IL17F regulates inflammatory responses by inducing cytokines (e.g., IL-6, IL-8) and chemokines (e.g., G-CSF, MCP-1) . It is expressed by Th17 cells, mast cells, and epithelial cells , contributing to:
Autoimmune diseases: Psoriasis, rheumatoid arthritis, and inflammatory bowel disease .
Infection defense: Neutrophil recruitment against Candida and Staphylococcus aureus .
IL17F antibodies are used in:
| Antibody Product | Clone | Application | Source |
|---|---|---|---|
| PE anti-mouse IL17F | eBio18F10 | Flow cytometry | |
| Human IL17F Antibody | MAB13352 | ELISA, Western blot | |
| IL17F antibody (ab168194) | Polyclonal | IHC, Western blot |
Bimekizumab, a humanized IgG1 antibody, simultaneously neutralizes IL17A and IL17F, showing efficacy in:
Psoriasis: Achieved 86.7% skin clearance (PASI 100) vs. placebo .
Arthritis: Reduced joint inflammation in collagen-induced arthritis models .
| Trial | Outcome | Source |
|---|---|---|
| Phase II Psoriasis Study | 80% ACR20 response vs. 16.7% placebo (p < 0.001) | |
| Collagen-Induced Arthritis | Combined IL17A/F blockade suppressed IL-6/G-CSF levels |
IL17F induces IL-6, IL-8, and TGF-β in epithelial cells , while anti-IL17F antibodies reduce neutrophil chemotaxis . Co-culture studies reveal cross-regulation: anti-IL17F antibodies downregulate IL17A production .
IL17F-deficient mice exhibit impaired defense against oropharyngeal candidiasis, highlighting its role in antifungal immunity .
IL-17F is a homodimeric, 34 kDa cytokine belonging to the IL-17 family, which includes IL-17A, IL-17B, IL-17C, IL-17D, and IL-17E (also known as IL-25). Among these family members, IL-17F shares the highest homology with IL-17A. Both IL-17A and IL-17F are predominantly produced by T helper 17 (Th17) cells, while other family members are produced more widely by different cell types .
Despite their structural similarities, IL-17F and IL-17A possess both overlapping and independent functions as demonstrated through knockout studies. IL-17F is involved in allergic airway inflammation and can induce several cytokines, chemokines, and adhesion molecules in bronchial epithelial cells, vein endothelial cells, fibroblasts, and eosinophils . The relative difference in potency between recombinant human IL-17A and IL-17F is approximately 100-fold, with IL-17A being more potent .
In inflammatory conditions, IL-17F may exist as a homodimer or as a heterodimer with IL-17A (IL-17A/F). Quantification studies in patients with psoriasis, psoriatic arthritis, and ankylosing spondylitis have revealed that IL-17F is significantly more abundant than IL-17A (>30-fold) .
To validate IL-17F antibody specificity, researchers should implement a multi-step approach:
Cross-reactivity testing: Evaluate binding to recombinant human IL-17 family members (IL-17A through IL-17E) using ELISA. Antibodies should demonstrate high specificity for IL-17F with minimal cross-reactivity to other family members, particularly IL-17A which shares the highest structural similarity .
Competitive binding assays: Perform competitive binding experiments where unlabeled IL-17F or other IL-17 family cytokines compete with labeled IL-17F for antibody binding. A specific antibody will show displacement only with unlabeled IL-17F.
Functional validation: Test the antibody's ability to neutralize IL-17F bioactivity in cell-based assays. For example, measure inhibition of IL-17F-induced IL-6 production in normal human dermal fibroblasts (NHDFs) stimulated with TNFα and IL-17F .
Western blot and immunoprecipitation: Confirm specificity by detecting endogenous or recombinant IL-17F at the expected molecular weight (~34 kDa for homodimer).
Knockout/knockdown controls: Include IL-17F knockout or knockdown samples as negative controls when possible to confirm specificity.
