Recombinant Human Interleukin-17A (IL17A) (Active)

Shipped with Ice Packs
In Stock

Description

Production Systems and Purification

Recombinant IL-17A is synthesized using multiple expression platforms:

Expression HostGlycosylation StatusPurification MethodActivity Validation
Escherichia coliNon-glycosylatedAffinity chromatography IL-6 induction in 3T3 fibroblasts
CHO/HEK293 CellsGlycosylatedProprietary chromatographic Neutrophil recruitment assays

Key Notes:

  • E. coli-derived IL-17A lacks glycosylation but retains bioactivity .

  • Mammalian cell systems (e.g., CHO, HEK293) produce glycosylated forms, mimicking native protein structure .

Biological Activity and Functional Assays

Recombinant IL-17A is validated through its ability to:

  • Induce IL-6, G-CSF, and CXCL chemokines in fibroblasts, epithelial cells, and synovial cells .

  • Enhance neutrophil recruitment via γδ T-cell activation .

  • Synergize with TNF-α or IL-22 to amplify inflammatory responses .

Activity Metrics:

Assay TypeED₅₀ RangeCell Line UsedSource
IL-6 secretion0.5–6 ng/mLMouse 3T3 fibroblasts
Neutrophil chemotaxis1–10 ng/mLPrimary human neutrophils
Antimicrobial peptide induction5–20 ng/mLKeratinocytes, epithelial cells

Host Defense and Infection

  • Bacterial Clearance: Enhances neutrophil-mediated eradication of Klebsiella pneumoniae, Escherichia coli, and Staphylococcus aureus in sepsis models .

  • Antiviral Activity: Promotes CXCL1/2 production to limit viral replication in mucosal tissues .

Autoimmune and Inflammatory Diseases

  • Pathogenic Role: Elevated levels correlate with rheumatoid arthritis, psoriasis, and multiple sclerosis .

  • Therapeutic Targeting: Neutralizing IL-17A antibodies (e.g., secukinumab) are FDA-approved for psoriasis .

Tissue Repair

  • Epithelial Regeneration: Stimulates keratinocyte proliferation and tight junction formation in gut and skin injury models .

  • Metabolic Regulation: Modulates CPT1A and itaconate pathways to balance inflammation and repair .

Key Research Findings

  1. Dual Role in Sepsis: IL-17A deficiency increases mortality in murine sepsis models by impairing neutrophil recruitment . Conversely, excessive IL-17A exacerbates organ damage via TNF-α synergy .

  2. Autoimmunity Mechanisms: Drives autoreactive germinal center formation in lupus models by enhancing plasma cell survival via Bcl-xL stabilization .

  3. Metabolic Cross-Talk: Itaconate derivatives inhibit IL-17A-induced IκBζ activation, suggesting therapeutic potential for inflammatory diseases .

Product Specs

Buffer
Lyophilized from a 0.2 µm filtered PBS, pH 7.4.
Form
Liquid or Lyophilized powder
Lead Time
Generally, we can ship the products within 1-3 working days after receiving your order. Delivery time may vary depending on the purchasing method or location. For specific delivery time, please consult your local distributors.
Note: All of our proteins are shipped with standard blue ice packs. If you require dry ice shipping, please inform us in advance as additional fees will apply.
Shelf Life
Shelf life is influenced by various factors such as storage conditions, buffer ingredients, temperature, and the inherent stability of the protein.
Generally, the shelf life of the liquid form is 6 months at -20°C/-80°C. The lyophilized form has a shelf life of 12 months at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is recommended for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
C-terminal 6xHis-tagged
Synonyms
CTLA 8; CTLA-8; CTLA8; cytotoxic T lymphocyte associated antigen 8; Cytotoxic T lymphocyte associated protein 8; Cytotoxic T lymphocyte associated serine esterase 8; Cytotoxic T-lymphocyte-associated antigen 8; IL 17; IL 17A; IL-17; IL-17A; IL17; IL17_HUMAN; Il17a; Interleukin 17 (cytotoxic T lymphocyte associated serine esterase 8); interleukin 17A; Interleukin-17A; interleukin17; Interleukin17A; OTTHUMP00000016597; OTTMUSP00000046003
Datasheet & Coa
Please contact us to get it.
Expression Region
24-155aa
Mol. Weight
15.9 kDa
Protein Length
Full Length of Mature Protein
Purity
Greater than 95% as determined by SDS-PAGE.
Research Area
Immunology
Source
Mammalian cell
Species
Homo sapiens (Human)
Target Names
Uniprot No.

