IL 22 Mouse, PEG

Interleukin-22 Mouse Recombinant, Pegylated
Shipped with Ice Packs
In Stock

Description

Pegylated Interleukin-22 Mouse Recombinant produced in E.Coli is a single, non-glycosylated homodimeric polypeptide chain containing 147 amino acids and an aditional Ala amino acid at N-terminus having a molecular mass of 36 kDa as determioned by mass spectometry. However due to enlarged hydrodymanic volume it runs on the SDS-PAGE as a 50 kDa protein and in gel-filtration on Superdex 200 as over 200 kDa protein. The Murine IL-22 is Mono-pegylated (with 20 kDa PEG) purified by proprietary chromatographic techniques.

Product Specs

Introduction
Interleukin-22 (IL-22), also called IL-10-related T cell-derived inducible factor (IL-TIF), is a cytokine initially discovered in mouse T cells and mast cells. Its production is stimulated by IL-9. The mouse IL-22 protein is 179 amino acids long, including a 33 amino acid signal peptide. After cleavage of this signal peptide, the mature protein is 147 amino acids long. Mouse IL-22 shares about 79% and 22% amino acid sequence identity with human IL-22 and IL-10, respectively. IL-22 has been observed to activate STAT-1 and STAT-3 in several hepatoma cell lines and increase the production of acute phase proteins. In normal mouse T cells, IL-22 is produced upon Con A activation. Additionally, mouse IL-22 expression is induced in various organs following lipopolysaccharide injection, suggesting a potential role in inflammatory responses. The functional IL-22 receptor complex is composed of two receptor subunits: IL-22R (previously known as the orphan receptor CRF2-9) and IL-10Rβ (previously known as CRF2-4). These receptors belong to the class II cytokine receptor family.
Description
Recombinant pegylated mouse Interleukin-22 is produced in E. coli. It is a non-glycosylated homodimeric polypeptide chain comprised of 147 amino acids, with an additional alanine residue at the N-terminus. Mass spectrometry analysis determined its molecular mass to be 36 kDa. However, due to its large hydrodynamic volume, it appears as a 50 kDa protein on SDS-PAGE and as a protein exceeding 200 kDa on gel filtration using Superdex 200. This murine IL-22 is mono-pegylated with a 20 kDa PEG and is purified using proprietary chromatographic techniques.
Physical Appearance
White, lyophilized (freeze-dried) powder, sterile-filtered.
Formulation
Lyophilized from a 0.65 mg/ml solution containing 0.003 mM NaHCO3.
Solubility
To reconstitute the lyophilized pegylated mouse Interleukin-22, it is recommended to dissolve it in sterile 0.4% NaHCO3 adjusted to a pH of 8-9, at a concentration of at least 100 µg/ml. This solution can be further diluted with other aqueous solutions as needed.
Stability
Lyophilized pegylated murine IL-22 remains stable at room temperature for several days, but for long-term storage, it should be kept desiccated at temperatures below -20°C. After reconstitution at a concentration of 0.1 mg/ml to 2 mg/ml, followed by filtration and sterilization, the protein can be stored at 4°C for several weeks, making it suitable for long-term infusion studies using osmotic pumps. For storage at lower concentrations, it is recommended to add a carrier protein such as 0.1% HSA or BSA. Avoid repeated freeze-thaw cycles.
Purity
Purity exceeds 98.0% as determined by: (a) Gel-Filtration Analysis, and (b) SDS-PAGE Analysis.
Biological Activity
The ED50 was determined by a STAT3 phosphorylation assay using HepG cells. The in vitro activity of pegylated mouse IL-22 was found to be approximately 10% of the activity observed for non-pegylated mouse IL-22.
Synonyms
IL-TIF, TIFa, IL-10-related T-cell-derived-inducible factor, IL-22, ILTIF, IL-D110, zcyto18, MGC79382, MGC79384, TIFIL-23.
Source
Escherichia Coli.

Q&A

What are the primary tissue targets of IL-22 in mice and how do they compare to humans?

IL-22 primarily targets epithelial cells in barrier tissues, with significant effects observed in the pancreas, colon, liver, and skin of mice. These tissues express varying levels of the IL-22 receptor complex components, particularly IL-22Rα and IL-10Rβ, which determines their responsiveness to IL-22 signaling. Research has shown that IL-10Rβ expression is highest in the mouse pancreas, followed by the colon, skin, and liver, which directly correlates with tissue responsiveness to engineered IL-22 variants . This differential expression pattern explains tissue-selective signaling responses observed in murine studies. Human and mouse IL-22 receptor distribution patterns share similarities, though species-specific differences in receptor expression levels may exist across certain tissues, which researchers should consider when translating findings from mouse models to human applications.

How does IL-22 signaling differ from other interleukins in mouse models?

IL-22 exhibits several unique signaling characteristics that distinguish it from other interleukins in mouse models. First, IL-22 selectively targets non-hematopoietic cells expressing IL-22Rα, predominantly epithelial cells, unlike many other interleukins that target immune cells . Second, IL-22 activates both STAT1 and STAT3 pathways with distinct activation thresholds, with STAT3 typically requiring lower signaling strength than STAT1 for activation . This creates a dual functionality where low levels of IL-22 signaling can selectively activate STAT3-mediated tissue protection pathways without triggering STAT1-mediated inflammatory responses. Additionally, IL-22 utilizes the shared IL-10Rβ receptor subunit, which is also employed by IL-10 family cytokines including IL-10 and IL-26, creating potential for receptor competition in experimental settings. These unique characteristics necessitate careful experimental design when studying IL-22 in comparison to other interleukins.

What are the key STAT-dependent gene expression programs induced by IL-22 in mouse tissues?