For optimal IL-17F detection in biological samples, researchers should consider these methodological approaches:
Flow cytometry (intracellular staining):
Fix and permeabilize cells using a commercial kit designed for cytokine detection
Use 5 μL (0.5 μg) of IL-17F antibody per test (10^5 to 10^8 cells) in 100 μL final volume
Include protein transport inhibitors during cell stimulation (e.g., brefeldin A or monensin)
For optimal results with PE-conjugated antibodies, use excitation at 488-561 nm and emission detection at 578 nm
ELISA detection:
Coat high-binding ELISA plates with recombinant IL-17F or capture antibody
For direct binding studies, add antibody titrations starting at 10 μg/mL
Cell stimulation protocols for IL-17F induction:
For T cells: Stimulate with PMA/ionomycin for 4-6 hours with protein transport inhibitor
For Th17 polarization: Culture naive CD4+ T cells with TGF-β, IL-6, IL-1β, IL-23, anti-IFN-γ, and anti-IL-4 for 5-6 days
Sample preparation considerations:
Serum/plasma: Centrifuge blood samples within 30 minutes of collection
Tissue samples: Homogenize in PBS with protease inhibitors before analysis
Cell culture supernatants: Collect after appropriate stimulation period
Distinguishing between IL-17F homodimers and IL-17A/F heterodimers requires sophisticated experimental approaches:
Antibody-based discrimination:
Use combination of specific antibodies targeting epitopes unique to IL-17F homodimers versus those present on heterodimers
Implement sandwich ELISA systems with capture antibodies specific for one subunit and detection antibodies for the other subunit
Affinity purification:
Perform sequential immunoprecipitation with anti-IL-17A and anti-IL-17F antibodies
Analyze the resulting fractions by western blot to identify homodimers versus heterodimers
Functional characterization:
Utilize differential receptor binding characteristics (IL-17RC binds IL-17F with higher affinity than IL-17A)
Compare neutralization effects of selective antibodies against homodimers versus heterodimers in bioassays
Mass spectrometry:
Implement proteomics approaches to identify unique peptide signatures from homodimers versus heterodimers
Use crosslinking mass spectrometry to characterize dimeric interfaces
Research has shown that in vitro cultured Th17 cells can express IL-17F and IL-17A homodimers and IL-17A/F heterodimers depending on culture conditions and differentiation state . This heterogeneity must be considered when designing experimental systems targeting IL-17F biology.
When designing experiments to compare IL-17A versus IL-17F neutralization in disease models, researchers should consider these methodological factors:
Antibody selection and validation:
Ensure comparable affinity and neutralizing potency of anti-IL-17A and anti-IL-17F antibodies
Validate neutralization capacity using in vitro bioassays before in vivo application
Dosing considerations:
Account for differences in potency between IL-17A and IL-17F (~100-fold in humans)
Implement dose-response studies to determine optimal antibody concentrations
For combined neutralization studies, consider using 100 μg of each specific antibody daily via intraperitoneal administration (based on mouse studies)
Experimental design:
Include appropriate control groups: isotype controls, single neutralization (anti-IL-17A or anti-IL-17F alone), and combination (anti-IL-17A + anti-IL-17F)
Consider the temporal expression patterns: IL-17A may be induced earlier than IL-17F in disease progression
Readout selection:
Measure both direct neutralization outcomes and downstream effects
Monitor disease-specific parameters (e.g., arthritic score, airway inflammation)
Assess changes in cytokine profiles, particularly IL-6 and G-CSF which are often suppressed following effective IL-17A neutralization
Evaluate cellular infiltration patterns in affected tissues
Analysis of IL-17A/F ratios:
Determine baseline ratios of IL-17F to IL-17A in your model system
Consider that when the ratio of IL-17F to IL-17A is ≥10-fold, dual neutralization may show differentiated effects compared to IL-17A inhibition alone
Affinity maturation can dramatically enhance IL-17F antibody performance in research applications, as evidenced by the development of dual-specific antibodies like bimekizumab:
Effects on binding kinetics:
The affinity maturation process can significantly improve binding constants. For example, the bimekizumab development process demonstrated a 43-fold increase in affinity for IL-17F (from KD of 1510 pM to 35 pM) and a 4-fold increase in affinity for IL-17A (from KD of 29 pM to 7 pM) compared to its parent antibody .
Improvements in neutralization potency:
Affinity-matured antibodies demonstrate enhanced neutralization capacity in functional assays. In fibroblast stimulation assays with TNFα and IL-17F, the affinity-matured antibody 496.g3 showed significantly greater inhibition of IL-6 production compared to its parent antibody 496.g1 .