Target Background

Function
Interleukin-17A (IL-17A) is an effector cytokine of both innate and adaptive immune systems. It plays a crucial role in antimicrobial host defense and maintaining tissue integrity. IL-17A signals through the IL17RA-IL17RC heterodimeric receptor complex, triggering homotypic interaction of IL17RA and IL17RC chains with the TRAF3IP2 adapter. This interaction leads to downstream TRAF6-mediated activation of the NF-κB and MAPkinase pathways, ultimately resulting in transcriptional activation of cytokines, chemokines, antimicrobial peptides, and matrix metalloproteinases. These processes contribute to potent immune inflammation.

IL-17A is a key player in connecting T cell-mediated adaptive immunity and acute inflammatory response. It acts as a signature effector cytokine of T-helper 17 cells (Th17), primarily inducing neutrophil activation and recruitment at infection and inflammatory sites. In airway epithelium, it mediates neutrophil chemotaxis through induction of CXCL1 and CXCL5 chemokines. In secondary lymphoid organs, IL-17A contributes to germinal center formation by regulating the chemotactic response of B cells to CXCL12 and CXCL13, enhancing B cell retention within the germinal centers, B cell somatic hypermutation rate, and selection towards plasma cells.

IL-17A is also an effector cytokine of a subset of γδ T cells that functions as part of an inflammatory circuit downstream of IL1B, TLR2, and IL23A-IL12B, promoting neutrophil recruitment for efficient bacterial clearance. It serves as an effector cytokine of innate immune cells including invariant natural killer cell (iNKT) and group 3 innate lymphoid cells, mediating initial neutrophilic inflammation. IL-17A is involved in maintaining the integrity of epithelial barriers during homeostasis and pathogen infection. Upon acute injury, it plays a direct role in epithelial barrier formation by regulating OCLN localization and tight junction biogenesis.

