytokine stimulation experiments: Assess how IL10RB-dependent signals modulate responses to other cytokines.
Phospho-flow cytometry: Track activation of shared signaling molecules like STAT3 in response to multiple stimuli.
Temporal studies: Examine how pre-treatment with one cytokine affects responses to subsequent stimulation.
Transcriptional profiling: Identify common and unique gene sets regulated by different cytokines sharing IL10RB.
Systems biology approaches: Apply mathematical modeling to predict pathway interactions.
Selective receptor component blockade: Use antibodies or genetic approaches to block specific receptor chains while leaving others functional.
These approaches help decipher how IL10RB participates in the complex network of cytokine signaling that regulates immune responses.
Several technical challenges affect IL10RB detection:
Challenge | Recommended Solution |
---|---|
Low surface expression | Use high-sensitivity detection methods; consider signal amplification techniques |
Epitope masking by protein interactions | Try multiple antibody clones targeting different epitopes |
Processing-induced changes | Analyze fresh samples when possible; standardize processing times |
High background in flow cytometry | Optimize blocking conditions; use appropriate isotype controls |
Inconsistent quantification | Implement quantitative flow cytometry with calibration beads |
Donor-to-donor variability | Increase biological replicates; normalize to stable reference markers |
Validation across multiple detection platforms and careful optimization of protocols for each cell type under study are essential for reliable IL10RB expression analysis.
Distinguishing between cytokines sharing IL10RB requires careful experimental design:
Use neutralizing antibodies specific to individual cytokines or their unique receptor components
Compare responses in cells with selective receptor component knockouts
Employ recombinant cytokines with altered receptor binding specificities
Analyze temporal response patterns that may differ between cytokines
Examine downstream gene expression signatures unique to each signaling pathway
Conduct experiments in defined media conditions to eliminate confounding cytokines
Consider using cytokine-deficient mouse models for in vivo studies
These approaches help isolate IL-10-specific effects from those of other cytokines that utilize IL10RB as a signaling component.
For studying IL10RB in complex tissues:
Single-cell RNA sequencing: Reveals cell-type specific expression patterns and response heterogeneity
Spatial transcriptomics: Maps IL10RB expression within tissue architecture
Multiplexed immunofluorescence: Visualizes IL10RB alongside cell type markers and signaling components
Mass cytometry (CyTOF): Simultaneously measures IL10RB with dozens of other proteins
Cell sorting prior to analysis: Enables focused study of specific populations
Laser capture microdissection: Isolates regions of interest for molecular analysis
In situ proximity ligation assays: Detects IL10RB interactions with other proteins in tissue context
These technologies overcome the limitations of bulk analysis methods that can mask important cellular heterogeneity in IL10RB expression and function within tissues.
Emerging evidence suggests epigenetic regulation plays a significant role in IL10RB function:
DNA methylation patterns at the IL10RB promoter can influence expression levels
Histone modifications affect chromatin accessibility at IL10RB-responsive genes
Non-coding RNAs may post-transcriptionally regulate IL10RB expression
Epigenetic reprogramming during cellular differentiation can alter IL10RB responsiveness
Researchers investigating epigenetic aspects should consider:
Chromatin immunoprecipitation sequencing (ChIP-seq) to map histone modifications
Bisulfite sequencing to analyze DNA methylation patterns
ATAC-seq to assess chromatin accessibility
Functional studies using epigenetic modifying compounds
Understanding these mechanisms could reveal new therapeutic approaches targeting IL10RB pathway regulation rather than the receptor itself.
Several therapeutic strategies targeting IL10RB signaling show potential:
Recombinant IL-10 or stabilized IL-10 analogs to enhance anti-inflammatory signaling
Cell-specific delivery systems to target IL-10 to relevant tissues
Gene therapy approaches to correct IL10RB mutations in VEO-IBD
Small molecule modulators of downstream signaling components
Combination therapies targeting multiple aspects of the pathway
Microbiome-based interventions that promote IL-10 production
When developing such therapies, researchers must consider:
Potential effects on other cytokine pathways sharing IL10RB
Cell-type specific responses that may influence efficacy and side effects
Patient stratification based on IL10RB pathway biomarkers
Delivery strategies to ensure targeting of relevant tissues
Therapeutic success will likely require personalized approaches based on individual patient genetics and disease mechanisms.
The interplay between IL10RB signaling and cellular metabolism represents an emerging research frontier:
IL-10 signaling through IL10RB influences metabolic pathways in macrophages, shifting from glycolysis toward oxidative phosphorylation
Metabolic state of cells affects their responsiveness to IL-10 and expression of IL10RB
Tissue-specific metabolic environments may modulate IL10RB function
Mitochondrial dynamics appear to be regulated by and regulate IL10RB signaling
Researchers investigating these interactions should consider:
Metabolomic profiling alongside IL10RB functional assays
Real-time metabolic flux analysis in IL10RB-sufficient versus deficient cells
How nutrient availability affects IL10RB signaling outcomes
The potential for metabolic interventions to enhance IL10RB signaling in inflammatory conditions
This emerging field may reveal new therapeutic targets at the interface of immunity and metabolism.
Interleukin-10 receptor beta (IL-10RB), also known as IL-10R2, is a crucial component of the interleukin-10 receptor complex. It plays a significant role in the immune system by mediating the anti-inflammatory effects of interleukin-10 (IL-10). This receptor is part of the cytokine receptor family and is essential for the proper functioning of the immune response.
IL-10RB is a type II cytokine receptor that is ubiquitously expressed in various cell types. It is a shared receptor subunit for several cytokines, including IL-10, IL-22, IL-26, IL-28, and IFN-λ. The receptor complex is tetrameric, consisting of two alpha (IL-10RA) and two beta (IL-10RB) subunits .
The IL-10RB subunit functions as the signaling component of the receptor complex. Upon binding of IL-10 to the IL-10RA subunit, a conformational change occurs in IL-10RB, allowing it to bind to IL-10 as well. This interaction leads to the formation of a heterotetrameric complex that activates the JAK1 and TYK2 kinases . These kinases phosphorylate the IL-10RA subunit, recruiting and phosphorylating STAT3, which then dimerizes and translocates to the nucleus to initiate gene transcription .
Recombinant human IL-10RB is produced using various expression systems, including insect cells and mammalian cells. The recombinant protein is typically glycosylated and has a molecular mass of approximately 50-70 kDa . It is used in research to study the signaling pathways and biological functions of IL-10 and other related cytokines.
IL-10RB is essential for the anti-inflammatory effects of IL-10, which include the inhibition of pro-inflammatory cytokine production and the suppression of antigen presentation. The receptor is also involved in the regulation of gastrointestinal immune responses, particularly in the mucosal layer . Defects in IL-10RB have been associated with early onset inflammatory bowel disease (IBD) and other immune-related disorders .
Mutations in the IL10RB gene can lead to severe immune dysregulation and increased susceptibility to infections and inflammatory diseases. Understanding the structure and function of IL-10RB is crucial for developing therapeutic strategies targeting IL-10 signaling pathways. Recombinant IL-10RB is also used in various assays to study its interactions with other cytokines and to develop potential treatments for immune-related disorders .