Impact on experimental sensitivity:
Higher affinity antibodies can:
Detect lower concentrations of target protein
Remain effective at lower dosages
Provide more consistent results across experimental replicates
Increase signal-to-noise ratio in detection applications
Methodological considerations:
When using affinity-matured antibodies, researchers should:
Re-validate optimal working concentrations
Consider potential off-target effects from increased binding to related proteins
Adjust incubation times and washing protocols according to binding kinetics
Document the specific clone and its affinity parameters in experimental reports
Developing therapeutic antibodies targeting both IL-17A and IL-17F presents several important technical and biological considerations:
Target biology understanding:
Different tissue expression patterns of IL-17A and IL-17F
Relative contributions of each cytokine to specific disease pathophysiology
Potential redundancy or synergy between IL-17A and IL-17F functions
Antibody engineering approaches:
Starting point selection: Researchers may begin with an IL-17A-specific antibody and enhance IL-17F binding through targeted mutations
Affinity maturation strategies: For bimekizumab development, five mutations in the light chain variable region of the parent antibody enhanced binding to both cytokines
Epitope selection: Target conserved epitopes between IL-17A and IL-17F while maintaining specificity within the IL-17 family
Pharmacokinetic/pharmacodynamic considerations:
Higher antibody concentrations may be required for dual targeting compared to single-cytokine neutralization
When the ratio of IL-17F to IL-17A is ≥10-fold (as observed in many inflammatory conditions), dual neutralization shows differentiated benefits
Target-mediated drug disposition modeling helps predict tissue penetration and neutralization capacity
Functional validation strategies:
Compare single versus dual neutralization in complex bioassays
Test varying ratios of IL-17F:IL-17A to determine optimal neutralization conditions
Evaluate inhibition across multiple cell types and readouts relevant to disease biology
The dual-targeting approach is supported by research showing that IL-17F is significantly more abundant than IL-17A in inflammatory conditions (>30-fold), and that when the ratio of IL-17F to IL-17A is ≥10-fold, dual neutralization demonstrates superior inflammatory suppression compared to IL-17A inhibition alone .
Several critical factors can affect IL-17F antibody performance in intracellular staining experiments:
Cell stimulation protocols:
Optimal stimulation conditions: PMA (50 ng/mL) + ionomycin (1 μg/mL) for 4-6 hours
Protein transport inhibitor selection: Brefeldin A works better for most cytokines, including IL-17F
Timing: IL-17F expression peaks later than some other cytokines, requiring longer stimulation
Fixation and permeabilization:
Fixative selection: Paraformaldehyde (4%) preserves cellular morphology while maintaining epitope accessibility
Permeabilization agent: Saponin-based buffers typically work well for cytokine detection
Incubation times: Excessive permeabilization can reduce signal intensity
Antibody parameters:
Optimal concentration: 5 μL (0.5 μg) per test for flow cytometric analysis
Clone selection: The SHLR17 monoclonal antibody clone has been validated for intracellular staining of human IL-17F
Fluorophore selection: PE conjugates (excitation: 488-561 nm; emission: 578 nm) provide good signal-to-noise ratio
Technical considerations:
Cell number optimization: Test range from 10^5 to 10^8 cells per 100 μL test
Buffer composition: Presence of serum proteins can reduce non-specific binding
Washing steps: Insufficient washing can increase background
To optimize IL-17F neutralization experiments for studying inflammatory pathways:
Cell model selection:
Normal human dermal fibroblasts (NHDFs) respond robustly to IL-17F stimulation, especially in combination with TNFα
Bronchial epithelial cells are appropriate for studying airway inflammation models
Primary cells generally provide more physiologically relevant responses than cell lines
Stimulation conditions:
Use IL-17F in combination with TNFα for synergistic induction of inflammatory mediators
