IL-17A drives proinflammatory responses through:
Chemokine induction: IL-8, CXCL1, and CCL20 recruit neutrophils and monocytes .
Cytokine amplification: Synergizes with TNF-α and IL-22 to enhance IL-6, G-CSF, and antimicrobial peptides .
Barrier maintenance: Strengthens epithelial tight junctions in mucosal tissues .
Autoimmunity: Elevated in rheumatoid arthritis, psoriasis, and multiple sclerosis .
Type 1 diabetes (T1D): IL-17+ CD4+ T cells exacerbate islet inflammation and apoptosis .
Cancer: Dual role—promotes tumor growth via angiogenesis while enhancing antitumor immunity .
SLE (lupus): IL-17 correlates with disease severity and promotes anti-dsDNA IgG production in plasma cells .
T1D: IL-17 synergizes with IFN-γ to induce islet cell apoptosis in vitro .
Therapeutic targeting: IL-17A-neutralizing antibodies (e.g., secukinumab) are FDA-approved for psoriasis and ankylosing spondylitis .
Antimicrobial defense: Essential for controlling Candida albicans and extracellular bacteria via neutrophil recruitment .
Gut homeostasis: Regulates microbiota composition through β-defensin secretion .
IL-17 Family Member | Sequence Homology to IL-17A | Primary Function |
---|---|---|
IL-17F | 55% | Redundant with IL-17A in inflammation |
IL-17E (IL-25) | 17% | Promotes type 2 immunity |
IL-17C | 23% | Epithelial immunity |
Human IL-17A is a 155 amino acid precursor protein that undergoes cleavage of a 19 amino acid signal peptide to yield a mature 136 amino acid protein. It functions primarily as a disulfide-linked homodimer with one potential N-linked glycosylation site . The protein signals through the obligate IL-17RA and IL-17RC heterodimeric receptor complex, which activates downstream inflammatory pathways .
For experimental purposes, recombinant IL-17A is often produced with histidine tags to facilitate purification using affinity chromatography. When designing experiments, researchers should consider:
Using both tagged and untagged versions to confirm biological activity isn't affected by the tag
Validating protein folding and dimerization through size exclusion chromatography
Confirming biological activity through cell-based assays measuring IL-6 or IL-8 production by fibroblasts
IL-17A is primarily produced by activated T cells, particularly the CD4+ T helper 17 (Th17) subset, which is characterized by expression of the transcription factor RORγt. Other sources include CD8+ (Tc17) cells, γδ T cells, natural killer T (NKT) cells, group 3 innate lymphoid cells (ILC3), and 'natural' Th17 cells .
To experimentally verify IL-17A production:
Flow cytometry: Stimulate PBMCs with PMA (50 ng/mL) and calcium ionophore (250 ng/mL) for 16 hours, then perform intracellular staining with anti-IL-17A antibodies
Western blot: Detect IL-17A in cell lysates and conditioned media from differentiated Th17 cells (approximately 15 kDa under reducing conditions)
Immunohistochemistry: Visualize IL-17A in tissue sections, where it localizes to lymphocytes in structures like tonsils
RT-PCR: Quantify IL-17A mRNA expression in sorted or cultured cell populations
Multiple techniques are available for detecting and quantifying human IL-17A:
ELISA/HTRF: For supernatants and biological fluids
Western blotting: For cell and tissue lysates
Flow cytometry: For cellular analysis
Immunohistochemistry: For tissue localization
When designing IL-17A detection experiments, researchers should:
Include recombinant IL-17A standards for quantification
Be aware of potential cross-reactivity (e.g., some antibodies show ~30% cross-reactivity with canine IL-17)
Validate detection in the specific biological matrix being studied
This differential potency has several experimental implications:
When studying IL-17 signaling, researchers must explicitly determine which family member is being targeted
Knockout or neutralization studies should account for potential compensation by other family members
Receptor blocking experiments need to consider the shared receptor components
For comprehensive signaling studies, researchers should:
Use specific neutralizing antibodies that distinguish between IL-17A, F, and A/F
Consider genetic approaches (CRISPR, siRNA) targeting individual ligands or receptor components
Employ receptor reporter systems to quantify signaling strength differences
IL-17A plays complex and seemingly paradoxical roles in viral infections, acting as both friend and foe in antiviral immunity . When designing studies to investigate these roles:
Timing considerations:
Early vs. late infection phases may show different IL-17A functions
Kinetic analyses of IL-17A production are essential to understand temporal roles
Viral specificity:
Tissue compartmentalization:
IL-17A functions differ between circulation and tissue microenvironments
Sample both blood and affected tissues when possible
Experimental systems:
In vitro cell culture systems should include relevant target cells
Animal models must be chosen carefully as IL-17A responses may differ between species
Human samples provide clinical relevance but experimental control is limited
Methodological approach:
Combine IL-17A neutralization, knockout, and overexpression approaches
Measure both direct antiviral effects and inflammatory consequences
Assess viral load, tissue damage, and immunopathology concurrently
IL-17A is implicated in multiple autoimmune diseases, requiring specialized experimental approaches:
Disease-specific considerations:
Human sample analysis:
Collect paired blood and affected tissue samples when possible
Consider disease stage and treatment status as confounding variables
Use multiparameter approaches to correlate IL-17A with other inflammatory markers
Experimental design:
Data interpretation:
Distinguishing between IL-17A homodimers and IL-17A/F heterodimers presents technical challenges due to structural similarities and cross-reactivity:
Immunoprecipitation approach:
ELISA-based methods:
Sandwich ELISA with capture antibody against one subunit and detection antibody against the other
Heterodimer-specific ELISA kits with validated specificity
Mass spectrometry:
Liquid chromatography-mass spectrometry can differentiate variants based on peptide mass and sequence
Requires sample purification but provides highest specificity
Functional bioassays:
Generating stable and reproducible Th17 cultures for IL-17A production can be challenging:
Optimal differentiation protocol:
Purify naïve CD4+ T cells (CD4+CD45RA+CD45RO-CD62L+)
Activate with plate-bound anti-CD3 (5 μg/mL) and soluble anti-CD28 (2 μg/mL)
Supplement with cytokine cocktail: IL-6 (20-30 ng/mL), TGF-β (1-5 ng/mL), IL-1β (10 ng/mL), IL-23 (20 ng/mL)
Block antagonistic pathways with anti-IFN-γ and anti-IL-4 antibodies
Critical variables to control:
Donor variability: Use consistent donor sources or pooled samples
Serum batch effects: Test and select optimal serum lots
Cell density: Maintain 1-2 × 10^6 cells/mL
Culture duration: Peak IL-17A production typically occurs at 5-7 days
Validation approaches:
Troubleshooting low IL-17A production:
Check cytokine quality and bioactivity
Optimize T cell activation strength
Consider alternative activation methods (e.g., αCD3/CD28 beads)
Exclude contaminating regulatory T cells
IL-17A affects various non-immune cells, including fibroblasts, epithelial cells, and endothelial cells:
Receptor expression verification:
Confirm IL-17RA and IL-17RC expression by flow cytometry or western blot
qPCR can quantify receptor expression levels
Consider receptor upregulation after inflammatory priming
Experimental design considerations:
Dose-response studies: IL-17A typically used at 1-100 ng/mL
Timing: Monitor responses at multiple timepoints (early: 0.5-6h; late: 24-72h)
Combined stimulation: Test IL-17A alone and with TNF-α for synergistic effects
Readout selection:
Transcriptional responses: RNA-seq or targeted gene panels
Protein secretion: ELISA for IL-6, IL-8, chemokines
Functional assays: Proliferation, migration, barrier function
Controls and validation:
Receptor blocking antibodies as specificity controls
Heat-inactivated IL-17A as negative control
siRNA knockdown of downstream mediators (ACT1, TRAF6)
IL-17A exhibits context-dependent roles in cancer, requiring nuanced experimental approaches:
Cancer-type specific considerations:
Experimental models:
Patient-derived xenografts maintain human tumor-infiltrating lymphocytes
Syngeneic mouse models preserve intact immune responses
In vitro co-cultures of tumor cells with IL-17A-producing cells
Methodological approaches:
Single-cell RNA sequencing to identify IL-17A-producing and responding populations
Spatial transcriptomics to map IL-17A activity within tumor microenvironment
IL-17A blockade or supplementation in immunotherapy combination studies
Clinical correlations:
Multiplex immunohistochemistry for IL-17A, cell type markers, and effector molecules
Correlation with patient outcomes and treatment responses
Integration with other immune parameters
With IL-17A-targeted therapeutics now in clinical use for autoimmune diseases, standardized preclinical evaluation methods are critical:
In vitro neutralization assays:
Inhibition of IL-17A-induced IL-6/IL-8 production by fibroblasts
Dose-response curves with fixed IL-17A concentration (typically 10 ng/mL)
Calculate IC50 values for comparing different neutralizing antibodies
Binding characterization:
Surface plasmon resonance for affinity and kinetics
Epitope mapping to determine binding sites
Cross-reactivity testing with other IL-17 family members
Animal model testing:
Psoriasis models: Imiquimod-induced or IL-23-induced
Arthritis models: Collagen-induced arthritis
Humanized models for human-specific therapeutics
