Recombinant Mouse Interleukin-22 protein (Il22) (Active)

Shipped with Ice Packs
In Stock

Description

Epithelial Regeneration and Barrier Integrity

  • Promotes epithelial cell proliferation and survival via STAT3 activation, critical for mucosal repair in colitis and lung injury .

  • Synergizes with IL-17A to amplify neutrophil-active chemokines (e.g., CXCL1, CXCL5) in colonic organoids, driving neutrophil recruitment .

Inflammatory Regulation

  • Pro-inflammatory Effects: Upregulates acute-phase proteins in hepatoma cells and enhances microbial recognition pathways .

  • Anti-inflammatory Balance: IL-22 binding protein (IL-22BP) constrains IL-22 activity, preventing excessive tissue damage .

Neutrophil Recruitment in Colitis

  • IL-22 overexpression in ulcerative colitis (UC) correlates with CXCR2+ neutrophil infiltration, worsening disease severity. Neutralizing IL-22 reduces colitis scores in murine models .

  • Transcriptional profiling links IL-22-driven chemokine networks to non-response to ustekinumab in UC patients .

Metabolic and Antimicrobial Roles

  • Restores mucosal immunity in diabetic mice by enhancing antimicrobial peptide production .

  • Improves bacterial clearance in pneumococcal pneumonia by downregulating macrophage oxidative phosphorylation and boosting glycolysis .

Protein Engineering Insights

  • Single-amino-acid muteins (Y51A, N54A) lose agonistic activity but retain antagonistic potential, enabling selective pathway inhibition .

  • Pegylation reduces IL-22 receptor affinity by 50–75%, highlighting structural sensitivity for therapeutic optimization .

Applications in Experimental Models

ApplicationModel SystemOutcomeSource
Goblet cell hyperplasiaHelminth-infected miceIL-22 mediates worm expulsion via REGIIIβ induction
Keratinocyte differentiationIn vitro co-cultureSynergy with IL-17A/TNFα inhibits differentiation
Corneal healingMurine injury modelIL-22/CCL20 axis enhances γδ T cell-mediated repair
Vaccine adjuvantsInfluenza-challenged miceIL-1/IL-22 co-administration boosts protective immunity

Clinical and Therapeutic Implications

  • Biomarker Potential: IL-22-responsive gene signatures predict ustekinumab response in UC, with high enrichment scores linked to non-remission .

  • Therapeutic Modulation: Recombinant IL-22 administration accelerates epithelial repair but risks exacerbating neutrophil-driven pathology in chronic inflammation .

Product Specs

Buffer
Lyophilized from a 0.2 µm filtered PBS, pH 7.4.
Form
Lyophilized powder
Lead Time
5-10 business days
Notes
Repeated freezing and thawing is not recommended. Store working aliquots at 4°C for up to one week.
Reconstitution
We recommend that this vial be briefly centrifuged prior to opening to bring the contents to the bottom. Please reconstitute the protein in deionized sterile water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% of glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our default final concentration of glycerol is 50%. Customers may use this as a reference.
Shelf Life
The shelf life is dependent on numerous factors, including storage state, buffer ingredients, storage temperature, and the stability of the protein itself. Generally, the shelf life of liquid form is 6 months at -20°C/-80°C. The shelf life of lyophilized form is 12 months at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt; aliquoting is necessary for multiple use. Avoid repeated freeze-thaw cycles.
Tag Info
Tag-Free
Synonyms
Il22; Il22a; Iltif; IltifaInterleukin-22; IL-22; IL-10-related T-cell-derived-inducible factor; IL-TIF; IL-TIF alpha; Interleukin-22a; IL-22a
Datasheet & Coa
Please contact us to get it.
Expression Region
34-179aa
Mol. Weight
16.6 kDa
Protein Length
Full Length of Mature Protein
Purity
>96% as determined by SDS-PAGE.
Research Area
Immunology
Source
E.coli
Species
Mus musculus (Mouse)
Target Names
Uniprot No.

