IL17F Antibody

Shipped with Ice Packs
In Stock

Description

Biological Functions

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 .

  • Cancer immunity: Modulation of tumor microenvironments .

Research Tools

IL17F antibodies are used in:

  • Flow cytometry: Detecting intracellular IL17F in Th17 cells (e.g., PE-conjugated eBio18F10 clone) .

  • ELISA: Quantifying IL17F in serum or tissue lysates .

  • Western blotting: Analyzing IL17F protein expression .

Antibody ProductCloneApplicationSource
PE anti-mouse IL17FeBio18F10Flow cytometry
Human IL17F AntibodyMAB13352ELISA, Western blot
IL17F antibody (ab168194)PolyclonalIHC, Western blot

Dual IL17A/F Neutralization

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 .

TrialOutcomeSource
Phase II Psoriasis Study80% ACR20 response vs. 16.7% placebo (p < 0.001)
Collagen-Induced ArthritisCombined IL17A/F blockade suppressed IL-6/G-CSF levels

Inflammatory Pathways

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 .

Infection Models

IL17F-deficient mice exhibit impaired defense against oropharyngeal candidiasis, highlighting its role in antifungal immunity .

Product Specs

Buffer
PBS with 0.02% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze/thaw cycles.
Lead Time
Typically, we can ship your order within 1-3 business days after receiving it. Delivery times may vary based on shipping method and location. For specific delivery timelines, please consult your local distributor.
Synonyms
CANDF6 antibody; Cytokine ML 1 antibody; Cytokine ML-1 antibody; IL 17F antibody; IL 24 antibody; IL-17F antibody; IL-24 antibody; Il17f antibody; IL17F_HUMAN antibody; Interleukin 17F antibody; Interleukin 24 antibody; Interleukin-17F antibody; Interleukin-24 antibody; ML 1 antibody; ML1 antibody; Mutant IL 17F antibody; OTTHUMP00000016602 antibody
Target Names
Uniprot No.