IL-22 induces distinct STAT-dependent gene expression programs in mouse tissues, which can be categorized into protective and inflammatory responses:

STAT3-dependent programs (tissue protective):

  • Antimicrobial peptide genes (e.g., S100 family proteins, defensins)

  • Tissue repair and regeneration genes (e.g., Reg3g, Reg3b)

  • Anti-apoptotic genes (e.g., Bcl2, Bcl-xL)

  • Mucus production genes in intestinal tissues

STAT1-dependent programs (pro-inflammatory):

  • Interferon-stimulated genes (ISGs) including Isg15, Apobec, and Tmem173 (STING)

  • Pro-inflammatory TNF family members (Tnfsfr1b/Tnfr2, Tnfsf10/Trail)

  • Chemokines such as Cxcl1, which contributes to pathological effects in the GI tract

Research has demonstrated that engineered IL-22 variants like 22-B3 can selectively induce STAT3-dependent tissue protective genes while avoiding STAT1-mediated pro-inflammatory gene expression . This selective activation pattern explains how modified IL-22 variants can promote epithelial repair without triggering inflammatory cascades in mouse models.

How do different IL-10Rβ expression levels across mouse tissues affect IL-22 signaling outcomes?

Differential IL-10Rβ expression is a critical determinant of tissue-specific IL-22 signaling outcomes in mice. Research has established a clear correlation between IL-10Rβ expression levels and the signaling profile elicited by both wild-type IL-22 and engineered variants . In tissues with high IL-10Rβ expression (pancreas), even engineered IL-22 variants with weakened IL-10Rβ binding (like 22-B3) can achieve sufficient receptor complex formation to activate both STAT3 and STAT1, resulting in only mild STAT3 bias. In tissues with intermediate IL-10Rβ expression (colon), the same variants exhibit strong STAT3-biased signaling, activating STAT3 robustly while failing to reach the threshold for STAT1 activation . In tissues with low IL-10Rβ expression (liver and skin), these variants cannot form sufficient receptor complexes to activate either STAT pathway, functioning as neutral antagonists .

This phenomenon has been experimentally verified through both in vivo phosphorylation studies across multiple tissues and in vitro studies where increasing IL-10Rβ expression levels reduced the STAT3 bias of engineered variants . These findings provide a mechanistic framework for understanding tissue-selective cytokine signaling and suggest that cellular modulation of IL-10Rβ expression may serve as a natural mechanism for tuning IL-22 responses in different physiological or pathological states.

What molecular mechanisms explain the different activation thresholds for STAT1 versus STAT3 downstream of the IL-22 receptor?

The different activation thresholds for STAT1 versus STAT3 downstream of the IL-22 receptor complex involve several molecular mechanisms that researchers should consider in experimental design:

  • Intrinsic binding affinity differences: STAT3 typically has higher affinity for phosphotyrosine sites on the IL-22 receptor than STAT1, allowing preferential STAT3 recruitment at lower signaling strengths .

  • Receptor complex stability requirements: Complete and stable IL-22Rα–IL-10Rβ complex formation is more critical for STAT1 activation than for STAT3 activation. Destabilizing this interaction through mutations in the IL-10Rβ binding interface selectively impairs STAT1 signaling while maintaining substantial STAT3 activation .

  • Signal amplification differences: The STAT3 pathway typically includes more robust signal amplification mechanisms than the STAT1 pathway, allowing effective signal transmission even with reduced receptor activation.

  • Feedback regulation: STAT1 and STAT3 are subject to different feedback regulation mechanisms, including distinct phosphatase recruitment patterns and negative regulators like SOCS proteins.

These mechanisms explain why partial agonists like 22-B2 and 22-B3, which destabilize the IL-22–IL-10Rβ interaction, effectively uncouple STAT3 and STAT1 activation . Understanding these differential thresholds enables researchers to design IL-22 variants with precise STAT activation profiles for specific experimental or therapeutic applications.

How does the structural configuration of the IL-22–IL-22Rα–IL-10Rβ ternary complex determine downstream signaling specificity?

The IL-22–IL-22Rα–IL-10Rβ ternary complex structure, resolved to 2.6 Å resolution, provides crucial insights into signaling specificity mechanisms . The complex exhibits a non-symmetric arrangement where IL-22 first binds with high affinity to IL-22Rα through site 1, followed by lower-affinity binding to IL-10Rβ through site 2, creating a functional signaling complex.

Key structural determinants of signaling specificity include:

  • The central role of IL-22 residue Tyr51 in the IL-10Rβ binding interface, which when mutated to alanine (Y51A) completely abolishes both STAT1 and STAT3 activation .

  • Peripheral contact residues like Glu117, Asn46, Gln116, Lys124, and Gln128, which when strategically mutated (as in variants 22-B1 through 22-B5) can calibrate signal strength to selectively activate STAT3 over STAT1 .

  • The relative positioning of the receptor intracellular domains, which determines the efficiency of Janus kinase (JAK) activation and subsequent STAT recruitment patterns.

Crystallographic studies using engineered high-affinity IL-22 super-agonists (Super-22a and Super-22b) were crucial for stabilizing the typically weak IL-22–IL-10Rβ interaction and enabling structure determination . This structural information provided the foundation for rational design of functionally selective IL-22 variants with calibrated STAT activation profiles. Understanding these structural determinants is essential for researchers designing experiments to investigate IL-22 signaling specificity or developing novel IL-22-based therapeutics with refined functional properties.

What approaches have been successful for engineering STAT-biased IL-22 agonists, and how do they compare?

Several successful approaches have been employed to engineer STAT-biased IL-22 agonists:

These approaches have generated IL-22 variants with distinct functional profiles:

The structure-guided approach has proven most effective for generating biased agonists, as it allows precise tuning of receptor complex stability to exploit different STAT activation thresholds while maintaining sufficient activity.

How can researchers verify STAT signaling bias of IL-22 variants across different experimental systems?

