IL-22 Human, Sf9 is generated through the Baculovirus Expression System (BES):
The final product is lyophilized or provided as a liquid solution, optimized for bioactivity assays.
IL-22 Human, Sf9 binds to the heterodimeric receptor complex IL-22R1/IL-10RB, activating STAT3-dependent pathways.
Tissue Repair: Promotes epithelial regeneration in the gut, skin, and liver.
Antimicrobial Defense: Enhances barrier integrity against pathogens.
Inflammation Modulation: Exerts pro-inflammatory effects in autoimmune diseases (e.g., psoriasis) and anti-inflammatory roles in asthma.
Receptor Subunit | Function | Sources |
---|---|---|
IL-22R1 | Specific binding to IL-22; expressed on non-immune cells (e.g., epithelial, fibroblasts). | |
IL-10RB | Shared subunit with IL-10; critical for signal transduction. |
Inflammatory Bowel Disease (IBD): IL-22 promotes epithelial survival but exacerbates colitis in murine models. Neutralizing antibodies (e.g., ABR167) reduce IL-1β, IL-6, and IL-17A in DSS-induced colitis .
Psoriasis: IL-22 upregulates chemokines and cytokines in keratinocytes, driving skin inflammation .
Glioblastoma: IL-22/IL-22R signaling enhances tumor cell survival via STAT3 activation .
Multiple Sclerosis: Elevated IL-22 levels correlate with astrocyte activation and neuroinflammation .
Sf9-derived IL-22 is preferred for studies requiring post-translational modifications.
Sf9, Baculovirus cells.
ADPAPISSHC RLDKSNFQQP YITNRTFMLA KEASLADNNT DVRLIGEKLF HGVSMSERCY LMKQVLNFTL EEVLFPQSDR FQPYMQEVVP FLARLSNRLS TCHIEGDDLH IQRNVQKLKD TVKKLGESGE IKAIGELDLL FMSLRNACIH HHHHH.
IL-22 Human produced in Sf9 Baculovirus cells is a single, glycosylated polypeptide chain consisting of 155 amino acids (residues 34-179) with a 6-amino acid histidine tag at the C-terminus. The protein has a molecular mass of approximately 17.8 kDa and appears as a sterile filtered colorless solution when purified . The amino acid sequence begins with ADPAPISSHC and continues through a series of amino acid residues ending with the histidine tag HHHHHH, which facilitates purification and detection in research applications . The glycosylation pattern in Sf9-derived IL-22 may differ from mammalian cell expression systems, which should be considered when evaluating biological activity and stability in experimental designs.
IL-22 signaling operates through a heterodimeric receptor complex comprising the specific receptor IL-22RA1 and the shared subunit IL-10RB . The binding process follows a sequential mechanism where IL-22 first binds to IL-22RA1 with high affinity, creating a complex that subsequently enables binding of the IL-10RB subunit . This completed receptor complex then initiates intracellular signaling primarily through the JAK-STAT pathway, leading to the phosphorylation of STAT3 at tyrosine residue 705 and sometimes at serine residue 727 . Additionally, IL-22 binding activates the MAPK and p38 pathways, which contribute to the diverse cellular responses observed in target tissues . When designing experiments, researchers should consider that IL-22 primarily targets non-hematopoietic cells, particularly epithelial cells, as these express the necessary IL-22RA1 receptor subunit.
For optimal preservation of IL-22 Human, Sf9 biological activity, store the protein at 4°C if the entire vial will be used within 2-4 weeks . For longer-term storage, maintain the protein at -20°C in an environment that minimizes freeze-thaw cycles, as these can significantly degrade protein structure and activity . To enhance stability during long-term storage, it is recommended to add a carrier protein such as 0.1% human serum albumin (HSA) or bovine serum albumin (BSA) . The standard formulation of IL-22 protein solution (1mg/ml) contains Phosphate Buffered Saline (pH 7.4) with 10% glycerol, which helps maintain stability during freeze-thaw processes . When designing experiments that will span several weeks, consider aliquoting the stock solution to avoid repeated freeze-thaw cycles of the entire preparation.
The biological activity of IL-22 Human, Sf9 can be quantified through its ability to induce IL-10 secretion in COLO 205 human colorectal adenocarcinoma cells, where the effective dose for 50% response (ED50) is typically ≤ 1.2 ng/ml . Alternative bioassays include measuring STAT3 phosphorylation in responsive cell lines through Western blotting or flow cytometry, which directly assesses the primary signaling pathway activated by IL-22 . Additionally, researchers can evaluate IL-22-induced expression of antimicrobial peptides and mucins in epithelial cell lines, or assess proliferation rates in target cells as IL-22 promotes cell survival and proliferation through STAT3, ERK1/2, and PI3K/AKT pathways . When designing these assays, include appropriate positive and negative controls, and consider dose-response experiments to establish the effective concentration range for your specific experimental system.