Pharmacokinetic/pharmacodynamic studies:
Serum concentration monitoring
Target engagement biomarkers (free vs. bound IL-17A)
Downstream inflammatory marker suppression
Safety evaluation:
Monitor for increased susceptibility to infections
Evaluate for immunogenicity
Check for compensatory cytokine production
Modern IL-17A research increasingly requires integration with systems-level approaches:
Network analysis methods:
Protein-protein interaction networks centered on IL-17A signaling components
Pathway enrichment analysis of IL-17A-induced transcriptional changes
Integration of IL-17A with other cytokine networks (IL-23, TNF, IL-1)
Multi-omics integration:
Combine transcriptomics, proteomics, and metabolomics data
Correlate IL-17A levels with global metabolic signatures
Integrate with epigenetic modifications at IL-17A-responsive genes
Computational modeling:
Agent-based models of IL-17A in tissue microenvironments
Ordinary differential equations to model IL-17A signaling dynamics
Machine learning to predict IL-17A responses from baseline parameters
Practical approach:
Design experiments that capture multiple parameters simultaneously
Include time-course elements to capture dynamic responses
Collaborate with computational biologists for advanced analyses
IL-17A research presents several common challenges:
Antibody specificity issues:
Cell source variations:
IL-17A production is heterogeneous across different T cell subpopulations
Clearly define and characterize the source cells in experiments
Account for donor-to-donor variability in human samples
Protein stability concerns:
IL-17A can degrade during storage and experimental manipulation
Use carrier proteins for dilute solutions
Avoid repeated freeze-thaw cycles
Validate activity of stored recombinant IL-17A periodically
Context-dependent functions:
IL-17A effects vary dramatically by tissue and disease context
Include relevant tissue-specific cells in experimental systems
Consider the inflammatory milieu when interpreting results
Detection sensitivity:
IL-17A may be present at low concentrations in some samples
Employ sensitive detection methods with appropriate lower limits of quantification
Consider sample concentration techniques for dilute biological fluids
Ensuring reproducibility in IL-17A research requires:
Standard operating procedures:
Detailed protocols for cell isolation, culture, and stimulation
Consistent reagent sources and lot numbers
Standardized detection methods with calibrated standards
Quality control measures:
Include positive and negative controls in every experiment
Use validated recombinant IL-17A standards
Implement blinding when analyzing samples
Validation approaches:
Confirm key findings with multiple detection methods
Replicate experiments with cells from different donors
Use genetic approaches (siRNA, CRISPR) to complement neutralization studies
Reporting standards:
Follow field-standard reporting guidelines
Provide detailed methods including antibody clone numbers
Share raw data when possible to enable reanalysis
Interleukin-17 (IL-17) is a proinflammatory cytokine produced by activated T cells. It plays a crucial role in the immune response by regulating the activities of NF-kappaB and mitogen-activated protein kinases . IL-17 stimulates the expression of other cytokines such as IL-6 and cyclooxygenase-2 (PTGS2/COX-2), and enhances the production of nitric oxide (NO) .
The human recombinant IL-17 with a His tag is produced in Escherichia coli (E. coli). It is a single, non-glycosylated polypeptide chain containing 132 amino acids, with a molecular weight of approximately 19.62 kDa . The His tag, which is a sequence of histidine residues, is fused to the amino-terminal end of the protein, adding an additional 4.5 kDa to the molecular weight .
The recombinant IL-17 is purified using proprietary chromatographic techniques to achieve a purity greater than 95% as determined by SDS-PAGE . The protein is supplied as a sterile filtered clear solution in 25 mM sodium acetate (pH 4.8) and 50% glycerol . For optimal stability, it should be stored at 4°C if used within 2-4 weeks, or frozen at -20°C for longer periods .
IL-17 is known for its role in promoting inflammation. It can stimulate the production of other proinflammatory cytokines and chemokines, which recruit immune cells to sites of infection or injury . High levels of IL-17 are associated with several chronic inflammatory diseases, including rheumatoid arthritis, psoriasis, and multiple sclerosis .
ProSpec’s recombinant IL-17 is intended for laboratory research use only and should not be used as drugs, agricultural or pesticidal products, food additives, or household chemicals . Proper safety protocols should be followed when handling this protein to avoid contamination and ensure accurate experimental results .