Target Background

Function
Interleukin-17F (IL-17F) is a crucial effector cytokine involved in both innate and adaptive immune responses. It plays a vital role in antimicrobial host defense and maintaining tissue integrity. IL-17F, along with IL-17A, signals through the IL-17RA-IL-17RC heterodimeric receptor complex. This interaction triggers the binding of IL-17RA and IL-17RC chains to the TRAF3IP2 adapter protein through SEFIR domains. This leads to the activation of downstream signaling pathways, including the TRAF6-mediated activation of NF-κB and MAPkinase pathways. Ultimately, these signaling cascades result in the transcription of various genes encoding cytokines, chemokines, antimicrobial peptides, and matrix metalloproteinases, which contribute to immune inflammation. IL-17F primarily contributes to host defense against extracellular bacteria and fungi by inducing neutrophilic inflammation. As a hallmark effector cytokine of T-helper 17 cells (Th17), IL-17F primarily induces neutrophil activation and recruitment at infection and inflammatory sites. IL-17F stimulates the production of antimicrobial beta-defensins DEFB1, DEFB103A, and DEFB104A by mucosal epithelial cells, limiting the entry of microbes through epithelial barriers. IL-17F homodimers can signal via the IL-17RC homodimeric receptor complex, leading to the activation of the TRAF6 and NF-κB signaling pathway. Through IL-17RC, IL-17F induces the transcriptional activation of IL-33, a potent cytokine that stimulates group 2 innate lymphoid cells and adaptive T-helper 2 cells involved in pulmonary allergic responses to fungi. IL-17F, likely via IL-17RC, promotes sympathetic innervation of peripheral organs by coordinating communication between gamma-delta T cells and parenchymal cells. IL-17F also stimulates sympathetic innervation of thermogenic adipose tissue by driving TGFB1 expression. Furthermore, IL-17F plays a role in regulating the composition of intestinal microbiota and immune tolerance by inducing antimicrobial proteins that specifically control the growth of commensal Firmicutes and Bacteroidetes.
Gene References Into Functions
  1. Research indicates that IL-17F genetic polymorphism is not associated with the development of rheumatic heart disease in the South Indian population. PMID: 29985710
  2. The expression of IL-6 gene and protein was significantly induced by IL-17F. IL-17F activates TAK1 and NF-κB in airway smooth muscle cells. PMID: 28474507
  3. IL-17FA7488G polymorphism was not significantly associated with colorectal cancer risk. PMID: 29970680
  4. A dietary pattern reflecting inflammation was significantly associated with colorectal cancer risk, and this association could be modified based on the IL-17F rs763780 genotype and anatomic site. PMID: 29874787
  5. Malignant T cells activate endothelial cells via IL-17F. PMID: 28731459
  6. The AA genotype on the 7489A/G single nucleotide polymorphism of IL-17F and the A allele might be associated with a lower risk of acute rejection and better graft survival in kidney transplant recipients. PMID: 29407292
  7. Findings suggest that IL-17F rs1889570 gene polymorphisms are significantly associated with susceptibility to severe EV71 infection in Chinese Han children. PMID: 29549443
  8. P-TEFb is involved in IL-17F-induced IL-8 expression, and steroids diminish it via the inhibition of CDK9 phosphorylation. PMID: 29649811
  9. The IL-17F (+7488A/G) genotype revealed a significantly increased risk of accelerated silicosis. The IL-17F (+7488 G) allele was associated with an increased risk of accelerated silicosis. PMID: 28481151
  10. Ultrasensitive methods were developed for measuring IL-17A and IL-17F in human serum samples. Serum from psoriasis patients had higher and a broader range of concentrations of both IL-17 proteins compared to healthy volunteers. PMID: 28534291
  11. SNPs of rs3819024 in IL-17A and rs763780 in IL-17F were weakly related to a prognosis of tuberculosis. PMID: 27339100
  12. IL17F (rs2397084) and IL10 (rs1800871) genes are associated with functional dyspepsia. PMID: 28965252
  13. A mutation in IL-17F is associated with susceptibility to recurrent aphthous stomatitis. PMID: 29458167
  14. The G allele at rs763780 (IL-17F) was significantly associated with Takayasu Arteritis in the Asian Indian population. PMID: 28438554
  15. Levels of mRNA IL-17F and IL17F might be useful parameters for the diagnosis of atopic asthma patients. PMID: 28606156
  16. IL-17F rs763780 polymorphisms may be associated with the development of primary immune thrombocytopenia in a Chinese Han population. PMID: 26620416
  17. In this study, IL-17F was demonstrated to have functions comparable to IL-17A in human keratinocytes. PMID: 27576147
  18. Serum IL-17F predicted increased knee bone marrow lesion scores in females only among patients with knee osteoarthritis. PMID: 27836676
  19. IL-17A and IL-17F polymorphisms therefore have the potential to act as predictive biomarkers for cervical cancer risk. PMID: 28621613
  20. The results suggest the possible involvement of the polymorphisms of IL17A G197A (rs2275913) and IL17F T7488C (rs763780) in the susceptibility to chronic Chagas disease and in the development and progression of cardiomyopathy. PMID: 28470012
  21. A meta-analysis of the function of IL-17A rs2275913 and IL-17F rs763780 genetic variations in inflammatory diseases risk indicated that these two genetic variations were risk factors for inflammatory diseases, particularly rheumatoid arthritis. PMID: 28186427
  22. This meta-analysis involving seven articles with 697 patients diagnosed with chronic periodontitis, 188 patients diagnosed with aggressive periodontitis, and 655 control patients (a total of 1540 participants) showed a non-significant association between the rs2275913 polymorphism in the IL-17A gene and the rs763780 polymorphism in the IL-17F gene with the risk of chronic periodontitis or aggressive periodontitis. PMID: 29027636
  23. The study provides evidence that functional IL-23R rs1884444 G/T and IL-17F rs763780 A/G polymorphisms may be a new genetic susceptibility factor to systemic lupus erythematosus, particularly in the Polish population. PMID: 27320770
  24. IL23R rs10889677 and IL17A rs2275913 were not associated with the susceptibility to Necrotizing enterocolitis (NEC). In conclusion, data suggest that a variant of IL17F (rs763780) may contribute to the development of NEC. PMID: 28224332
  25. The results confirmed IL17A and IL17F as potential candidate genes involved in rheumatoid arthritis. They play pivotal roles in the susceptibility and in clinical features of rheumatoid arthritis. Responses to rheumatoid arthritis treatments are differently conditioned by polymorphisms in IL17A and IL17F genes. PMID: 28143790
  26. IL17A and IL17F gene polymorphism are not significant factors associated with susceptibility and some clinical parameters of rheumatoid arthritis in a Polish population. PMID: 27169372
  27. The aim of this study was to investigate the associations between the 7383A/G and 7488A/G polymorphisms of the interleukin (IL)-17F gene with disease activity and clinical outcomes in Turkish patients with ankylosing spondylitis. PMID: 27155445
  28. Using an in-vitro migration assay, B cells were shown to migrate towards both IL-17A and IL-17F. These observations indicate a direct chemotactic effect of IL-17 cytokines on primary peripheral blood B cells, with a higher effect being on asthmatic B cells. PMID: 25494178
  29. IL-17-related cytokine expression was amplified in bronchial/nasal mucosa of neutrophilic asthma prone to exacerbation, suggesting a pathogenic role of IL-17F in frequent exacerbators. PMID: 27931975
  30. IL-17A (-197G/A) and IL-17F (7488T/C) SNPs were not associated with susceptibility to rheumatoid arthritis or secondary Sjogrens syndrome (sSS, p > 0.05 for both SNPs). Additionally, they did not influence RA activity or clinical markers of SS. PMID: 26232893
  31. It can be stated that the IL17A and IL17F polymorphisms are not markers of susceptibility to psoriasis. However, the IL17F polymorphism may affect the response to NB-UVB therapy. PMID: 27591988
  32. Elevated autoantibodies against IL-17F correlate with disease activity in patients with early rheumatoid arthritis. PMID: 26087054
  33. The study provides evidence that polymorphisms of both IL-17A and IL-17F may increase lung cancer risk in the Chinese population. PMID: 26073462
  34. This study shows that Korean patients with psoriasis show a strong association for IL17F single nucleotide polymorphism. PMID: 27774581
  35. The association of three polymorphism loci (rs2275913, 197 G/A; rs3748067, 383 A/G; and rs763780, 7488 T/C) of IL-17A and IL-17F with laryngeal cancer was analyzed. Allele and genotype frequencies of IL-17A rs2275913 were significantly different between patients and controls, with rs2275913 (197 G/A) AA and GA+AA genotypes compared to the GG genotype significantly higher in patients. PMID: 28362993
  36. These findings identify a novel biological function for IL-17A/F as an indirect angiogenic agent. PMID: 27594509
  37. IL-17F was correlated with increased autoantibody levels and disease activity in primary Sjogren's syndrome and is more clinically relevant than IL-17A. PMID: 28210632
  38. No relationship was found between IL17F rs763780 and rs9463772 polymorphisms and Henoch-Schonlein purpura susceptibility. PMID: 27021337
  39. Single nucleotide polymorphisms in IL-17F A7488G but not IL-17A are associated with the development of chronic immune thrombocytopenia in China. PMID: 27312555
  40. A comprehensive meta-analysis of the role of IL-17A rs2275913 and IL-17F rs763780 polymorphisms in cancer risk demonstrated that these two polymorphisms significantly increase the risk of developing cancer, particularly gastric cancer. PMID: 26843459
  41. Levels of IL-17FF were significantly higher in rheumatoid arthritis sera and showed a trend of increase in relapsing remitting multiple sclerosis, compared with normal healthy subjects. PMID: 27620302
  42. Polymorphism of IL-17 rs3748067 and rs763780 is closely associated with gastric cancer development. Polymorphism of IL-17 rs2275913 and rs4711998 may be correlated with the risk for gastric cancer. PMID: 27097946
  43. ELISA analysis verified high levels of Th17-associated proinflammatory cytokines such as interleukin-17A/F, interleukin-6, and interleukin-23 and low levels of inflammatory inhibitory factors including interleukin-10 and transforming growth factor-beta in primary immune thrombocytopenia patients compared with normal controls. PMID: 26484642
  44. The GGAGAA combined genotype and the GGA haplotype of IL-17A rs2275913, IL-17F rs763780, and rs2397084 can be considered risk factors for the development of systemic lupus erythematosus in Egyptian children. PMID: 26515887
  45. The GA genotype of the rs11465553 IL17F gene polymorphism may be associated with a significantly higher risk of graft function loss and return to dialysis after kidney transplantation. PMID: 26447633
  46. No associations were found between rs8193036, rs2275913, and rs3748067 in IL-17A and rs763780 in IL-17F SNPs and myasthenia gravis in Chinese patients. PMID: 26337284
  47. No evidence of association was observed between rs3748067, rs3819025, rs763780, rs9382084, and rs1266828 polymorphisms and the risk of cervical cancer. PMID: 26505366
  48. A higher expression level was observed in chronic lymphocytic leukemia patients. PMID: 26478573
  49. In vitro, CSE stimulation significantly increased IL-17F and IL-17R in 16HBE (2.5%) and A549 (5%), while IL-17A and IL-17F in PBMC (10%). IL-17A and CSE stimulation, rather than CSE or rhIL-17A alone, increased proliferation in 16HBE and apoptosis in A549. PMID: 26198032
  50. A G/G genotype of rs766748 in IL-17F, and a C/C or C/A genotype of rs1883136 in TRAF3IP2. PMID: 26558270

Show More

Hide All

Database Links

HGNC: 16404

OMIM: 606496

KEGG: hsa:112744

STRING: 9606.ENSP00000337432

UniGene: Hs.272295

Involvement In Disease
Candidiasis, familial, 6 (CANDF6)
Protein Families
IL-17 family
Subcellular Location
Secreted.
Tissue Specificity
Expressed in T-helper 1 and T-helper 2 cells, basophils and mast cells.

Q&A

What is IL-17F and how does it differ from other IL-17 family cytokines?

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) .

How should researchers design experiments to validate IL-17F antibody specificity?

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.

What are the recommended protocols for IL-17F detection in biological samples?

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

  • Measure optical density at 450 nm

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

How can researchers distinguish between IL-17F homodimers and IL-17A/F heterodimers in experimental systems?

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.

What methodological considerations are important when comparing IL-17A versus IL-17F neutralization in disease models?

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

How does affinity maturation impact IL-17F antibody performance in research applications?

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

What are the key considerations for developing therapeutic antibodies targeting both IL-17A and IL-17F?

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 .

What factors influence IL-17F antibody performance in intracellular staining experiments?

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

How can researchers optimize IL-17F neutralization experiments to study inflammatory pathways?

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

What are the optimal strategies for evaluating IL-17F antibody efficacy in animal models of inflammation?

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

How should researchers interpret contradictory findings between IL-17F neutralization studies?

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 .

What quantitative approaches best measure IL-17F antibody efficacy in complex biological systems?

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)

  • Neutralization efficacy at different IL-17F:IL-17A ratios

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 .

What are emerging techniques for studying IL-17F biology with enhanced precision?

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.

How might understanding of IL-17F biology inform development of next-generation therapeutic antibodies?

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.

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 2025 TheBiotek. All Rights Reserved.