Verifying STAT signaling bias of IL-22 variants requires a multi-faceted approach across different experimental systems:

  • Cell-based phosphorylation assays:

    • Measure phosphorylation of STAT1 (Y701) and STAT3 (Y705) by western blot or phospho-flow cytometry

    • Calculate pSTAT3/pSTAT1 ratios across a concentration range to quantify bias

    • Test in multiple cell lines with different IL-10Rβ expression levels to assess context-dependence

  • Dose-response analysis:

    • Generate full dose-response curves for each STAT pathway

    • Calculate EC50 values and maximal responses to characterize both potency and efficacy differences

    • Compare bias factors across various IL-22 variants

  • Transcriptional profiling:

    • Analyze expression of STAT1-selective genes (e.g., ISGs like Isg15, Apobec, and Tmem173) versus STAT3-selective genes (e.g., antimicrobial peptides, tissue repair factors)

    • Use RNA-seq to comprehensively assess pathway activation

  • In vivo tissue analysis:

    • Examine pSTAT1 and pSTAT3 levels across multiple tissues after administration

    • Correlate signaling patterns with IL-10Rβ expression levels

    • Compare acute signaling (30 minutes) with gene expression changes (6-24 hours)

  • Receptor modulation studies:

    • Artificially increase or decrease IL-10Rβ levels to confirm mechanistic basis of bias

    • Test antagonistic properties by co-administration with wild-type IL-22

By systematically employing these complementary approaches, researchers can establish robust evidence for signaling bias and characterize how this bias translates across different biological contexts.

What are the critical quality control parameters for ensuring consistent functional properties of engineered IL-22 variants?

Ensuring consistent functional properties of engineered IL-22 variants requires rigorous quality control across multiple parameters:

  • Protein structural integrity:

    • Circular dichroism (CD) spectroscopy to confirm secondary structure preservation

    • Thermal stability assessment via differential scanning calorimetry (DSC) or thermal shift assays

    • Size-exclusion chromatography (SEC) to verify monomeric state and absence of aggregation

  • Receptor binding characteristics:

    • Surface plasmon resonance (SPR) to measure binding kinetics (kon, koff) and affinity (KD) for both IL-22Rα and IL-10Rβ

    • Cell-based binding assays with fluorescently labeled proteins to confirm native receptor recognition

    • Competition binding assays against wild-type IL-22

  • Functional consistency:

    • Standardized phospho-STAT3 and phospho-STAT1 assays in reference cell lines

    • Batch-to-batch comparison of STAT3/STAT1 activation ratios

    • Assessment of EC50 values across multiple preparations

  • Endotoxin control:

    • Endotoxin removal using techniques like the NoEndoHC Column Kit

    • Verification of endotoxin levels (<0.1 EU/mg) using the Chromogenic Endotoxin Quantification assay

    • Control experiments with heat-inactivated proteins to rule out endotoxin effects

  • Storage stability:

    • Accelerated stability testing at various temperatures

    • Activity retention assessment after freeze-thaw cycles

    • Formulation optimization to maintain functional properties

Researchers should establish specific acceptance criteria for each parameter based on intended applications, and systematically document these quality attributes across different production batches to ensure experimental reproducibility and reliable interpretation of biological effects.

What are the optimal experimental protocols for evaluating tissue-specific effects of IL-22 in mouse models?

Optimal protocols for evaluating tissue-specific effects of IL-22 in mouse models should incorporate multiple complementary approaches:

  • Administration route selection:

    • Intraperitoneal injection for systemic exposure assessment across multiple tissues

    • Local administration (e.g., intratracheal, rectal, or intradermal) for tissue-specific targeting

    • Genetic approaches using tissue-specific promoters for sustained expression

    • Standardize dosing based on protein activity rather than mass (typically 10-50 μg/mouse for acute studies)

  • Timepoint selection:

    • Acute signaling: 15-60 minutes post-administration for STAT phosphorylation analysis

    • Early gene expression: 2-6 hours for primary transcriptional responses

    • Functional changes: 24-72 hours for tissue remodeling and barrier function effects

    • Multiple timepoints to capture both immediate signaling and downstream biological consequences

  • Tissue collection and processing:

    • Flash freezing in liquid nitrogen for signaling analysis

    • RNA preservation in RNAlater for transcriptional profiling

    • Fixed tissue preparation for histological and immunofluorescence analysis

    • Single-cell preparations for flow cytometry or cell sorting

    • Standardization of tissue sections (e.g., proximal vs. distal colon)

  • Comprehensive readouts:

    • Western blot or ELISA for phospho-STAT quantification

    • RNA-seq or qPCR for gene expression profiling

    • Histological assessment for tissue architecture and cell composition

    • Functional assays (e.g., epithelial barrier integrity, antimicrobial activity)

    • Cytokine/chemokine profiling to assess secondary inflammatory responses

  • Controls and comparisons:

    • Wild-type IL-22 at equivalent doses for direct comparison

    • Vehicle controls matched for all excipients

    • IL-22 knockout mice or IL-22BP administration for loss-of-function analysis

    • Interferon controls to distinguish IL-22-specific from general STAT1-mediated effects

Adopting this multi-parameter approach enables comprehensive characterization of both the molecular mechanisms and physiological consequences of IL-22 signaling across different tissues.

How should PEGylation strategies be optimized for in vivo studies of IL-22 in mouse models?

Optimizing PEGylation strategies for IL-22 mouse studies requires careful consideration of several key parameters:

  • PEG conjugation site selection:

    • Map the IL-22 crystal structure to identify solvent-exposed residues distant from receptor binding interfaces

    • Avoid Tyr51, Glu117, and other residues critical for IL-10Rβ interaction

    • Consider introducing non-native cysteine residues at strategic positions for site-specific conjugation

    • Evaluate multiple conjugation sites to identify positions that preserve biological activity

  • PEG size and structure optimization:

    • Test linear PEGs ranging from 5-40 kDa to balance circulatory half-life extension with tissue penetration

    • Compare branched versus linear PEG structures for optimal pharmacokinetic profile

    • Consider releasable PEG linkages that can detach in target tissues

    • Systematically evaluate how different PEG sizes affect IL-22's ability to activate STAT3 versus STAT1 pathways

  • Activity characterization:

    • Compare EC50 values of PEGylated versus non-PEGylated IL-22 for both STAT3 and STAT1 activation

    • Determine if PEGylation introduces or modifies signaling bias

    • Assess tissue distribution patterns using fluorescently labeled PEG-IL-22

    • Measure circulatory half-life through time-course sampling and bioactivity assays

  • Formulation considerations:

    • Optimize buffer composition to prevent aggregation of PEGylated proteins

    • Evaluate freeze-thaw stability of PEGylated IL-22 preparations

    • Determine compatible administration routes (IV, IP, SC) based on formulation properties

    • Assess endotoxin removal efficiency from PEGylated preparations

Based on established cytokine PEGylation studies, researchers should expect PEGylation to extend IL-22's circulatory half-life from approximately 2-3 hours to potentially 24-48 hours depending on PEG size, which necessitates adjustment of dosing schedules in experimental protocols. The impact of PEGylation on tissue distribution should be carefully evaluated, as it may alter IL-22's ability to access certain tissue compartments, potentially enhancing systemic effects while reducing local tissue concentrations.