As part of the mucosal immune response induced by commensal bacteria, IL-17A enhances the host's ability to resist pathogenic bacterial and fungal infections by promoting neutrophil recruitment and antimicrobial peptide release. In synergy with IL17F, it mediates the production of antimicrobial β-defensins DEFB1, DEFB103A, and DEFB104A by mucosal epithelial cells, limiting the entry of microbes through the epithelial barriers. IL-17A is also involved in antiviral host defense through various mechanisms. It enhances immunity against West Nile virus by promoting T cell cytotoxicity. IL-17A may play a beneficial role in influenza A virus (H5N1) infection by enhancing B cell recruitment and immune response in the lung. It contributes to influenza A virus (H1N1) clearance by driving the differentiation of B-1a B cells, providing for production of virus-specific IgM antibodies at the first line of host defense.
Gene References Into Functions
  1. that proliferation potential-related protein promotes esophageal cancer cell proliferation and migration, and suppresses apoptosis by mediating the expression of p53 and IL-17 PMID: 30223275
  2. The pooled estimate revealed an association between IL-17A rs2275913 polymorphism and the risk of gastric cancer (GC) under all genetic models (A vs. G, OR 1.187, 95% CI 1.086-1.297, P < 0.001; GA vs. GG, OR 1.108, 95% CI 1.008-1.218, P = 0.033; AA vs. GG, OR 1.484, 95% CI 1.236-1.781, P < 0.001), while no evidence of association was found with IL-17A rs3748067 or IL-17F rs763780 polymorphisms. PMID: 29860554
  3. Over-expression of IL-17 and IL-27 are involved in the pathogenesis of liver damage in children with human cytomegalovrius infection. PMID: 30022757
  4. IL23 and IL17 have roles in the pathogenesis of Tunisian pemphigus foliaceus PMID: 30116153
  5. our findings supported the association between IL-17 SNPs and the risk of asthma in Chinese Han population from central China. GA genotype of rs3748067 and the C carries (CT+CC) of rs763780 were associated with a higher risk of asthma PMID: 30036556
  6. IL17A and HPSE may promote tumor angiogenesis and cell proliferation and invasion in cervical cancer, possibly via the NF-κB signaling pathway. PMID: 30066843
  7. the results suggest that IL17A (rs2275913) polymorphism is associated with the development of rheumatic heart disease in South Indian population PMID: 29985710
  8. This study demonstrated the alteration of IL-17 levels in aseptic non-vasculitic cerebral sinovenous thrombosis PMID: 30246697
  9. In this Brazilian population, TNF and IL17 gene polymorphisms responsible for the expression of important inflammatory cytokines were associated with overall spondyloarthritis and, specifically, with ankylosing spondylitis and psoriatic arthritis, regardless of gender and HLA-B27 PMID: 29849482
  10. The single nucleotide polymorphism rs2275913 in the IL-17A gene is associated with susceptibility to viral myocarditis. PMID: 29530464
  11. IL-17A197AA polymorphism is associated with the risk of colorectal cancer. PMID: 29970680
  12. our findings demonstrated that the AA genotype from the IL-17A rs2275913 SNP is positively associated with protection to active tuberculosis but related to higher disease severity in the Argentinean population. PMID: 28098168
  13. Our results from experiments suggest that the effects of IL-17 mediate activation of STAT3 signaling in breast cancer cells. Taken together, our study shows that myeloid-derived suppressor cells can be a new type of prognostic marker in breast cancer patients. Targeting IL-17/Stat3 signaling may be a promising strategy for BC treatment. PMID: 29655056
  14. This study draws two main conclusions: 1) The presence of IL-17 polymorphism rs2275913 is closely related to a more severe form of the disease and as a result, a higher number of disease-modifying anti rheumatic drugs required to control it, 2) The presence of IL-17 polymorphism rs2275913 may confer a risk of developing rheumatoid arthritis in Mexican carriers PMID: 28379210
  15. Polymorphisms of IL-17 are associated with host susceptibility to some bacterial pathogen. [review] PMID: 29345518
  16. secreted IL-17A is not responsible for the second hit in acute pancreatitis PMID: 29422392
  17. In carriage, an increased IL-17 and IFN-gamma levels were observed following stimulation with S. aureus strains. PMID: 29311230
  18. IL17A G197A is associated with a higher susceptibility of developing OLP and these patients seem to present a considerable increase in IL17A serum levels. PMID: 28741807
  19. These findings highlight a regulatory pathway of Tiam1/Rac1 in Th17 cells and suggest that it may be a therapeutic target in multiple sclerosis. PMID: 27725632
  20. Interleukin 17A (IL-17[G197G]) was associated with preterm birth (PTB), and the PTB group had lower IL-17A expression compared to the full-term group . PMID: 29431293
  21. In studies of mouse and human pancreatic tumors and precursors, we found that immune cell-derived IL17 regulated development of tuft cells and stem cell features of pancreatic cancer cells via increased expression of DCLK1, POU2F3, ALDH1A1, and IL17RC. PMID: 29604293
  22. Expression of miR-135a in the cancer cells isolated from nasopharyngeal tumors was significantly lower than that in NP69 cells, and suppression of IL-17 by miR-135a mimic resulted in significant inhibition of NPC cell proliferation. PMID: 29734196
  23. The expression of IL-17 and IL-12 in patients with lupus miliaris disseminatus faciei is reported in patients and healthy controls. PMID: 27515793