To study IL-22 interactions with its receptor complex, researchers can employ surface plasmon resonance (SPR) to measure binding kinetics and affinity between IL-22 and IL-22RA1, which exhibits a dissociation constant (KD) of approximately 20 nM . Co-immunoprecipitation assays can be used to confirm the formation of the IL-22-IL-22RA1-IL-10RB complex in cell models, while FRET (Fluorescence Resonance Energy Transfer) or BRET (Bioluminescence Resonance Energy Transfer) techniques can provide real-time visualization of receptor interactions in living cells . For functional studies, researchers can use siRNA knockdown or CRISPR-Cas9 targeting of either receptor component to assess the impact on downstream signaling. When investigating the stabilizing effect of GSK3β phosphorylation on IL-22R1, phospho-specific antibodies can be employed in Western blot analysis to detect receptor phosphorylation at serine residues 410 and 414 .
When comparing Sf9-derived IL-22 with proteins from other expression systems (e.g., E. coli, mammalian cells), researchers should first standardize protein quantification using methods like BCA or Bradford assays to ensure equivalent concentrations . Purity assessment by SDS-PAGE or HPLC is essential, with Sf9-derived IL-22 typically exhibiting greater than 90% purity . Functional comparisons should include dose-response experiments measuring canonical activities such as STAT3 phosphorylation, antimicrobial peptide induction, or IL-10 secretion in COLO 205 cells . Glycosylation analysis using mass spectrometry or specialized glycoprotein staining is particularly important, as the insect cell glycosylation pattern differs from mammalian systems and may affect protein stability, half-life, or receptor binding kinetics . For comprehensive characterization, circular dichroism spectroscopy can assess secondary structure differences that might impact biological activity.
To investigate IL-22's role in epithelial barrier function, develop an experimental framework incorporating both in vitro and in vivo approaches. For in vitro studies, establish trans-epithelial electrical resistance (TEER) assays using polarized epithelial monolayers (such as Caco-2 or T84 cells) treated with IL-22 . Complement TEER measurements with paracellular permeability assays using fluorescently labeled dextrans of various molecular weights to assess barrier integrity at different levels. Immunofluorescence microscopy focusing on tight junction proteins (claudins, occludin, ZO-1) can visualize IL-22-induced barrier modifications . For in vivo approaches, utilize dextran-FITC gavage in mouse models with compromised barriers (e.g., DSS colitis) treated with IL-22 to measure barrier recovery. Integrate measurements of antimicrobial peptide expression (e.g., Reg3β, Reg3γ) which are upregulated by IL-22 and contribute to barrier defense . For mechanistic insights, incorporate phosphorylation inhibitors targeting STAT3, ERK1/2, or PI3K/AKT pathways to determine which signaling cascade is critical for the barrier-protective effects of IL-22 .
Distinguishing direct IL-22 effects from secondary responses requires a multi-layered experimental approach. First, establish cell type-specific expression of IL-22RA1 in your experimental system using immunohistochemistry, flow cytometry, or single-cell RNA sequencing, as only cells expressing this receptor can respond directly to IL-22 . Implement time-course experiments measuring early responses (0-1 hour: STAT3 phosphorylation) versus later events (6-24 hours: gene expression changes; 24-72 hours: phenotypic alterations) to separate primary from secondary effects . To definitively identify direct targets, perform chromatin immunoprecipitation sequencing (ChIP-seq) for phosphorylated STAT3 following IL-22 stimulation, revealing immediate transcriptional targets . In complex tissue systems, utilize conditional knockout models where IL-22RA1 is deleted in specific cell populations to determine which phenotypic changes require direct IL-22 sensing. Alternatively, ex vivo organ cultures comparing responses in wild-type versus IL-22RA1-deficient tissues can isolate direct effects while maintaining tissue architecture .
To investigate cross-talk between IL-22 and other cytokine signaling pathways, design experiments that systematically address pathway interactions at multiple levels. Begin with combinatorial stimulation experiments treating cells with IL-22 alone, companion cytokines alone (e.g., IL-17, IFNγ, TNFα), and combinations at physiologically relevant ratios . Analyze signaling pathway activation through multiplexed phosphoprotein analysis (e.g., Luminex, reverse phase protein array) that can simultaneously detect activation of STAT3 (IL-22), NF-κB (TNFα), STAT1 (IFNγ), and other relevant pathways . For transcriptional cross-talk, perform RNA-seq following single and combined cytokine treatments, applying bioinformatic approaches to identify synergistic or antagonistic gene regulation patterns . To determine receptor-level interactions, assess whether pre-treatment with one cytokine alters receptor expression for another, potentially using flow cytometry or qPCR to quantify receptor dynamics . For mechanistic insights, utilize CRISPR-Cas9 to selectively disrupt components of one pathway while monitoring the other, thereby establishing dependency relationships. Include analysis of epigenetic modifications (e.g., histone acetylation at key regulatory regions) to understand how cytokine combinations might uniquely alter the chromatin landscape .