What approaches are most effective for measuring IL-22 pharmacokinetics and biodistribution in mouse models?

Effective measurement of IL-22 pharmacokinetics and biodistribution in mouse models requires a multi-modal approach:

  • Serum concentration determination:

    • ELISA: Develop sandwich ELISAs using anti-IL-22 antibodies that recognize epitopes preserved after potential modifications

    • Functional bioassays: Measure serum-induced STAT3 phosphorylation in reporter cell lines

    • Western blot: For size comparison of intact versus degraded IL-22 species

    • Mass spectrometry: For absolute quantification and modification site identification

    • Sample collection timepoints: 5min, 15min, 30min, 1h, 2h, 4h, 8h, 24h, 48h, 72h post-administration

  • Tissue biodistribution analysis:

    • Tissue homogenization and ELISA quantification across multiple organs

    • Immunohistochemistry to visualize tissue localization patterns

    • Flow cytometry to assess cellular association in disaggregated tissues

    • In vivo imaging using fluorescently labeled or radiolabeled IL-22

    • Single-cell analysis to identify specific cellular targets

  • Advanced tracking strategies:

    • Genetic tagging approaches (e.g., FLAG, HA, or His tags) for distinguishing exogenous from endogenous IL-22

    • Click chemistry-compatible IL-22 analogs for post-administration visualization

    • Complexation with IL-22BP to assess receptor-bound versus free IL-22 fractions

    • Receptor occupancy assays using fluorescent anti-receptor antibodies

  • Comparative analysis frameworks:

    • Wild-type versus engineered variants to correlate structural modifications with PK/PD properties

    • PEGylated versus non-PEGylated IL-22 to assess impact of half-life extension

    • Healthy versus disease models to evaluate context-dependent distribution

    • Dose-proportionality studies across a 10-100 fold concentration range

  • Mathematical modeling:

    • Compartmental PK modeling to determine volume of distribution, clearance, and half-life

    • Physiologically-based PK modeling to predict tissue concentrations

    • PK/PD integration to correlate exposure with downstream biological responses

    • Allometric scaling for translation between mouse data and other species

These approaches should be tailored based on the specific IL-22 variant being studied, as modifications like PEGylation or mutations affecting receptor binding will significantly alter pharmacokinetic properties and necessitate customized analytical strategies.

How does endogenous IL-22BP affect experimental outcomes in IL-22 mouse studies?

Endogenous IL-22BP (IL-22 binding protein) can significantly impact experimental outcomes in IL-22 mouse studies through several mechanisms:

  • Neutralization of administered IL-22:

    • IL-22BP binds IL-22 with approximately 1000-fold higher affinity than the IL-22 receptor complex

    • Endogenous IL-22BP can sequester exogenously administered IL-22, reducing effective concentrations

    • Expression levels vary across tissues and disease states, creating site-specific neutralization effects

    • Higher doses of IL-22 may be required to overcome IL-22BP neutralization in certain experimental contexts

  • Tissue-specific IL-22BP expression patterns:

    • Primarily expressed in lymphoid tissues, intestine, breast, and skin

    • Expression regulated by inflammatory stimuli and varies between homeostatic and diseased states

    • Differential expression creates tissue-specific sensitivity to exogenous IL-22

    • Changes in IL-22BP levels during disease progression can affect experimental reproducibility

  • Experimental design considerations:

    • Measure endogenous IL-22BP levels in target tissues to inform dosing strategies

    • Consider co-administration of IL-22BP-neutralizing antibodies for controlled IL-22 bioavailability

    • Use IL-22BP knockout mice to eliminate this variable in mechanistic studies

    • Evaluate binding affinity of engineered IL-22 variants to IL-22BP, as modifications may alter this interaction

  • Differential effects on IL-22 variants:

    • Engineered variants with modifications at the IL-10Rβ interface may retain IL-22BP binding

    • Super-agonists like Super-22a/b may compete more effectively with IL-22BP due to enhanced receptor binding

    • PEGylation may sterically hinder IL-22BP binding, potentially increasing bioavailability

    • Systematic evaluation of variant sensitivity to IL-22BP is essential for accurate interpretation of in vivo results

Researchers should systematically account for endogenous IL-22BP effects by measuring its levels in experimental tissues, considering genetic approaches to modulate its expression, and evaluating how different IL-22 variants interact with IL-22BP to ensure reproducible and interpretable experimental outcomes.

What are the most robust approaches for evaluating IL-22-mediated tissue protection versus inflammation in mouse models?

Robust evaluation of IL-22-mediated tissue protection versus inflammation requires comprehensive experimental approaches:

  • Challenge models with dual readouts:

    • Intestinal injury: Dextran sodium sulfate (DSS) colitis model - measure both epithelial integrity (protection) and immune cell infiltration (inflammation)

    • Liver injury: Acetaminophen or carbon tetrachloride models - assess both hepatocyte survival (protection) and inflammatory cytokine production

    • Pulmonary infection: Influenza challenge - evaluate barrier function and viral clearance versus inflammatory damage

    • Skin wound healing: Measure both re-epithelialization (protection) and inflammatory cell recruitment

  • Molecular pathway discrimination:

    • STAT3-dependent protective markers: Reg3γ, Reg3β, antimicrobial peptides, mucins, anti-apoptotic genes

    • STAT1-dependent inflammatory markers: ISGs (Isg15, STING), chemokines (Cxcl1), TNF family members (Tnfr2, Trail)

    • Transcriptional profiling to globally assess protective versus inflammatory gene signatures