  24. these findings suggest that the variants +2199 A/C IL-23R and -197 G/A IL-17A could contribute to rheumatoid arthritis development in the studied population PMID: 28547498
  25. IL-17A rs2275913 polymorphism did not seem to influence RA susceptibility in Tunisian population. PMID: 29584788
  26. results suggest that IL-17A stimulated keratinocytes activated PI3K/AKT/mTOR signaling and inhibited autophagy by simultaneously inhibiting autophagosome formation and enhancing autophagic flux. PMID: 29432814
  27. In the present review, we have discussed the cellular sources, modes of action and regulation of IL-17 and IL-33 in the context of hypersensitive diseases [Review] PMID: 29153708
  28. Findings identify interleukin-17A as a potential mediator of neuroanatomical remodeling of the gut innervation during inflammatory bowel diseases. PMID: 28560787
  29. This review discusses recent discoveries related to the pro-inflammatory cytokine IL17A, and its potential role in the pathogenesis of COPD. We propose that an intervention strategy targeting IL-17 signaling offers an exciting opportunity to mitigate inflammatory processes, and prevent the progression of tissue pathologies associated with COPD [Review] PMID: 28438639
  30. rs2275913 gene polymorphism associated with a risk of Bacillus-Calmette-Guerin osteitis after vaccination PMID: 28731539
  31. Study shows that higher baseline levels of Interleukin 17 are selectively associated with greater symptomatic reduction in depressed patients treated with bupropion-SSRI combination. PMID: 28698115
  32. IL-23/IL-17 axis and biochemical markers in the pathogenesis of Type 2 Diabetes PMID: 28757426
  33. Higher levels of TGF-beta mRNA were observed in biopsies taken from healthy controls, and the IL-23 mRNA levels were significantly increased in the peri-implantitis group (P < 0.0001). No differences in IL-17 mRNA levels were observed between the two groups (P > 0.05). PMID: 27062688
  34. Studied interleukin-17 (IL-17) expression levels in blood and skin of atopic dermatitis (AD) patients and controls. PMID: 28279075
  35. semen IL-17 and IL-18 levels in diabetes mellitus males were significantly higher than those in normal males and were positively correlated with blood glucose level and sperm DNA fragmentation index PMID: 28858634
  36. clinical significance of IL-17 and IL-23 in the pathogenesis of different types of gastric neoplasms in humans, is reported. PMID: 27869179
  37. The IL-17A (-832A/G) polymorphism was not associated with accelerated silicosis. PMID: 28481151
  38. IL-17A was significantly upregulated in patients with the uncontrolled and refractory status. Therefore, IL-17A may play important roles in asthmatic exacerbation, and its high level, in combination with upregulated Th2 and other cytokines, may indicate the refractory endotype of asthma. PMID: 28840844
  39. results suggest that IL17A participates in the immune response of the human host against M. tuberculosis through the activation of the autophagy process in correlation with the severity of the disease PMID: 28581888
  40. developed ultrasensitive methods for measuring IL-17A and IL-17F in human serum samples and found that serum from psoriasis patients had higher and a broader range of concentrations of both IL-17 proteins compared to healthy volunteers PMID: 28534291
  41. SNPs of rs3819024 in IL-17A and rs763780 in IL-17F were weakly related to a prognosis of tuberculosis. PMID: 27339100
  42. this study shows that aberrant NKG2D expression with IL-17 production of CD4+ T subsets in patients with type 2 diabetes PMID: 27168217
  43. Luciferase assay using the 5'-UTR of the IL-17 F gene revealed transcriptional regulation. Induced IL-17 F production was further confirmed at the protein level by ELISA. Smad1/5/8 inhibitor pretreatment decreased IL-17 F expression levels in the cells. PMID: 28812969
  44. Study shows increased IL-17A expression in early tendinopathy and proposes IL-17A as an inflammatory regulator in tendon remodeling. PMID: 27263531
  45. Several studies identified IL-17A as a pro-inflammatory player in atherosclerosis while its expression was associated with increased inflammation and plaque vulnerability in human atherosclerotic lesions. Moreover, IL-17A induced a pro-inflammatory, pro-thrombotic, plaque-destabilizing, and cell-attracting response of the inflammatory milieu of human plaque tissue samples. [review] PMID: 28034277
  46. asymmetric cell divisions in psoriasis are IL17A-dependent. PMID: 28600817
  47. Serum IL-23 and IL-17 levels were elevated in patients with aneurysmal subarachnoid hemorrhage (aSAH) showing upregulation of IL-23/IL-17 inflammatory axis after aSAH. Serum IL-23 and IL-17 showed differential correlations with post hemorrhagic complications and no correlation with clinical outcome. PMID: 28609751
  48. Data showed that IL-17A levels were sustained in respiratory samples from cystic fibrosis patients infected by P. aeruginosa. IL-17 mediated-immunity plays a double-edged found during chronic airways infection: in one hand, it contributes to the control of P. aeruginosa burden, modulating host resistance, while, on the other, it alters host tolerance, propagating exacerbated pulmonary neutrophilia and tissue remodeling. PMID: 27189736
  49. gamma delta T cells, rather than Th17cells, are the principal producers of IL-17 in acute gouty arthritis patients during the acute gout flares. PMID: 29476737
  50. high expression of both IL17A and IL32 leads to enhancement of T cell responses in breast tumors PMID: 28470472