For human cell models: IL-17F (25 nM) combined with TNFα (0.025 nM) effectively stimulates IL-6 production
Pre-incubate neutralizing antibodies with cytokines for 1 hour before addition to cells
Standard stimulation period: 18-20 hours for robust cytokine production
Readout selection:
Primary readouts: IL-6 production serves as a reliable indicator of IL-17F activity
Additional informative markers: CXCL1, CXCL8, and CCL20 production
Detection method: Homogeneous time-resolved fluorescence provides sensitive quantification
Experimental controls:
Include IL-17F alone, TNFα alone, and combination conditions to demonstrate synergy
Include isotype control antibodies at equivalent concentrations
For dual-neutralization studies, include single-neutralization conditions (anti-IL-17A or anti-IL-17F alone)
Antibody titration:
Perform dose-response experiments with neutralizing antibodies
For combined IL-17A/F neutralization, test different antibody ratios and concentrations
For evaluating IL-17F antibody efficacy in animal disease models, researchers should implement these strategic approaches:
Model selection considerations:
Collagen-induced arthritis (CIA) provides a well-characterized model for studying IL-17 biology
House dust mite (HDM) and toluene diisocyanate (TDI) models are effective for studying airway inflammation
Consider species differences: The potency difference between mouse IL-17A and IL-17F is ~10,000-fold, compared to ~100-fold in humans
Administration protocol:
Dosage: 100 μg of specific antibody daily via intraperitoneal route
Control groups: Include isotype control antibodies (100 μg) for each specific antibody
For evaluating combined effects: Administer 100 μg each of anti-IL-17A and anti-IL-17F antibodies
Timing: In CIA models, initiate treatment from day 20 post-immunization
Assessment parameters:
Clinical scoring: Monitor disease-specific parameters (e.g., arthritic score)
Cytokine profiling: Measure serum levels of IL-6, G-CSF, IFN-γ, IL-1β, TNF-α, and GM-CSF
Cellular analysis: Evaluate inflammatory cell infiltration by flow cytometry
Histopathological evaluation: Assess tissue damage and inflammation
Temporal considerations:
Monitor the differential induction timing of IL-17A and IL-17F
In CIA models, IL-17A is induced first (during initiation phase), while IL-17F appears with the onset of arthritis
Only a small percentage of CD4+ T cells co-express both IL-17A and IL-17F
Data interpretation:
Compare single versus combined neutralization effects
Evaluate systemic versus local effects of neutralization
Consider unexpected findings: Some studies report increased IL-6 levels with anti-IL-17F antibody treatment alone
When faced with contradictory findings in IL-17F neutralization studies, researchers should systematically evaluate these factors:
Species and model differences:
Mouse versus human IL-17F potency varies dramatically (~10,000-fold difference in mice versus ~100-fold in humans compared to IL-17A)
Disease model selection affects cytokine dominance patterns
Genetic background of animal models influences IL-17 pathway activities
Antibody characteristics:
Affinity differences between antibodies used in different studies
Epitope specificity affecting neutralization of different forms (monomer, homodimer, heterodimer)
Isotype differences impacting in vivo half-life and effector functions
Experimental timing:
IL-17A is induced earlier than IL-17F in some inflammatory models
Intervention timing relative to disease phase (preventive versus therapeutic)
Duration of neutralization treatment
Readout selection:
Direct versus indirect measures of IL-17F activity
Cellular versus molecular endpoints
Acute versus chronic outcome measures
Contextual cytokine environment:
Ratio of IL-17F to IL-17A in the specific model (~30-fold higher IL-17F in human inflammatory conditions)
Presence of synergizing factors like TNFα
Compensatory mechanisms in chronic neutralization settings
For example, in collagen-induced arthritis studies, anti-IL-17F antibody treatment alone did not reduce arthritis severity, while anti-IL-17A or combined anti-IL-17A/F antibody treatment significantly reduced disease. Surprisingly, IL-6 levels were actually increased in mice receiving anti-IL-17F antibody compared to isotype controls, suggesting complex regulatory mechanisms .
Quantitative assessment of IL-17F antibody efficacy in complex biological systems requires multi-dimensional analysis approaches:
Pharmacokinetic/pharmacodynamic modeling:
Target-mediated drug disposition models predict antibody distribution and IL-17F occupancy in different tissues
Allometric scaling to predict human pharmacokinetic parameters from animal data
Simulation of percentage binding at different doses and intervals
Dose-response relationship analysis:
EC50 determination for neutralization of IL-17F-induced cellular responses
Comparison of potency shifts in the presence of other cytokines (e.g., TNFα)
Multiparameter biomarker assessment:
Cytokine network analysis: Measure changes in multiple downstream cytokines (IL-6, G-CSF)
Transcriptomics: Evaluate global gene expression changes following neutralization
Phosphoprotein analysis: Quantify changes in IL-17 signaling pathway activation
Mathematical modeling of pathway inhibition:
Systems biology approaches to model IL-17 pathway dynamics
Network analysis to identify key nodes affected by IL-17F neutralization
Predictive modeling of therapeutic outcomes based on baseline parameters
Standardized reporting metrics:
Neutralization potency (IC50 values)
Receptor occupancy percentages in different tissues
Biomarker modulation indices (percent change from baseline)
Area under the effect curve (AUEC) for temporal integration of responses
For example, when developing bimekizumab, researchers used target-mediated drug disposition modeling to predict that following 160 mg IV dosing every 4 weeks, IL-17A would be completely bound in plasma and >95% bound in skin, but IL-17F would show <50% occupancy in skin. This informed subsequent affinity maturation efforts to improve IL-17F binding .
Several cutting-edge approaches are enhancing precision in IL-17F research:
Single-cell analysis technologies:
Single-cell RNA sequencing to identify discrete IL-17F-producing cell populations
Mass cytometry (CyTOF) for high-dimensional characterization of IL-17F-producing cells
Imaging mass cytometry to visualize IL-17F production in tissue microenvironments
Advanced protein engineering:
Bispecific antibody formats targeting IL-17F and synergistic partners
Conditional activation systems for temporal control of IL-17F neutralization
Tissue-targeted antibody delivery to improve local neutralization efficiency
In vivo imaging techniques:
Immuno-PET with radiolabeled anti-IL-17F antibodies to track tissue distribution
Intravital microscopy to visualize IL-17F neutralization dynamics in real-time
Bioluminescence reporters to monitor IL-17F pathway activation longitudinally
Computational approaches:
Machine learning algorithms to predict IL-17F-dependent gene networks
Molecular dynamics simulations of antibody-cytokine interactions
Systems pharmacology models integrating multi-omics data
CRISPR-based methodologies:
Precise genetic manipulation of IL-17F and receptor components
CRISPR activation/repression systems for temporal control of IL-17F expression
Base editing to introduce specific mutations in IL-17F pathway components
These advanced technologies will help address remaining questions about IL-17F biology, including: tissue-specific roles, temporal dynamics of expression, heterodimer versus homodimer functional differences, and optimal targeting strategies for different inflammatory conditions.
Deepening understanding of IL-17F biology is driving several key areas for next-generation therapeutic antibody development:
Pathway-selective targeting:
Designing antibodies that selectively inhibit specific downstream pathways activated by IL-17F
Developing antibodies that preferentially neutralize either homodimers or heterodimers
Creating antibodies with differential tissue distribution profiles
Combination targeting strategies:
Dual-variable domain antibodies targeting IL-17F and synergistic cytokines
Bispecific antibodies neutralizing IL-17F while simultaneously engaging immune modulatory receptors
Antibody-cytokine fusion proteins combining IL-17F neutralization with delivery of anti-inflammatory cytokines
Enhanced tissue targeting:
Antibody engineering for improved blood-brain barrier penetration in neuroinflammatory conditions
Pulmonary delivery systems for targeting airway inflammation
pH-responsive antibodies for enhanced function in inflammatory microenvironments
Potency and selectivity optimization:
Structure-guided design targeting unique epitopes on IL-17F
Affinity maturation strategies focusing on tissue-specific efficacy
Engineering for improved binding kinetics rather than equilibrium binding constants
Predictive biomarkers:
Developing companion diagnostics to identify patients likely to benefit from IL-17F targeting
Establishing IL-17F:IL-17A ratio thresholds predictive of dual-targeting efficacy
Identifying genetic polymorphisms affecting IL-17F pathway activation
The understanding that IL-17F is significantly more abundant than IL-17A in inflammatory conditions (>30-fold) , and that dual neutralization shows superior effects at IL-17F:IL-17A ratios ≥10:1, suggests that antibodies effectively targeting both cytokines may provide enhanced therapeutic benefits for conditions like psoriasis, psoriatic arthritis, and ankylosing spondylitis.