    • Pathway inhibitors to selectively block STAT3 or STAT1 signaling components

  • Cellular and functional assessments:

    • Epithelial barrier integrity: FITC-dextran permeability, transepithelial electrical resistance, tight junction protein analysis

    • Tissue proliferation: Ki67 staining, BrdU incorporation, organoid formation efficiency

    • Inflammatory cell recruitment: Flow cytometry quantification of neutrophils, monocytes, T cells

    • Histological scoring systems that separately quantify tissue damage and inflammatory infiltration

  • Comparative interventions:

    • Wild-type IL-22 versus STAT3-biased variants (e.g., 22-B3) to dissociate protective and inflammatory effects

    • Combination with anti-inflammatory agents to assess additive versus redundant effects

    • Temporal separation of administration (prevention versus treatment paradigms)

    • Genetic approaches using conditional deletion of STAT3 versus STAT1 in epithelial cells

  • Translation-relevant biomarkers:

    • Serum biomarkers of epithelial damage (e.g., I-FABP, citrulline) versus inflammation (CRP, SAA)

    • Non-invasive imaging approaches for longitudinal assessment

    • Correlation of molecular markers with functional outcomes

    • Cross-validation in multiple disease models

This multi-parametric approach enables discrimination between the protective and inflammatory functions of IL-22, providing mechanistic insight into the context-dependent roles of this pleiotropic cytokine across different tissues and disease states.

How can data conflicts between acute versus chronic IL-22 exposure be reconciled in mouse models?

Reconciling data conflicts between acute versus chronic IL-22 exposure in mouse models requires systematic experimental approaches and careful interpretation:

  • Temporal transition mechanisms:

    • Track STAT activation kinetics over extended timeframes (minutes to weeks)

    • Monitor development of feedback inhibition via SOCS proteins and other negative regulators

    • Assess receptor downregulation/desensitization after repeated exposure

    • Evaluate changes in IL-22BP expression levels with prolonged IL-22 presence

  • Experimental design considerations:

    • Implement both pulse (single high-dose) and sustained (continuous low-dose) exposure paradigms

    • Utilize controlled-release systems (osmotic pumps, PEGylation, hydrogels) for consistent exposure

    • Compare genetic models (IL-22 transgenic mice) with pharmacological approaches

    • Design washout periods to assess reversibility of effects

  • Context-dependent outcome assessment:

    • Characterize differential responses across homeostatic versus injury settings

    • Evaluate tissue-specific transitions from protective to inflammatory states

    • Assess impact of concurrent inflammatory signals that may shift STAT1/STAT3 balance

    • Monitor pathological changes associated with prolonged exposure (fibrosis, dysplasia)

  • Mechanistic resolution approaches:

    • Use STAT3-biased IL-22 variants to determine if chronic inflammatory effects are STAT1-dependent

    • Implement conditional genetic systems for temporal control of specific pathway components

    • Combine IL-22 with pathway-specific inhibitors at different timepoints

    • Use computational modeling to predict signaling network adaptation over time

  • Reconciliation framework:

    • Establish clear temporal boundaries for protective versus pathological phases

    • Identify biomarkers predictive of transition from beneficial to detrimental effects

    • Develop integrated models incorporating dose, duration, tissue context, and disease state

    • Compare outcomes from IL-22 receptor partial agonists versus full agonists across acute and chronic settings

This comprehensive approach can help resolve apparent contradictions, revealing that acute IL-22 exposure primarily activates tissue-protective STAT3 pathways, while chronic exposure may lead to STAT1 pathway predominance, receptor desensitization, or activation of compensatory inflammatory mechanisms. Understanding these temporal dynamics is essential for translating IL-22 biology into therapeutic applications with appropriate dosing regimens.

What are the critical parameters for successful expression and purification of functional recombinant mouse IL-22?

Successful expression and purification of functional recombinant mouse IL-22 requires optimization of several critical parameters:

  • Expression system selection:

    • Mammalian expression (e.g., Expi293F cells): Provides proper folding and post-translational modifications, ideal for signaling studies

    • Baculovirus/insect cell system: Balances yield with proper folding, suitable for structural studies

    • E. coli: Highest yield but requires refolding protocols, appropriate for initial screening

    • Choose based on experimental requirements for activity, yield, and purity

  • Construct design optimization:

    • Signal peptide selection: HA signal peptide for mammalian systems, GP64 for baculovirus

    • Tag configuration: C-terminal 6xHis tag preserves N-terminal structure critical for function

    • Glycosylation management: Consider N68Q, N97Q mutations for structural studies to reduce heterogeneity

    • Codon optimization for expression system of choice

  • Purification strategy:

    • Immobilized metal affinity chromatography (IMAC): Ni-NTA resin for initial capture

    • Size exclusion chromatography (SEC): Critical for removing aggregates and ensuring monomeric state

    • Consider additional polishing steps (ion exchange, hydrophobic interaction) for highest purity

    • Buffer optimization to maintain stability (typically HEPES-buffered saline, pH 7.4)

  • Quality control parameters:

    • SDS-PAGE and western blot to confirm identity and purity

    • SEC-MALS to verify monomeric state and absence of aggregation

    • Endotoxin removal and testing (<0.1 EU/mg) for in vivo applications

    • Functional verification through cell-based STAT phosphorylation assays

  • Stability considerations:

    • Avoid repeated freeze-thaw cycles

    • Optimal storage concentration: 0.5-1 mg/mL

    • Addition of stabilizers: Consider 0.1% BSA for dilute solutions

    • Aliquoting strategy to maintain consistent freeze-thaw history across experiments

For structural studies requiring the complete receptor complex, co-expression or co-purification with receptor components may be necessary, as demonstrated in the IL-22–IL-22Rα–IL-10Rβ ternary complex crystallization, which required overnight incubation of purified components and carboxypeptidase treatment to achieve complex stability .

How can researchers address variability in IL-22 receptor expression across different mouse strains and tissues?

Addressing variability in IL-22 receptor expression across mouse strains and tissues requires systematic characterization and experimental design approaches:

  • Comprehensive receptor profiling:

    • Quantitative PCR to measure IL-22Rα and IL-10Rβ mRNA levels across tissues and strains

    • Western blot analysis with validated antibodies for protein-level confirmation

    • Flow cytometry of disaggregated tissues to assess cellular distribution patterns

    • Immunohistochemistry for spatial localization within tissue architecture

    • Single-cell RNA-seq to identify specific cell populations expressing receptor components

  • Standardization strategies:

    • Establish baseline receptor expression profiles for common mouse strains (C57BL/6, BALB/c, etc.)

    • Develop normalization procedures based on receptor expression for dose adjustment

    • Consider ex vivo stimulation of tissue explants to control for receptor variability

    • Use receptor overexpression or knockdown approaches to establish causal relationships

  • Experimental design considerations:

    • Include matched strain controls in all experiments

    • Pilot dose-finding studies for each strain/tissue combination

    • Report receptor expression data alongside functional outcomes

    • Consider receptor visualization techniques (e.g., fluorescently tagged IL-22) to confirm target engagement

  • Addressing functional consequences:

    • Correlate IL-10Rβ expression levels with STAT1/STAT3 activation ratios across tissues

    • Evaluate how strain differences in receptor expression affect therapeutic responsiveness

    • Assess sensitivity to engineered variants (e.g., 22-B3) across strains with different receptor profiles

    • Develop predictive models relating receptor levels to functional outcomes

  • Genetic approaches:

    • Generate receptor reporter mice for visualization of expression patterns

    • Consider conditional knockout models for specific receptor components

    • Use receptor humanization approaches for translational studies

    • Implement CRISPR-based approaches to equalize receptor expression for mechanistic studies

This systematic approach enables researchers to account for and leverage the natural variability in IL-22 receptor expression, transforming a potential confounding factor into an opportunity to understand context-dependent signaling mechanisms and improve translational predictions.

What techniques are most effective for distinguishing direct IL-22 effects on epithelial cells from indirect effects mediated by other cell types?

Distinguishing direct IL-22 effects on epithelial cells from indirect effects requires sophisticated experimental approaches:

  • Cell-specific receptor deletion models:

    • Generate conditional IL-22Rα knockout mice using epithelial-specific Cre lines (e.g., Villin-Cre for intestine, K14-Cre for skin)

    • Compare phenotypes with global IL-22Rα knockouts to identify epithelial-specific versus indirect effects

    • Use inducible systems (e.g., tamoxifen-inducible CreERT2) for temporal control

    • Implement cell-specific STAT3 deletion to distinguish direct signaling consequences

  • Ex vivo and in vitro approaches:

    • Isolate primary epithelial cells or organoids to assess direct responses in controlled environments

    • Conduct co-culture experiments with immune cells to identify paracrine signaling circuits

    • Use transwell systems to physically separate epithelial and immune compartments

    • Compare epithelial-only responses with those from complex tissue environments

  • Molecular and temporal discrimination:

    • Conduct rapid timepoint analysis (15-30 minutes) to capture direct signaling events before secondary responses

    • Utilize phospho-flow cytometry with epithelial markers to identify direct cellular targets

    • Perform single-cell RNA-seq to delineate cell-specific transcriptional responses

    • Use epithelial-specific reporters (e.g., STAT3-responsive elements driving fluorescent proteins in epithelial cells)

  • Pharmacological approaches:

    • Local administration to minimize systemic effects

    • Combination with cell-specific inhibitors to block potential indirect pathways

    • Use of STAT3-biased IL-22 variants (e.g., 22-B3) that preferentially target epithelial repair functions

    • Comparative studies with other epithelial-targeted cytokines (e.g., IL-20)

  • Human translation models:

    • Develop humanized mouse models with human epithelial grafts

    • Compare results between species-specific IL-22 variants

    • Utilize patient-derived organoids to confirm direct effects in human tissues

    • Correlate mouse findings with human biopsy analyses

These complementary approaches allow researchers to definitively attribute observed effects to direct IL-22 action on epithelial cells versus indirect mechanisms involving intermediate cell types or signaling molecules, providing clearer understanding of IL-22's biological functions and therapeutic potential.

How predictive are IL-22 mouse models for human therapeutic applications, and what are the key cross-species differences?

The predictive value of IL-22 mouse models for human applications must be evaluated in the context of several key cross-species differences:

What strategies can optimize the translational potential of engineered IL-22 variants from mouse to human studies?

Optimizing translational potential of engineered IL-22 variants from mouse to human studies requires multi-faceted strategies:

  • Structure-guided design with cross-species compatibility:

    • Focus engineering efforts on highly conserved regions of the IL-22-receptor interface

    • Validate mutations in both mouse and human IL-22 proteins in parallel

    • Confirm that key mutations (e.g., those in 22-B3) produce similar functional effects across species

    • Utilize the high-resolution structure of the IL-22–IL-22Rα–IL-10Rβ complex to guide rational design

  • Comprehensive cross-species validation:

    • Test engineered variants on both mouse and human cell lines

    • Compare STAT activation profiles and gene expression signatures

    • Validate in primary human tissues or organoids derived from target disease tissues

    • Evaluate species-specific differences in IL-22BP binding and neutralization

  • Targeted improvement of human-specific functions:

    • Optimize for human receptor binding while maintaining rodent activity for preclinical testing

    • Address human-specific immune recognition and immunogenicity concerns

    • Consider human-specific proteolytic processing and stability differences

    • Fine-tune STAT3/STAT1 bias ratios based on human tissue responses

  • Translational model development:

    • Generate humanized mice expressing human IL-22 receptor components

    • Develop surrogate molecules that engage mouse receptors but maintain human-optimized properties

    • Establish ex vivo human tissue systems for direct testing of human-optimized variants

    • Design mouse models that recapitulate human disease mechanisms

  • Pharmaceutical optimization:

    • Adapt PEGylation strategies based on human-specific pharmacokinetic requirements

    • Address human-specific manufacturing and stability challenges

    • Implement quality control assays that predict human biological activity

    • Develop formulations suitable for intended clinical administration routes

The translational success of engineered IL-22 variants depends critically on maintaining the core functional properties observed in mouse models while addressing species-specific differences. The structural conservation of the IL-22 receptor complex between species provides a solid foundation for translation, particularly for variants targeting the IL-10Rβ interface to modulate STAT signaling bias . This mechanistic approach, focused on conserved signaling pathways rather than specific sequence identity, enhances the probability of successful translation from mouse models to human therapeutic applications.

How can PEGylation be leveraged to create therapeutically optimized IL-22 variants with enhanced pharmacokinetic properties?

PEGylation can be strategically leveraged to create therapeutically optimized IL-22 variants with enhanced properties:

  • Site-directed PEGylation strategies:

    • Structure-guided selection of PEGylation sites that preserve receptor binding interfaces

    • Avoidance of residues involved in IL-22Rα binding (site 1) and IL-10Rβ binding (site 2)

    • Consideration of N-terminal, C-terminal, or internal site-specific conjugation

    • Systematic comparison of different attachment chemistries (maleimide-cysteine, click chemistry, enzymatic approaches)

  • PEG architecture optimization:

    • Evaluation of linear versus branched PEG structures for optimal pharmacokinetic profile

    • Molecular weight titration (5-40 kDa) to balance circulatory persistence with tissue penetration

    • Assessment of releasable PEG linkers for targeted activity in diseased tissues

    • Consideration of site-specific multi-PEGylation for maximal half-life extension

  • Functional impact characterization:

    • Comprehensive analysis of how PEGylation affects STAT3/STAT1 activation ratios

    • Assessment of whether PEGylation enhances or diminishes signaling bias of engineered variants

    • Determination of tissue-specific distribution changes resulting from PEGylation

    • Evaluation of how PEGylation impacts IL-22BP binding and neutralization

  • Disease-specific optimization:

    • Acute conditions: Focus on rapid-acting formulations with moderate half-life extension

    • Chronic conditions: Maximize half-life for sustained activity with minimal dosing frequency

    • Inflammatory bowel disease: Optimize for gastrointestinal targeting and mucosal penetration

    • Systemic conditions: Balance distribution properties with circulatory persistence

  • Combination with biased signaling:

    • PEGylation of STAT3-biased variants (e.g., 22-B3) for enhanced therapeutic index

    • Development of PEG-conjugated super-agonists (e.g., Super-22a/b) for applications requiring potent receptor activation

    • Fine-tuning of PEGylation to compensate for any activity reductions in biased variants

    • Exploration of tissue-selective PEGylation patterns to align with disease-specific needs

Advanced PEGylation approaches can extend IL-22's approximately 2-3 hour half-life to potentially 1-5 days, dramatically improving dosing convenience and maintaining more consistent exposure levels. This pharmacokinetic enhancement, when combined with engineered signaling specificity (as in STAT3-biased variants), creates opportunities for IL-22-based therapeutics with improved efficacy, reduced side effects, and enhanced patient compliance. Importantly, PEGylation strategies should be customized for each engineered variant to ensure preservation of its unique signaling properties and therapeutic advantages.

What emerging technologies will advance understanding of tissue-specific IL-22 signaling networks in mouse models?

Several emerging technologies promise to revolutionize our understanding of tissue-specific IL-22 signaling networks:

  • Advanced single-cell and spatial transcriptomics:

    • Single-cell RNA-seq to identify cell-specific responses to IL-22 within heterogeneous tissues

    • Spatial transcriptomics (e.g., Visium, MERFISH) to map IL-22 responses within tissue architecture

    • Single-cell ATAC-seq to reveal chromatin accessibility changes mediating STAT1 versus STAT3 responses

    • Spatial proteomics to visualize protein-level changes in IL-22-responsive tissues

  • Real-time in vivo signaling visualization:

    • STAT3/STAT1 translocation reporter mice for dynamic pathway activation imaging

    • Intravital microscopy with fluorescent IL-22 variants to track receptor binding in real-time

    • Bioluminescence resonance energy transfer (BRET) systems for monitoring receptor dimerization

    • Nanobody-based biosensors for tracking immediate signaling events in live tissues

  • Precision genetic engineering approaches:

    • CRISPR-based screens to identify novel components of IL-22 signaling networks

    • Base editing to introduce precise receptor mutations without disrupting expression

    • Tissue-specific inducible expression systems for temporal control of pathway components

    • Receptor component knockin models with fluorescent tags at endogenous loci

  • Advanced structural and interaction methodologies:

    • Cryo-electron microscopy of complete signaling complexes in near-native states

    • Hydrogen-deuterium exchange mass spectrometry to map dynamic protein interactions

    • Protein-protein interaction screening with engineered IL-22 variant libraries

    • Structural prediction with AI tools to design novel functional IL-22 variants

  • Integrative systems biology approaches:

    • Multi-omics integration to build comprehensive tissue-specific signaling networks

    • Mathematical modeling of STAT activation kinetics across tissues with variable receptor expression

    • Network analysis to identify key nodes controlling protective versus inflammatory outcomes

    • Machine learning approaches to predict tissue-specific responses to engineered IL-22 variants

These technologies will enable unprecedented resolution of how IL-22 signaling proceeds in different tissue environments, revealing the molecular basis for differential outcomes across tissues with varying IL-10Rβ expression levels . This deeper understanding will inform the design of next-generation IL-22-based therapeutics with enhanced tissue selectivity and functional specificity.

How might combining IL-22 variants with emerging drug delivery technologies enhance therapeutic applications?

Combining IL-22 variants with emerging drug delivery technologies presents transformative opportunities for enhanced therapeutic applications:

  • Targeted nanoparticle delivery systems:

    • Lipid nanoparticles encapsulating IL-22 or IL-22-encoding mRNA for extended release

    • Polymer-based nanocarriers decorated with tissue-specific targeting ligands

    • Stimuli-responsive nanoparticles that release IL-22 in response to disease-specific triggers

    • Layer-by-layer nanoparticles for sequential delivery of IL-22 with complementary therapeutics

  • Hydrogel-based local delivery platforms:

    • Injectable thermoresponsive hydrogels for sustained local IL-22 release

    • Adhesive hydrogels for mucosal surface delivery in IBD applications

    • Electrospun fiber scaffolds incorporating IL-22 for wound healing applications

    • Biologically responsive hydrogels that degrade and release IL-22 based on inflammatory signals

  • Engineered cell-based delivery approaches:

    • Modified probiotics secreting STAT3-biased IL-22 variants for intestinal delivery

    • Engineered stem cells providing sustained IL-22 production at sites of tissue damage

    • CAR-T cells modified to produce IL-22 in response to disease-specific antigens

    • Exosomes loaded with IL-22 or IL-22-inducing factors for enhanced tissue penetration

  • Device-based precision delivery:

    • Microneedle patches for controlled transdermal IL-22 delivery

    • Implantable microelectromechanical systems (MEMS) for programmable release

    • Ultrasound-triggered release from acoustically responsive carriers

    • 3D-printed drug-eluting implants customized for specific anatomical locations

  • Hybrid combinatorial approaches:

    • Co-delivery of STAT3-biased IL-22 variants with anti-inflammatory agents

    • Sequential delivery systems releasing IL-22BP antagonists followed by IL-22

    • Tissue-selective targeting combined with PEGylated IL-22 for optimized pharmacokinetics

    • Multi-compartment delivery systems releasing different IL-22 variants based on disease stage

These advanced delivery technologies can address key challenges in IL-22 therapeutics by: (1) providing tissue-specific targeting to enhance efficacy and reduce systemic exposure; (2) enabling sustained release to overcome the short half-life of native IL-22; (3) protecting STAT3-biased variants from degradation or neutralization; and (4) allowing precise spatiotemporal control of IL-22 activity to align with disease dynamics. Integration of these delivery technologies with engineered IL-22 variants like 22-B3 could revolutionize treatment approaches for inflammatory bowel disease, acute lung injury, and other conditions where targeted epithelial protection is therapeutic.

What are the most promising research directions for developing next-generation engineered IL-22 variants with enhanced therapeutic properties?

The development of next-generation engineered IL-22 variants presents several promising research directions:

  • Enhanced functional selectivity engineering:

    • Development of pure STAT3-exclusive agonists through comprehensive mutagenesis of the IL-10Rβ interface

    • Creation of variants with calibrated STAT3/STAT5 activation ratios for specific therapeutic applications

    • Engineering tissue-selective variants that exploit differential receptor expression patterns

    • Design of switchable IL-22 variants responsive to disease-specific environmental cues

  • Multi-functional cytokine engineering:

    • Creation of IL-22/IL-10 chimeric proteins combining epithelial protection with immune regulation

    • Development of bifunctional molecules linking IL-22 activity with targeted antagonism of pro-inflammatory pathways

    • Engineering of IL-22-receptor targeted delivery of other therapeutic payloads

    • Design of variants that can sequentially activate distinct signaling pathways based on disease stage

  • Advanced protein engineering approaches:

    • Computational protein design to create super-stable IL-22 variants with extended half-life

    • Directed evolution in mammalian display systems to optimize human receptor engagement

    • Circuit-based cellular screening systems to identify variants with precise signaling properties

    • De novo design of minimal IL-22 mimetics retaining key functional properties

  • Disease-specific optimization:

    • Variants optimized for intestinal barrier enhancement in IBD

    • Lung-targeted variants for acute respiratory distress syndrome and viral pneumonia

    • Liver-specific variants for metabolic liver diseases

    • Tissue-regenerative variants for wound healing applications

  • Translation-focused development:

    • Humanized variants with minimized immunogenicity risk

    • Production optimization for consistent glycosylation and post-translational modifications

    • Stability engineering for enhanced formulation options

    • Synergistic combinations with established therapeutics for specific indications

The structure-function insights gained from the high-resolution IL-22–IL-22Rα–IL-10Rβ complex provide a solid foundation for rational design of these next-generation variants. Particularly promising are approaches building on the STAT3-biased agonist platform (exemplified by 22-B3), which has demonstrated the ability to uncouple tissue-protective functions from inflammatory effects . Future variants could further refine this selective signaling while incorporating enhanced pharmaceutical properties through rational protein engineering, potentially yielding transformative therapeutics for conditions characterized by epithelial barrier dysfunction and inflammatory damage.

Product Science Overview

Introduction

Interleukin-22 (IL-22) is a cytokine that plays a crucial role in the immune response, particularly in the regulation of inflammatory processes and the maintenance of tissue homeostasis. It is also known as IL-10-related T cell-derived inducible factor (IL-TIF). IL-22 was initially identified as a gene induced by IL-9 in mouse T cells and mast cells .

Structure and Production

Mouse IL-22 cDNA encodes a protein consisting of 179 amino acid residues, including a 33 amino acid signal peptide that is cleaved to generate a mature protein of 147 amino acids . The recombinant form of IL-22 is produced in Escherichia coli (E. coli) and is a single, non-glycosylated homodimeric polypeptide chain .

Pegylation

Pegylation refers to the process of attaching polyethylene glycol (PEG) chains to a protein or peptide. This modification enhances the stability and solubility of the protein, reduces immunogenicity, and prolongs its half-life in the bloodstream. The pegylated form of mouse IL-22 is mono-pegylated with a 20 kDa PEG chain . This modification results in a protein with a molecular mass of 36 kDa, which appears as a 50 kDa protein on SDS-PAGE due to its enlarged hydrodynamic volume .

Biological Activity

IL-22 is known to activate the STAT-1 and STAT-3 signaling pathways in various cell types, including hepatoma cell lines. It upregulates the production of acute-phase proteins and is involved in inflammatory responses . The functional IL-22 receptor complex consists of two subunits: IL-22R and IL-10Rβ, both of which belong to the class II cytokine receptor family .

Applications

Recombinant pegylated mouse IL-22 is used in various research applications, including studies on inflammation, immune response, and tissue regeneration. Its enhanced stability and prolonged half-life make it suitable for long-term infusion studies using osmotic pumps .

Storage and Stability

Lyophilized pegylated mouse IL-22 is stable at room temperature for several days but should be stored desiccated below -20°C for long-term storage. Upon reconstitution, the protein can be stored at 4°C for several weeks, making it suitable for extended experimental 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.