Show More

Hide All

Database Links

HGNC: 5981

OMIM: 603149

KEGG: hsa:3605

STRING: 9606.ENSP00000344192

UniGene: Hs.41724

Protein Families
IL-17 family
Subcellular Location
Secreted.
Tissue Specificity
Expressed in memory Th17 cells (at protein level).

Q&A

What is the molecular structure and basic characteristics of recombinant human IL-17A?

Recombinant human IL-17A is a disulfide-linked homodimeric secreted glycoprotein comprising 155 amino acid residues. The mature human IL-17A protein sequence typically spans from Gly24/Ile20 to Ala155, depending on the specific recombinant product . Under non-reducing conditions, IL-17A appears as a 28-38 kDa protein due to glycosylation and dimerization . When expressed in human cell lines, IL-17A exhibits proper glycosylation patterns with N-linked oligosaccharides that contribute to its stability and biological activity . Under reducing conditions, IL-17A can migrate as two bands at approximately 16 and 22 kDa on SDS-PAGE, reflecting different post-translational modifications . The protein has a predicted isoelectric point (pI) of approximately 8.62, indicating its slightly basic nature .

IL-17A adopts a conserved cystine knot fold shared among the IL-17 family cytokines. Besides forming homodimers, IL-17A can also form heterodimers with IL-17F, which may exhibit slightly different biological activities compared to IL-17A homodimers .

How does the expression system affect recombinant human IL-17A properties?

The expression system significantly impacts the structural and functional properties of recombinant human IL-17A. Two primary expression systems are commonly used:

Expression SystemAdvantagesLimitationsMolecular Characteristics
Human Cell Lines (e.g., HEK293)Native glycosylation patterns, Proper folding and disulfide bond formation, Higher biological activityHigher production costs, Potentially lower yield28-38 kDa (non-reduced), N-glycosylated
E. coliCost-effective, Higher yield, Simpler purificationLacks glycosylation, May require refolding, Potential endotoxin contaminationLower molecular weight, No glycosylation, Requires additional endotoxin removal

Human cell-expressed IL-17A typically exhibits ED50 values of 0.12-1.2 ng/mL in IL-6 induction assays, while E. coli-derived protein shows ED50 values of 0.4-4 ng/mL, indicating potential differences in specific activity . For experiments requiring physiologically relevant post-translational modifications, human cell-expressed IL-17A is preferred, while E. coli-derived protein may be sufficient for applications where glycosylation is not critical .

What are the recommended reconstitution and storage conditions for maintaining IL-17A activity?

Proper reconstitution and storage are crucial for maintaining the biological activity of recombinant IL-17A. The following methodological approach is recommended:

Reconstitution Protocol:

  • For carrier-containing formulations: Reconstitute at 100 μg/mL in PBS containing at least 0.1% human or bovine serum albumin .

  • For carrier-free formulations: Reconstitute at 100 μg/mL in sterile PBS without additives .

  • Allow the lyophilized protein to reach room temperature before reconstitution.

  • Gently swirl until completely dissolved; avoid vigorous vortexing that can cause protein denaturation.

Storage Recommendations:

  • Store lyophilized protein at 2-8°C before reconstitution .

  • For short-term use (≤1 month), store reconstituted protein at 4°C.

  • For long-term storage, prepare small aliquots and store at -18 to -20°C .

  • Use a manual defrost freezer and avoid repeated freeze-thaw cycles as they significantly reduce biological activity .

  • When preparing working solutions for experiments, dilute in buffers containing a carrier protein (0.1-0.5% BSA or HSA) to prevent adsorption to labware surfaces.

For HSA-containing formulations, the reconstituted solution typically contains 1% human serum albumin and 10% trehalose, which enhances stability during freeze-thaw cycles .

How can I design experiments to assess IL-17A signaling pathways in different cell types?

Experimental design for IL-17A signaling studies should consider cell type-specific responses and the temporal dynamics of downstream pathway activation. A comprehensive approach includes:

Cell Selection and Preparation:

  • Primary cells relevant to research question (e.g., aortic endothelial cells for vascular studies, fibroblasts for tissue remodeling) .

  • Culture cells to 70-80% confluence in appropriate serum-reduced medium for 12-24 hours before IL-17A treatment to reduce background signaling.

  • Include appropriate positive controls (e.g., TNF-α, IL-1β) and negative controls (vehicle only).

Concentration and Time-Course Optimization:

  • Perform dose-response experiments ranging from 0.1-100 ng/mL IL-17A.

  • For signaling studies, examine early timepoints (5, 15, 30, 60 minutes) for MAPK activation and later timepoints (1, 3, 6, 24 hours) for transcriptional responses .

Pathway Analysis Methods:

  • Western blotting for phosphorylated p38 MAPK, which is specifically activated by IL-17A in endothelial cells .

  • qRT-PCR to measure expression of IL-17A-responsive genes: IL-6, GM-CSF, CXCL1, and CXCL2 .

  • ELISA to quantify secreted inflammatory mediators.

  • Pathway inhibitors (e.g., p38 MAPK inhibitor) to confirm specific signaling mechanisms .

Functional Readouts:

  • Cell adhesion assays measuring monocyte adhesion to IL-17A-activated endothelial cells .

  • Intercellular adhesion molecule 1 (ICAM-1) expression by flow cytometry .

  • Transwell migration assays to assess chemotactic responses.

When investigating IL-17A signaling, it's important to address the potential heterodimeric interactions with IL-17F, as both IL-17 receptor A (IL-17RA) and IL-17 receptor C (IL-17RC) are required for responsiveness to IL-17A/F heterodimers .

What are the key methodological considerations for evaluating IL-17A bioactivity in experimental systems?

Accurate assessment of IL-17A bioactivity requires carefully standardized assays that reflect its physiological functions. Several methodological approaches are recommended:

Standard Bioactivity Assays:

  • IL-6 induction assay in NIH-3T3 mouse embryonic fibroblasts - a standard measure of IL-17A activity (typical ED50: 0.12-1.2 ng/mL for human cell-expressed; 0.4-4 ng/mL for E. coli-derived) .

  • IL-6 production in normal human dermal fibroblasts (NHDF) - measures physiologically relevant activity (typical ED50: 0.5-1.5 ng/mL) .

Critical Experimental Controls:

  • Heat-inactivated IL-17A (95°C for 5 minutes) as negative control.

  • Species-matched IL-17A for animal studies to account for species specificity.

  • Blocking antibodies against IL-17RA to confirm receptor specificity.

  • Inclusion of IL-17F and IL-17A/F heterodimers for comparative analyses.

Measuring Endothelial Cell Activation:

  • Flow cytometry for cell surface adhesion molecules (ICAM-1).

  • Monocyte adhesion assays under static or flow conditions.

  • Quantification of chemokines CXCL1 and CXCL2 by ELISA or multiplex assay .

Technical Considerations:

  • Use low-binding microcentrifuge tubes and pipette tips to prevent protein adsorption.

  • Prepare fresh working solutions for each experiment.

  • Include appropriate vehicle controls that match the IL-17A formulation buffer.

  • For long-term studies, perform stability assessment of the reconstituted protein at experimental conditions.

When comparing results across different recombinant IL-17A preparations, standardize activity units rather than absolute concentrations to account for batch-to-batch and source-to-source variations in specific activity .

How does IL-17A interact with other inflammatory mediators in experimental disease models?

IL-17A functions within a complex network of inflammatory mediators, requiring careful experimental design to elucidate its specific contributions. Key methodological approaches include:

Co-stimulation Experiments:

  • Combine IL-17A with TNF-α, IL-1β, or IFN-γ at submaximal concentrations to assess synergistic effects on target cell responses.

  • Compare sequential vs. simultaneous cytokine treatments to model priming effects.

  • Measure both transcriptional responses (mRNA levels) and protein outputs (secreted cytokines/chemokines).

In Vivo Models:

  • Using genetic approaches such as IL-17A knockout mice (e.g., ApoE−/−/IL-17A−/− double knockout) to study atherosclerosis progression .

  • Intravital microscopy to directly visualize leukocyte adhesion to endothelium in response to IL-17A .

  • Tissue-specific conditional knockout or overexpression systems to distinguish local vs. systemic IL-17A effects.

Mechanistic Pathway Analysis:

  • Specific inhibition of p38 MAPK pathway in endothelial cells to attenuate IL-17A-mediated activation, which regulates expression of proinflammatory cytokines, chemokines, and adhesion molecules .

  • Analysis of IL-17 receptor complex formation (IL-17RA-IL-17RC heterodimers) and TRAF3IP2 adapter recruitment in response to different inflammatory stimuli .

  • Investigation of transcriptional regulators downstream of IL-17A signaling.

Experimental Readouts:

  • Quantification of neutrophil chemotaxis and activation, a hallmark of IL-17A activity .

  • Assessment of antimicrobial peptide production in epithelial cells.

  • Measurement of tissue-specific inflammatory markers.

  • Evaluation of germinal center formation and B cell responses .

In cardiovascular disease models, IL-17A contributes to endothelial activation via transcriptional and post-translational mechanisms. Hyperlipidemic stress (e.g., oxidized LDL) up-regulates IL-17 receptors in aortic endothelial cells, enhancing their responsiveness to IL-17A and promoting leukocyte adhesion . The absence of IL-17A in mouse models reduces this endothelial activation and subsequent inflammatory responses.

What are common causes of variability in IL-17A activity assays and how can they be addressed?

Variability in IL-17A bioactivity assays can significantly impact experimental reproducibility. Researchers should consider these methodological solutions to common problems:

ProblemPossible CausesMitigation Strategies
Low or inconsistent activityProtein denaturation during reconstitutionReconstitute gently without vortexing; maintain recommended concentration (100 μg/mL)
Multiple freeze-thaw cyclesPrepare single-use aliquots upon reconstitution
Protein adsorption to labwareAdd carrier protein (0.1-0.5% BSA) to working solutions
Variable cell responsivenessStandardize cell passage number and density; synchronize cells by serum starvation
High backgroundSerum components interfering with assayReduce serum concentration during assay; use defined serum replacements
Endotoxin contaminationUse carrier-free preparations with certified low endotoxin levels (<1 EU/μg)
Cell stress from handlingMinimize trypsinization time; allow recovery period before experiments
Discrepancies between assay systemsDifferent receptor expression levelsValidate IL-17RA and IL-17RC expression in test cells
Species-specific differencesUse species-matched reagents; consider species cross-reactivity

When transitioning from in vitro to in vivo experiments, consider:

  • Differences in protein half-life and bioavailability in vivo.

  • Endogenous IL-17A/F background levels.

  • Compensatory mechanisms absent in simpler in vitro systems.

  • Timing of IL-17A administration relative to disease onset in experimental models.

For increased reproducibility across laboratories, standardize reporting of:

  • Specific recombinant IL-17A product and lot number.

  • Detailed reconstitution and storage protocol.

  • Cell types, passage numbers, and culture conditions.

  • Complete assay methodology including incubation times and detection systems.

How can I distinguish between IL-17A homodimer and IL-17A/F heterodimer effects in experimental systems?

Distinguishing the specific contributions of IL-17A homodimers versus IL-17A/F heterodimers requires specialized experimental approaches:

Protein Selection Strategies:

  • Use purified recombinant IL-17A homodimers and compare with equivalent molar concentrations of IL-17A/F heterodimers.

  • Include IL-17F homodimers as additional control.

  • Consider using site-directed mutants that preferentially form specific dimeric structures.

Receptor Targeting Approaches:

  • Use receptor-specific blocking antibodies:

    • Anti-IL-17RA blocks both IL-17A and IL-17A/F signaling.

    • Combined anti-IL-17RA and anti-IL-17RC completely inhibits both cytokines.

    • Selective IL-17RC blockade may differentially affect responses.

  • Employ siRNA knockdown of IL-17RA or IL-17RC to assess their relative contributions.

  • Use cell lines with defined receptor expression profiles.

Functional Discrimination:

  • Neutrophil migration assays - IL-17A induces neutrophil migration while IL-17F does not .

  • Dose-response analyses - IL-17A and IL-17A/F may show different potencies.

  • Kinetic studies - temporal differences in signaling activation.

  • Gene expression profiling to identify distinct transcriptional signatures.

Analytical Methods:

  • Western blotting under non-reducing conditions to visualize dimeric species.

  • Immunoprecipitation with dimer-specific antibodies if available.

  • Use of epitope-tagged recombinant proteins to track complex formation.

When interpreting results, consider that both IL-17RA and IL-17RC are required for responsiveness to IL-17A/F heterodimers, while IL-17A homodimers may signal through alternative receptor complexes including IL-17RA with either IL-17RC or IL-17RD .

What considerations are important when comparing different sources of recombinant IL-17A in research protocols?

Comparing results obtained with different recombinant IL-17A preparations requires careful consideration of several factors:

Source and Production Variables:

  • Expression system differences:

    • Human cell-expressed (HEK293) IL-17A shows proper glycosylation and typically exhibits higher specific activity (ED50: 0.12-1.2 ng/mL) than E. coli-derived protein (ED50: 0.4-4 ng/mL) .

    • E. coli-derived proteins lack glycosylation but may be suitable for many applications .

  • Sequence variations:

    • Some products include the signal peptide sequence while others start at mature protein sequence.

    • N-terminal methionine additions in E. coli-derived products .

    • Sequence boundaries vary slightly between products (Gly24-Ala155 vs. Ile20-Ala155) .

Formulation Differences:

  • Carrier protein presence/absence:

    • Carrier-containing formulations (with BSA) enhance stability but may interfere with some applications .