Target Background

Function
Cytokine that contributes to the inflammatory response in vivo.
Gene References Into Functions
  1. The level of anti-IL17A autoantibodies that develop in aged Aire-deficient mice is not sufficient for conferring susceptibility to oropharyngeal candidiasis. However, patient-derived monoclonal antibodies that cross-react with murine IL-22 increase the fungal burden on C. albicans infected mucosa. PMID: 29150834
  2. This study defines a critical IL-36/IL-23/IL-22 cytokine network instrumental for antimicrobial peptide production and host defense in intestinal mucosa damage using a mouse inflammatory bowel disease model. PMID: 29760082
  3. The level of lncRNA H19 is increased in inflamed intestinal tissues from mice and patients. The inflammatory cytokine IL22 induces expression of H19 in IECs, which is required for intestinal epithelial proliferation and mucosal healing. H19 lncRNA appears to inhibit p53 protein and microRNA 34a and let-7 to promote proliferation of IECs and epithelial regeneration. PMID: 29621481
  4. Despite the presence of all Notch pathway molecules in the kidney and a model-specific induction of Notch ligands, IL-22 was only up-regulated in acute inflammation, but rapidly down-regulated during regeneration. This implies that for targeting injury responses, e.g. via IL-22, species-specific differences, injury type, and time points have to be considered. PMID: 29054964
  5. Knockout of signal transducer and activator of transcription factor-3 (STAT3) in intestine epithelial cells resulted in a complete loss of IL-22 protection, demonstrating that STAT3 is required for intestine barrier protection following ethanol combined with injury. PMID: 28498296
  6. Cancer cells induce IL-22 production from memory CD4(+) T cells via activation of the NLRP3 inflammasome and the release of IL-1beta to promote tumor growth. PMID: 29150554
  7. Although IL-22 is expressed, it seems to play a minor role in protection and pathology during the acute systemic infection with the reticulotropic Tulahuen strain of T. cruzi. PMID: 27650379
  8. IL-22 amplifies the inflammatory response, induces endothelial dysfunction, and promotes blood pressure elevation in angiotensin II-induced hypertensive mice via STAT3 signaling. PMID: 28974499
  9. These results demonstrate a strong association between Hh-induced DNA damage and the development of dysplasia, and further suggest that IL-22-dependent induction of iNOS within crypt epithelial cells rather than macrophages is a driving force in this process. PMID: 28198364
  10. IL-22 plays an important role in CIA development, and neutralizing this cytokine seems an attractive new strategy in RA treatment. Most importantly, SPECT/CT imaging with 111In-28H1 can be used to specifically monitor therapy responses, and is potentially more sensitive in disease monitoring than the gold standard method of macroscopic arthritis scoring. PMID: 29361119
  11. Protease IV is responsible for the degradation of IL-22 by P. aeruginosa. The major anti-proteases molecules present in the lungs were unable to inhibit protease IV enzymatic activity. This so far undescribed cleavage of IL-22 by a bacterial protease is likely to be an immune-evasion strategy that contributes to P. aeruginosa-triggered respiratory infections. PMID: 27792459
  12. IL-22 seems to be the critical cytokine for the development of atopic dermatitis (AD) and is induced in AD model by epicutaneous sensitization with ovalbumin. PMID: 28655472
  13. The finding demonstrated that IL-22 could exert favorable effects on Diabetic nephropathy (DN) via simultaneously alleviating systemic metabolic syndrome and downregulating renal NLRP3/caspase-1/IL-1beta pathway, suggesting that IL-22 might have therapeutic potential for the treatment of DN. PMID: 28726774
  14. This study shows that IL-22 mediates protective immunity during chronic stages of Mycobacterium tuberculosis HN878 infection in mice. PMID: 28247861
  15. This study shows that hepatocyte responses to IL-22 stimulation are reduced in hypoxic environments. PMID: 27796296
  16. Epithelial IL-23R signaling enables protective IL-22 responses in experimental colitis. PMID: 27524624
  17. We conclude that IL-22 has an important role in controlling S. aureus nasal colonization through distinct mechanisms, with IL-22 mediating its effect exclusively by inducing AMP expression and controlling availability of staphylococcal ligands. PMID: 27007677
  18. Results reveal that IL-22 increases intestinal epithelial permeability by upregulating Claudin-2 expression through the JAK/STAT pathway. PMID: 28939759
  19. Investigated the function of Card9-mediated innate immunity in inflammation-associated colon carcinogenesis; report that Card9-signaling drives the production of IL-1beta within the damaged intestine and regulates the subsequent generation of IL-22 by group3 innate lymphoid cells, which promotes tumorigenesis via STAT3 activation within the transformed epithelium. PMID: 28586167
  20. These results demonstrate that IL-22 has a critical role in vaccine-induced protection against Helicobacter pylori. PMID: 27143303
  21. This study shows for the first time that a defect in IL-22 is involved in the acute exacerbation induced by non-typeable Haemophilus influenzae infection during experimental chronic obstructive pulmonary disease. PMID: 27143304
  22. Hypoxic IL-22 upregulation is dependent on HIF-1alpha. PMID: 27534553
  23. Macrophage-derived IL-22 protects hepatocytes from ethanol-induced cell death. IL-22 downregulation is a new regulatory target of LPS in the pathogenesis of AH. PMID: 28637673
  24. IL-22 inhibits acetaldehyde-induced hepatic stellate cells activation and proliferation, which may be related to nuclear translocation of Nrf2 and increased activity of the antioxidant axis Nrf2-keap1-ARE. PMID: 28373766
  25. Endogenous IL-22 and hepatic IL-22R signaling play critical roles in controlling pneumococcal lung burden, and systemic IL-22 decreases bacterial burden in the lungs and peripheral organs by potentiating C3 opsonization on bacterial surfaces, through the increase of hepatic C3 expression. PMID: 27456484
  26. Our data suggest that IL-20 subfamily cytokines, particularly IL-20, IL-22, and IL-24, might provide therapeutic benefit for patients with Diabetic foot ulcers (DFU). PMID: 28125663
  27. Innate immune cell-derived IL-22 is required for efficient liver regeneration, and secretion of IL-22 in the regenerating liver is modulated by the ATP receptor, P2X1. PMID: 26853442
  28. IL-22 and its receptor have a crucial role in the development and pathogenesis of uveitis by facilitating inflammatory cell infiltration. PMID: 27166675
  29. We therefore postulate IL-22 as an important enhancer of the GC reaction, maintaining chemokine levels for the persistence of GC reactions, essential for the production of autoantibody-secreting plasma cells. Blocking IL-22 might therefore prevent immune-complex deposition and destruction of joints in RA patients. PMID: 27067635
  30. AhR has a direct role in IL-22 production by Th17 cells in the mouse ear skin, but not by gammadelta T cells, CD4(-) CD8(-) TCRbeta(+) T cells and ILCs. PMID: 27000947
  31. Taken together, our results demonstrate that Dok-1 and Dok-2 negatively regulate intestinal inflammation, apparently through the induction of IL-17A and IL-22 expression. PMID: 27450811
  32. Upregulation of IL-22 in combination with a complete loss of its negative regulator IL-22BP, and increased downstream STAT3-signaling in K8(-/-) and K8(-/-)Apc(Min/+) colonic epithelia confirmed that the IL-22 pathway, important in inflammation, proliferation, and tissue regeneration. PMID: 27234655
  33. This study shows that serum IL-22/IL-22BP protein ratio strongly correlates with psoriasis severity. PMID: 28356382
  34. Data suggest that the influence of IL-22 on autoimmunity is determined in part by the local microenvironment. In particular, IL-22 deficiency exacerbates tissue injury in inflammatory bowel disease, but has no influence on either the hepatocytes or cholangiocytes in the same model. PMID: 27148790
  35. This study has demonstrated a crucial role for retinoic acid in promoting IL-22 production and tempering dendritic cell function through downregulating S100A4 protein during viral hepatitis. PMID: 28363907
  36. This study shows that losartan and dexamethasone may suppress inflammatory responses in IgA nephropathy by inhibiting IL-22 expression in Th22 cells. PMID: 27930971
  37. Exogenous recombinant IL-22 protects mice against L-arginine-induced severe acute pancreatitis-associated lung injury by enhancing the expression of anti-apoptosis genes through the STAT3 signaling pathway. PMID: 27275094
  38. Orally administered R848 triggers TLR-7 on CD11c(+) dendritic cells, inducing interleukin-23 (IL-23) expression followed by a burst of IL-22 secretion by innate lymphoid cells, leading to Reg3gamma expression and restoration of colonization resistance against vancomycin-resistant enterococcus. PMID: 26912904
  39. IL-22 Defect During Streptococcus pneumoniae Infection Triggers Exacerbation of Chronic Obstructive Pulmonary Disease. PMID: 26870795
  40. Rag-RORgammat-reporter and Rag KO mice undergoing ischemia reperfusion injury expressed high protein levels of both IL-22 and GFP (RORgammat). PMID: 26341825
  41. Overexpression of IL-22 significantly reduced the Klebsiella pneumonia infection in the liver and spleen. PMID: 26729763
  42. IL-22 restrains tapeworm-mediated protection against colitis via regulation of IL-25 expression. PMID: 27055194
  43. IL-22 is involved in plaque formation; IL-22 released by immune cells is involved in activation of vascular repair by stimulating medial SMC dedifferentiation into a synthetic phenotype. PMID: 26298743
  44. Cigarette smoke can inhibit the ROCK2-IRF4 axis and modulate T cell production of IL-22. PMID: 26882474
  45. IL-17A and IL-22 work synergistically to induce antimicrobials and chemokines such as IL-8, components of calprotectin (CP), lipocalin (LCN), and some beta-defensins in both human and primary mouse gastric epithelial cells (GEC) and gastroids. PMID: 26867135
  46. The study demonstrated the probable involvement of gamma delta T cells in the immune response of an organism via the secretion of IL-17 and IL-22. PMID: 26400286
  47. IL-22 is not required for type 1 diabetes pathogenesis; suggested that IL-22 may have a regenerative and protective role in the pancreatic islets. PMID: 26496462
  48. IL-23, but not IL-17a or IL-22, promotes neutrophil recruitment and inflammatory cytokine and chemokine expression in the colon in response to C. difficile infection. PMID: 26455347
  49. Data suggest that interleukin 22 (IL-22) plays a pro-inflammatory/pathogenic role in the onset of antigen-induced arthritis (AIA) through apoptosis-associated speck-like Pycard protein (ASC)-dependent stimulation of interleukin-1 beta (IL-1beta) production. PMID: 26330334
  50. IL-22 can play a previously unappreciated role in controlling leishmania-induced immunopathology. PMID: 26285207

Show More

Hide All

Database Links
Protein Families
IL-10 family
Subcellular Location
Secreted.

Q&A

What is the molecular structure of recombinant mouse IL-22 protein?

Recombinant mouse IL-22 is a 179 amino acid residue protein with a putative 33 amino acid signal peptide that is cleaved to generate a 147 amino acid mature protein. The commercially available recombinant protein typically spans amino acids 25-179. The protein shares approximately 79% amino acid sequence identity with human IL-22 and 22% with IL-10. The mouse IL-22 gene is located on chromosome 10 and exists as either a single copy or duplicated gene (IL-TIF alpha and IL-TIF beta with >98% sequence homology) depending on the mouse strain, with duplication occurring in C57B1/6, FVB, and 129 strains .

What are the key biological functions of IL-22 in mouse models?

IL-22 plays critical roles in modulating tissue responses during inflammation and is essential for the regeneration of epithelial cells to maintain barrier function after injury and prevent further tissue damage. Unlike most cytokines, IL-22 has no direct effect on immune cells. It signals through a heterodimeric receptor composed of the specific receptor IL-22RA1 (present on non-immune cells in multiple organs) and the shared subunit IL-10RB. This interaction activates the tyrosine kinases JAK1 and TYK2, which then activate STAT3. Consequently, IL-22 promotes cell survival and proliferation through STAT3, ERK1/2, and PI3K/AKT pathways, and promotes phosphorylation of GSK3B and CTTN .

How is IL-22 expression regulated in mouse tissues during inflammation?

IL-22 is produced by normal mouse T cells upon Concanavalin A (Con A) activation. Its expression is also induced in various organs following lipopolysaccharide (LPS) injection, suggesting its involvement in inflammatory responses. Type 3 Innate Lymphoid Cells (ILC3) are a significant source of IL-22, as demonstrated in lupus-prone MRL/lpr mice . The expression pattern indicates that IL-22 serves as an important mediator in the inflammatory cascade, bridging adaptive and innate immune responses in tissue inflammation scenarios.

What are the optimal conditions for using recombinant mouse IL-22 in in vitro experiments?

For in vitro experiments with mouse primary cells or cell lines, the recommended effective dose (ED50) of recombinant mouse IL-22 is typically 60-300 pg/mL . The protein should be reconstituted according to manufacturer specifications, usually in sterile filtered PBS containing at least 0.1% carrier protein such as BSA. When stimulating cells in culture, pretreatment with the protein for 24-48 hours is common in many experimental protocols. For instance, when testing IL-22's effects on kidney epithelial cells, researchers typically treat cells with recombinant IL-22 for 30 minutes to 24 hours, with phosphorylation of STAT3 observable within 30 minutes and changes in chemokine expression (CCL2, CXCL10) detectable within several hours .

How should researchers design experiments to study IL-22 signaling pathways?

When investigating IL-22 signaling pathways, researchers should consider:

  • Receptor expression analysis: Confirm expression of IL-22RA1 and IL-10RB in target cells before experiments

  • Pathway inhibitor controls: Include STAT3 pathway inhibitors (e.g., C188-9) as controls

  • Time-course experiments: Monitor both immediate (minutes to hours) and delayed (12-48 hours) responses

  • Phosphorylation analysis: Use Western blot or flow cytometry to detect phosphorylated STAT3, which is a key mediator of IL-22 signaling

  • Downstream target measurement: Assess expression of known IL-22-responsive genes such as CCL2 and CXCL10

Western blot analysis should be performed to detect phosphorylated STAT3 at 15, 30, 60, and 120 minutes post-stimulation, while qPCR for target genes should be performed at 6, 12, and 24 hours to capture the complete signaling cascade.

What functional assays can be used to assess IL-22 biological activity in research settings?

Several functional assays can be employed to assess IL-22 biological activity:

  • Cell proliferation/survival assays: MTT or WST-1 assays with epithelial cell lines

  • Wound healing assays: Scratch assays with epithelial monolayers

  • Chemokine induction: qPCR or ELISA measurement of CCL2, CXCL10 expression

  • Transwell migration assays: Using supernatant from IL-22-stimulated cells to assess immune cell recruitment

  • STAT3 activation: Phospho-flow cytometry or Western blot for phosphorylated STAT3

  • Barrier function: Transepithelial electrical resistance (TEER) measurements in epithelial cultures

In transwell assays, supernatant from primary kidney epithelial cells treated with recombinant IL-22 has been shown to recruit significantly more macrophages compared to controls, demonstrating IL-22's role in promoting immune cell chemotaxis through indirect mechanisms .

How does IL-22 function in mouse models of inflammatory bowel disease?

In mouse models of ulcerative colitis, IL-22 regulates pro-inflammatory pathways involved in microbial recognition, cancer, and immune cell chemotaxis, most prominently those involving CXCR2+ neutrophils. IL-22-mediated transcriptional regulation of CXC-family neutrophil-active chemokine expression is highly conserved across species, depends on STAT3 signaling, and is functionally important in recruiting CXCR2+ neutrophils into colonic tissue. The magnitude of enrichment of IL-22-regulated transcripts in colonic biopsies correlates with neutrophil infiltration in ulcerative colitis patients . When designing experiments to study IL-22 in colitis models, researchers should consider using organoid cultures alongside in vivo models to comprehensively assess epithelial-specific effects.

What is the role of IL-22 in lupus nephritis models, and how can this be studied?

In lupus nephritis models using MRL/lpr mice, IL-22 secreted primarily by Type 3 Innate Lymphoid Cells (ILC3) has been shown to play a pathogenic role. IL-22 binding to IL-22R on kidney epithelial cells activates the STAT3 signaling pathway, enhancing chemokine secretion (particularly CCL2 and CXCL10) and promoting macrophage infiltration into the kidney. Both IL-22 knockout and IL-22R knockout MRL/lpr mice exhibit decreased macrophage infiltration in the kidney, reduced proteinuria, improved renal function, and less severe pathological impairment compared to control mice. These mice also demonstrate milder lymphadenopathy and splenomegaly, suggesting IL-22 may also influence systemic immune responses .

For studying IL-22 in lupus nephritis, researchers should:

  • Compare wild-type, IL-22 KO, and IL-22R KO mice in the MRL/lpr background

  • Monitor survival, skin lesions, proteinuria, and renal function

  • Assess immune complex deposition and complement activation in kidney tissue

  • Quantify infiltrating immune cells using flow cytometry and immunohistochemistry

  • Measure autoantibody production and serum complement levels

How do knockout/knockin approaches compare with antibody neutralization for studying IL-22 function in vivo?

Both genetic approaches (knockout/knockin) and antibody neutralization have distinct advantages and limitations when studying IL-22 function:

Genetic approaches (IL-22 KO or IL-22R KO):

  • Advantages: Complete absence of target protein; no off-target effects from therapeutic agents; allows study of developmental effects

  • Limitations: May trigger compensatory mechanisms during development; cannot study temporal effects easily; strain-specific variations exist (e.g., IL-22 gene duplication in C57B1/6, FVB, and 129 strains)

Antibody neutralization:

  • Advantages: Can be administered at specific time points; dose-dependent inhibition possible; more translatable to therapeutic approaches

  • Limitations: Incomplete neutralization; potential off-target effects; antibody immunogenicity in long-term studies

In lupus nephritis studies, both IL-22 KO and anti-IL-22 monoclonal antibody treatment have shown similar protective effects in MRL/lpr mice, confirming the pathogenic role of IL-22 . For comprehensive understanding, researchers should consider using both approaches - genetic models for mechanistic insights and antibody neutralization for therapeutic potential assessment.

What expression systems are optimal for producing high-quality recombinant mouse IL-22?

Recombinant mouse IL-22 can be produced in various expression systems, each with distinct characteristics affecting protein quality and function:

  • E. coli expression systems: Yield high amounts of protein but may lack proper post-translational modifications. Commercial E. coli-derived mouse IL-22 proteins typically contain N-terminal methionine and span Leu34-Val179 .

  • Mammalian expression systems (HEK293): Produce proteins with proper folding and post-translational modifications, resulting in higher biological activity. Commercial HEK293-derived mouse IL-22 proteins typically achieve ≥95% purity with endotoxin levels ≤0.005 EU/μg .

For research requiring high biological activity and minimal endotoxin contamination, mammalian expression systems are preferred, particularly for in vivo studies and primary cell culture experiments.

How can researchers accurately quantify biologically active IL-22 in experimental samples?

Accurate quantification of biologically active IL-22 in experimental samples requires multiple complementary approaches:

  • ELISA: Measures protein concentration but not necessarily biological activity

  • Bioactivity assays: Cell-based assays measuring STAT3 phosphorylation in responsive cell lines

  • Western blot: For semi-quantitative analysis with specificity verification

  • Mass spectrometry: For precise identification and absolute quantification

  • Flow cytometry: For intracellular detection in specific cell populations

When designing quantification experiments, researchers should include appropriate standards and controls, and consider the specific context of their research question. For instance, detection of IL-22 in tissue lysates may require different approaches than detection in cell culture supernatants.

What are common pitfalls in experimental design when studying IL-22 signaling, and how can they be avoided?

Several common pitfalls occur in IL-22 signaling studies:

  • Failure to confirm receptor expression: Always verify IL-22RA1 and IL-10RB expression in target cells, as IL-22 effects are dependent on receptor presence.

  • Cross-reactivity issues: When using anti-IL-22 antibodies, validate specificity, as IL-22 shares structural similarities with other IL-10 family cytokines.

  • Overlooking strain differences: Consider that some mouse strains (C57B1/6, FVB, 129) have duplicated IL-22 genes (IL-TIF alpha and IL-TIF beta), which may affect knockout strategies and interpretation of results .

  • Endotoxin contamination: Ensure recombinant proteins have low endotoxin levels (<0.005 EU/μg), as endotoxin can independently activate inflammatory pathways .

  • Timing of measurements: IL-22 effects can be rapid (phosphorylation events within minutes) or delayed (gene expression changes over hours), so design time-course experiments accordingly.

To avoid these pitfalls, researchers should include appropriate positive and negative controls, validate reagents thoroughly, perform time-course experiments, and consider using multiple complementary approaches to confirm findings.

How can researchers effectively study the differential effects of IL-22 on various tissue-specific epithelial cells?