When encountering limited IL-22 Human, Sf9 biological activity, implement a systematic troubleshooting approach. First, verify protein integrity through SDS-PAGE under both reducing and non-reducing conditions to assess potential aggregation or degradation . Confirm target cell responsiveness by testing IL-22RA1 and IL-10RB expression using Western blot, flow cytometry, or qPCR, as receptor downregulation may occur in cultured cells . Examine buffer compatibility issues by performing small-scale dialysis into alternative buffers, noting that IL-22 formulation typically contains PBS with 10% glycerol at pH 7.4 . If aggregation is observed, consider adding low concentrations (0.01-0.05%) of non-ionic detergents like Tween-20. For diminished activity in serum-containing media, pre-incubate IL-22 with carrier proteins (0.1% BSA) to prevent non-specific binding . If glycosylation is crucial for your application but appears suboptimal in the Sf9 system, consider enzymatic deglycosylation followed by controlled renaturation protocols. Finally, optimize exposure time and concentration in dose-response experiments, as some experimental systems may require higher concentrations or extended exposure to IL-22 compared to the standard COLO 205 bioassay .
To minimize batch-to-batch variation in IL-22 Human, Sf9 experiments, implement standardized quality control procedures before initiating primary experiments. Establish a reference standard from a well-characterized batch and compare each new lot using analytical techniques including SDS-PAGE (for purity assessment), size exclusion chromatography (to detect aggregation), and circular dichroism (to verify proper folding) . Perform functional validation using a standardized bioassay, such as STAT3 phosphorylation or IL-10 induction in COLO 205 cells, calculating relative potency compared to your reference standard . Create internal standards by generating substantial stocks of validated IL-22 aliquoted and stored at -80°C to span multiple experimental series. Implement strict documentation procedures tracking source, lot number, reconstitution date, freeze-thaw cycles, and bioactivity measurements for each batch . To control for unavoidable variations, design experiments with internal normalization controls and consider including a standard curve of the reference protein in each experiment. If working with multiple batches is unavoidable for a single experimental series, stratify treatments to ensure all experimental conditions experience equivalent batch exposure.
Transitioning from in vitro to in vivo IL-22 research requires specific methodological adaptations. For in vivo administration, reformulate IL-22 Human, Sf9 into physiologically compatible buffers, typically phosphate-buffered saline without preservatives or stabilizers that might cause immunogenic reactions . Determine appropriate dosing regimens through preliminary pharmacokinetic studies measuring serum half-life, which is typically short for unmodified cytokines; consider using IL-22-Fc fusion proteins for extended half-life in circulation . Establish baseline expression of IL-22RA1 across target tissues in your model organism using immunohistochemistry or tissue-specific qPCR, as receptor distribution varies between species and dictates responsiveness . Develop relevant biomarkers for IL-22 activity in vivo, such as serum levels of regenerating islet-derived proteins (REG3A/REG3G) or liver-derived serum amyloid A . Include appropriate controls including vehicle-treated animals and, ideally, IL-22RA1 knockout models to confirm specificity of observed effects . For therapeutic applications, consider potential immunogenicity of Sf9-derived IL-22 due to insect cell-specific glycosylation patterns, which may necessitate modified formulations or alternative expression systems for repeated dosing studies .
Designing experiments to study IL-22-microbiome interactions requires integration of immunological and microbiological methodologies. Begin with gnotobiotic mouse models comparing IL-22-mediated responses in germ-free, specific pathogen-free, and defined microbial community settings . Implement 16S rRNA sequencing and metagenomic analysis to characterize microbial community shifts following IL-22 administration or in IL-22 knockout models, with particular attention to species capable of metabolizing host-derived antimicrobial peptides induced by IL-22 . Utilize ex vivo organ culture systems (e.g., intestinal organoids with microbiota) to study how IL-22 affects epithelial-microbial interactions in a controlled environment. For mechanistic studies, focus on antimicrobial peptides (Reg3β, Reg3γ) and mucins as key IL-22-regulated factors that shape microbial communities, using knockout models of these effectors to determine their necessity in IL-22-mediated microbiome effects . Consider metabolomic approaches to identify microbiome-derived metabolites that might be altered by IL-22 signaling, such as short-chain fatty acids or secondary bile acids. Design bidirectional experiments examining not only how IL-22 shapes the microbiome but also how microbial products regulate IL-22 production by immune cells, potentially through pattern recognition receptor signaling . For clinical translation, correlate IL-22 levels with microbiome composition in patient cohorts with inflammatory or metabolic conditions, utilizing multivariate statistical approaches to identify significant associations.
IL-22 is primarily produced by immune cells such as Th17, Th22, and innate lymphoid cells . It acts on non-hematopoietic cells, including epithelial cells and fibroblasts, to mediate its effects. The primary functions of IL-22 include:
Recombinant IL-22 (Mouse) is produced using the Sf9 insect cell expression system. This system is widely used for the production of recombinant proteins due to its ability to perform post-translational modifications similar to those in mammalian cells. The process involves the following steps:
Recombinant IL-22 has several applications in research and therapeutic development: