IL-22 antibodies are monoclonal antibodies (mAbs) designed to target interleukin-22 (IL-22), a cytokine in the IL-10 family. These antibodies are used in research and clinical settings to modulate IL-22 activity, which plays dual roles in host defense and inflammatory pathology . IL-22 is primarily secreted by Th17 cells, NK cells, and γδ T cells, acting on non-immune cells such as epithelial cells and keratinocytes . Therapeutic anti-IL-22 antibodies are being explored for autoimmune diseases, inflammatory disorders, and cancer .
IL-22 is a 20 kDa α-helical cytokine encoded on chromosome 12q15 in humans . It signals through a heterodimeric receptor complex (IL-22R1 and IL-10R2), activating the JAK-STAT pathway . A soluble receptor, IL-22BP, acts as a natural antagonist .
Anti-IL-22 antibodies bind to IL-22 with high specificity, blocking its interaction with IL-22R1/IL-10R2 and inhibiting downstream STAT3 phosphorylation . Key clones include:
Atopic Dermatitis (AD): Fezakinumab (anti-IL-22 mAb) reduced SCORAD scores by 21.6±3.8 in severe AD patients vs. 9.6±4.2 for placebo at 12 weeks .
Osteoarthritis (OA): Anti-IL-22/IL-22R antibodies reduced cartilage degradation and pain in murine models .
Graft-versus-Host Disease (GVHD): Anti-IL-22 treatment decreased IFN-γ+ and TNF-α+ T cells while expanding regulatory T cells (Tregs) .
IL-22 promotes epithelial proliferation and tumorigenesis. BioAtla’s CPS09 suppressed IL-22-induced STAT3 activation and reduced tumor growth in colitis-associated colorectal cancer models .
Dual Roles: IL-22 enhances antimicrobial defense in the gut and skin but exacerbates inflammation in diseases like psoriasis .
Th17 Axis Modulation: IL-22 synergizes with IL-17A/IL-17F to regulate keratinocyte proliferation and innate immunity .
Context-Dependent Effects: IL-22’s protective vs. pathogenic roles require patient stratification .
Limited Clinical Success: Fezakinumab showed efficacy only in severe AD subgroups .
Applications : IHC
Sample type: cells
Review: Regarding cellular inflammation, IL-22-induced increases in the levels of IL1β, IL17, and IL6 in the supernatant were significantly reduced by baicalin treatment.
IL-22 is a 20 kDa alpha-helical cytokine belonging to the IL-10 family, which includes IL-19, IL-20, IL-24, IL-26, IL-28, IL-29, and interferons types I and II . The IL-22 gene in humans is located on the q arm of chromosome 12 and shares structural homology with IL-10 . This cytokine is primarily secreted by activated T helper 17 (Th17) cells and natural killer (NK) cells, though other T cell subsets have been identified as additional sources .
In vitro Th17 culture systems have demonstrated that IL-22 expression is induced more robustly in response to IL-23 than to IL-6 or TGF-beta, suggesting that in vivo, this cytokine may be predominantly secreted by fully differentiated Th17 cells rather than developing ones . This pattern of expression indicates IL-22's important role in the later phases of adaptive immune responses.
IL-22 signals through a heterodimeric receptor complex consisting of two distinct components:
IL-22R1 (also known as IL-22 Ralpha1 and CRF2-9): A primary receptor component weighing approximately 65 kDa that functions as a transmembrane glycoprotein belonging to the type II cytokine receptor family .
IL-10R2: An accessory receptor that is shared with several other cytokines, including IL-10, IL-27, IL-28, and IL-29 .
Unlike many cytokine receptors found on immune cells, IL-22R1 expression is predominantly restricted to non-hematopoietic stromal cells, particularly epithelial cells of the lung and gastrointestinal tract . This distinct expression pattern creates a unique communication channel through which immune cells can signal to tissue cells. Interestingly, differential expression of IL-22R1 has been observed in cancer cell lines, with positive detection in HT-29 human colon adenocarcinoma cells but negative expression in HepG2 human hepatocellular carcinoma cells .
IL-22 antibodies serve several crucial functions in immunological research:
Detection and quantification of IL-22 expression through techniques such as intracellular staining followed by flow cytometric analysis .
Neutralization of IL-22 activity in experimental disease models, such as in acute graft-versus-host disease (aGVHD) studies .
Immunohistochemical analysis of tissues to examine IL-22 receptor expression patterns .
Mechanistic investigations of IL-22 signaling pathways and downstream effects on target cells.
For example, the 22URTI monoclonal antibody has been pre-titrated and validated for intracellular staining in flow cytometry analyses of restimulated, Th17-polarized CD4+ human peripheral blood cells .
When using IL-22 antibodies for flow cytometric analysis, researchers should consider the following parameters for optimal results:
Antibody concentration: For the 22URTI PE-conjugated antibody, 5 μL (0.01 μg) per test is recommended, where a test is defined as the amount of antibody needed to stain a cell sample in a final volume of 100 μL .
Cell preparation: Both Th17-polarized CD4+ normal human peripheral blood cells and stimulated total normal human peripheral blood cells have been validated as appropriate sample types .
Cell numbers: While empirical determination is ideal, cell numbers can range from 10^5 to 10^8 cells per test .
Optical parameters: For PE-conjugated antibodies, use excitation wavelengths of 488-561 nm and emission detection at 578 nm. Compatible lasers include blue, green, and yellow-green lasers .
Filtration: Ensure antibody preparations are 0.2 μm post-manufacturing filtered to avoid particulate contamination .
Administration of anti-IL-22 antibody has demonstrated significant immunomodulatory effects in experimental models such as acute graft-versus-host disease (aGVHD). In a mouse aGVHD model, IL-22 antibody treatment significantly reduced disease progression . The immunological mechanisms underlying this therapeutic effect involve several key alterations in immune cell populations:
Decreased percentages of IFN-γ+ and TNF-α+ T cells, indicating reduced pro-inflammatory T cell responses .
Increased numbers of Foxp3+ regulatory T cells (Tregs), suggesting enhanced immunoregulatory capacity .
Modulation of CD11b+ antigen-presenting cells, which appears essential for the protective effects of IL-22 antibody treatment .
These findings indicate that IL-22 antibody treatment not only neutralizes IL-22 activity directly but also induces broader changes in the immune microenvironment that promote immunoregulation over inflammation.
Anti-IL-22 antibody treatment induces complex changes in cellular signaling and cytokine networks. Research has identified several key molecular mechanisms:
Cytokine modulation: IL-22 antibody treatment reduces the expression of pro-inflammatory cytokines IL-6, IFN-γ, and IL-18 while enhancing the production of immunoregulatory cytokines IL-10 and TGF-β .
Altered co-stimulatory molecule expression: Treatment with IL-22 antibody downregulates expression of co-stimulatory molecules CD80, CD86, and MHC-II on antigen-presenting cells .
Modulation of dendritic cell maturation: IL-22 antibody treatment appears to regulate the transformation of mature dendritic cells to immature dendritic cells, as indicated by changes in CD83 expression .
Reduced STAT signaling: The phosphorylation of transcription factors STAT1 and STAT3 is downregulated following IL-22 antibody administration .
Decreased expression of IL-22-related transcription factors: Levels of β-defensin and Reg3γ are reduced after IL-22 antibody treatment .
These molecular changes collectively create an environment more conducive to Treg expansion and less supportive of effector T cell proliferation, both in vitro and in vivo .
IL-22 plays a critical role in maintaining epithelial tissue integrity, particularly at barrier surfaces such as the intestinal epithelium. IL-22 antibodies provide valuable tools for investigating these functions through:
In vivo blocking experiments to assess the impact of IL-22 signaling on epithelial regeneration following injury.
Immunohistochemical studies to correlate IL-22 receptor expression patterns with tissue responses, as demonstrated in the differential expression between HT-29 and HepG2 cancer cell lines .
Investigation of IL-22-mediated activation of epithelial STAT3 signaling, which promotes cell survival, proliferation, and wound healing .
Analysis of antimicrobial peptide expression (like β-defensin and Reg3γ) that is regulated by IL-22 signaling .
Exploration of the paradoxical roles of IL-22 in different disease models, such as its protective function in experimental colitis versus its pathogenic role in certain autoimmune conditions .
These applications enable researchers to decipher the context-dependent functions of IL-22 at epithelial surfaces.
Bibliometric analysis of IL-22 research from 2014 to 2023 reveals several significant trends:
Steady increase in research output: The number of publications on IL-22 has consistently grown over this period, reflecting increased interest in this cytokine .
Geographic distribution of research: The United States and China are the primary contributors to IL-22 research, with the Medical Research Institute (INSERM) and the University of California system being the most active institutions .
Publication patterns: Frontiers of Immunology published the most articles on IL-22 and received the highest number of citations in this field .
Citation metrics: IL-22 research publications have accumulated 122,063 citations, averaging 30.96 citations per article, with an H-index of 137 for all publications .
Research focus: Current IL-22 studies predominantly concentrate on immunology and cell biology, with increasing interest in therapeutic applications .
This bibliometric data suggests that IL-22 research remains a highly active and evolving field with significant potential for future discoveries.
IL-22 antibodies are instrumental in exploring the therapeutic potential of IL-22 pathway modulation across various disease contexts:
Autoimmune conditions: IL-22 antibodies help elucidate IL-22's role in diseases such as rheumatoid arthritis, psoriasis, and acute hepatitis, where IL-22 has been implicated as a pathogenic factor .
Transplantation medicine: Anti-IL-22 antibody treatment has demonstrated protective effects in aGVHD models, suggesting potential applications in transplantation medicine .
Inflammatory bowel disease: By blocking IL-22, researchers can investigate its paradoxical functions in intestinal inflammation, where it appears to both promote inflammatory responses and protect epithelial integrity .
Cancer research: The differential expression of IL-22 receptors across cancer cell lines (e.g., positive in HT-29 but negative in HepG2) suggests potential applications for IL-22 antibodies in studying cancer biology .
Precision medicine approaches: The development of humanized IL-22 antibodies may contribute to targeted therapeutic strategies that selectively modulate specific aspects of immune responses.
When conducting experiments with IL-22 antibodies, researchers should implement the following quality control measures:
Antibody validation: Confirm specificity using positive and negative controls, as demonstrated in the detection of IL-22 R alpha 1 in HT-29 cells (positive) versus HepG2 cells (negative) .
Titration optimization: Pre-titrate antibodies to determine optimal concentrations, as illustrated with the 22URTI antibody recommendation of 5 μL (0.01 μg) per test for flow cytometry .
Cell preparation protocols: Standardize cell isolation and stimulation protocols, particularly for detecting intracellular IL-22, which requires appropriate cell activation .
Species specificity: Verify cross-reactivity when working with animal models, as antibody specificity may vary across species.
Storage and handling: Maintain appropriate storage conditions and avoid repeated freeze-thaw cycles to preserve antibody functionality.
Experimental controls: Include isotype controls, FMO (fluorescence minus one) controls for flow cytometry, and biological negative and positive controls in each experiment.
To effectively investigate IL-22 signaling mechanisms, consider the following experimental design principles:
Receptor expression analysis: Confirm expression of both IL-22R1 and IL-10R2 components in target cells, as both are required for functional IL-22 signaling .
Signaling pathway assessment: Measure activation of key downstream pathways, particularly STAT3 phosphorylation, which is central to IL-22 signaling .
Temporal dynamics: Evaluate both acute and chronic effects of IL-22 signaling, as responses may evolve over time.
Cell-type specificity: Since IL-22R1 expression is restricted to non-hematopoietic cells, use appropriate epithelial or stromal cell models .
Context dependency: Consider the microenvironmental context, as IL-22 effects vary significantly depending on the presence of other cytokines and cellular interactions.
Loss-of-function and gain-of-function approaches: Combine IL-22 neutralization (using antibodies) with IL-22 supplementation experiments to comprehensively map signaling effects.