To study tissue-specific effects of IL-22, researchers should:

  • Isolate primary epithelial cells from different organs (intestine, kidney, liver, skin) using tissue-specific isolation protocols

  • Characterize IL-22 receptor expression on each cell type using flow cytometry or qPCR

  • Perform comparative transcriptomic analysis (RNA-seq) on different epithelial cell types after IL-22 stimulation to identify tissue-specific gene signatures

  • Use tissue-specific conditional knockout models of IL-22RA1 to examine in vivo relevance

  • Employ organoid cultures from different organs to maintain tissue-specific architecture

For example, when studying kidney epithelial cells versus colonic epithelial cells, researchers have observed that IL-22 induces CCL2 and CXCL10 in kidney cells, promoting macrophage recruitment , while in colonic cells, IL-22 regulates neutrophil-active chemokines, promoting neutrophil infiltration . These tissue-specific differences highlight the context-dependent nature of IL-22 signaling.

What approaches can be used to study the interplay between IL-22 and other cytokines in complex inflammatory environments?

Studying cytokine interplay requires sophisticated experimental designs:

  • Combinatorial cytokine stimulation: Treat cells with IL-22 alone or in combination with other cytokines (IL-17, TNFα, IL-1β) to identify synergistic or antagonistic effects

  • Multi-parametric flow cytometry: To simultaneously detect multiple phosphorylated signaling molecules

  • Single-cell RNA sequencing: To identify cell-specific responses in heterogeneous populations

  • Conditional knockout systems: Use Cre-lox systems to delete IL-22RA1 in specific cell types while exposing them to complex cytokine environments

  • In vivo cytokine blockade combinations: Combine anti-IL-22 antibodies with blockade of other cytokines

In inflammatory bowel disease models, the relationship between IL-22 and other cytokines (particularly IL-17) has been shown to significantly influence disease progression. Similarly, in lupus nephritis models, the interplay between IL-22 and type I interferons impacts disease severity .

How can researchers investigate the therapeutic potential of targeting IL-22 in autoimmune and inflammatory diseases?

To investigate IL-22 as a therapeutic target, researchers should:

  • Compare prophylactic versus therapeutic interventions: Administer anti-IL-22 antibodies before disease onset (prophylactic) or after disease establishment (therapeutic)

  • Develop tissue-targeted delivery systems: To specifically target IL-22 signaling in affected tissues

  • Explore combination therapies: Test IL-22 blockade alongside standard-of-care treatments

  • Monitor biomarkers of IL-22 activity: Develop assays to measure IL-22-regulated genes as pharmacodynamic markers

  • Consider biphasic effects: Design studies to account for potential protective versus pathogenic roles of IL-22 depending on disease stage

How should researchers interpret seemingly contradictory findings regarding IL-22 function in different disease models?

Contradictory findings regarding IL-22 function are common due to its context-dependent effects. When interpreting such findings, researchers should consider:

  • Disease stage: IL-22 may be protective during tissue repair but pathogenic during active inflammation

  • Genetic background: Different mouse strains (e.g., C57BL/6 vs. MRL/lpr) may show different IL-22 responses

  • Cellular source of IL-22: T cell-derived versus ILC3-derived IL-22 may have different implications

  • Target tissue receptor expression: Variation in IL-22RA1 expression across tissues affects response

  • Co-existing cytokine milieu: Presence of other cytokines may synergize with or antagonize IL-22 effects

For example, in inflammatory bowel disease, IL-22 has been reported to have both protective effects (promoting epithelial barrier repair) and pathogenic effects (enhancing neutrophil recruitment) . These seemingly contradictory roles likely reflect the complex biology of IL-22 rather than experimental artifacts.

What statistical approaches are most appropriate for analyzing IL-22-related experimental data in complex disease models?

For complex IL-22 experimental data, researchers should consider:

  • Mixed-effects models: To account for both fixed effects (treatment) and random effects (individual variation)

  • Longitudinal data analysis: For time-course experiments monitoring disease progression

  • Multivariate analysis: To examine relationships between multiple IL-22-regulated outcomes

  • Network analysis: To understand IL-22's position within broader cytokine networks

  • Machine learning approaches: For identifying patterns in complex datasets with multiple parameters

When comparing IL-22 KO, IL-22R KO, and control mice, researchers typically use one-way ANOVA for comparison among all groups, with post-hoc tests to identify specific group differences. For time-course data, repeated measures ANOVA or mixed-effects models are more appropriate .

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.