    • Carrier-free versions (CF) are preferred for applications where BSA could interfere .

  • Buffer composition differences between commercial sources.

  • Presence of stabilizers like trehalose in some formulations .

Standardization Approaches:

  • Normalize based on bioactivity rather than protein concentration:

    • Calculate EC50 values in standardized bioassay (e.g., IL-6 induction in NIH-3T3 cells).

    • Use biological activity units rather than absolute concentration.

  • Include internal standards across experiments.

  • Perform side-by-side comparisons when changing protein sources.

Application-Specific Considerations:

  • For in vivo studies, carrier-free preparations may be preferred to avoid immune responses to BSA.

  • For structural studies, glycosylation heterogeneity in human cell-expressed proteins may be problematic.

  • For functional assays measuring downstream signaling, human cell-expressed IL-17A may more accurately reflect physiological activity.

  • For ELISA standards, either formulation is typically suitable.

When transitioning between different IL-17A sources, validate the new product in your specific experimental system before collecting critical data. Document the specific product and lot number in publications to enhance reproducibility .

How does IL-17A contribute to tissue-specific inflammatory responses in different disease models?

IL-17A exhibits differential effects across tissues, contributing to disease pathogenesis through tissue-specific mechanisms:

Vascular System:

  • IL-17A activates aortic endothelial cells via the p38 MAPK pathway, promoting monocyte adhesion and potentially contributing to atherosclerosis .

  • Hyperlipidemic stress (oxidized LDL) up-regulates IL-17 receptors in aortic endothelial cells, enhancing their responsiveness to IL-17A .

  • In ApoE−/−/IL-17A−/− double knockout mice, leukocyte adhesion to endothelium is reduced, suggesting a direct role in vascular inflammation .

  • IL-17A induces expression of proinflammatory cytokines (IL-6, GM-CSF) and chemokines (CXCL1, CXCL2) in endothelial cells, creating a chemotactic gradient for neutrophil recruitment .

Mucosal Tissues:

  • IL-17A produces protective mucosal inflammation against microbial infection by inducing antimicrobial peptide production .

  • In intestinal inflammation, IL-17A can have dual roles:

    • Promoting neutrophil recruitment and antimicrobial defense.

    • Limiting chronic inflammation and protecting against colitis progression .

  • Experimental approaches should include tissue-specific knockout models and localized cytokine delivery to distinguish protective versus pathogenic roles.

Lymphoid Tissues:

  • IL-17A enhances germinal center formation by regulating B cell chemotaxis in response to CXCL12 and CXCL13 .

  • It increases B cell retention within germinal centers and enhances somatic hypermutation rates .

  • Research protocols should include B cell trafficking assays and analysis of affinity maturation in the presence of IL-17A.

Methodology for Tissue-Specific Studies:

  • Tissue-specific conditional knockout models.

  • Ex vivo tissue explant cultures with defined IL-17A exposure.

  • Tissue-specific reporter systems to track IL-17A-responsive cells.

  • Single-cell RNA sequencing to identify cell-specific responses within heterogeneous tissues.

When investigating IL-17A in tissue-specific inflammation, consider the temporal dynamics of acute versus chronic responses and the interaction with tissue resident cells versus recruited inflammatory cells .

What are the current methodological approaches for targeting IL-17A signaling in experimental disease models?

Research on IL-17A signaling inhibition employs various strategic approaches with distinct experimental considerations:

Direct IL-17A Neutralization:

  • Anti-IL-17A neutralizing antibodies:

    • Provide high specificity but variable tissue penetration.

    • Dose titration critical for partial versus complete inhibition.

    • Consider potential immunogenicity in long-term studies.

  • Soluble receptor constructs (IL-17RA-Fc):

    • Broader inhibition profile (blocks multiple IL-17 family members).

    • May better mimic therapeutic approaches being developed clinically.

Receptor Targeting Strategies:

  • Anti-IL-17RA or anti-IL-17RC blocking antibodies:

    • Allows differential blockade of receptor components.

    • IL-17RA blockade affects multiple IL-17 family members.

  • Genetic approaches:

    • Cell-type specific receptor knockout using Cre-lox systems.

    • Inducible knockout systems for temporal control.

    • CRISPR/Cas9 editing for precise receptor modifications.

Downstream Signaling Inhibition:

  • p38 MAPK inhibitors specifically attenuate IL-17A-mediated endothelial cell activation by:

    • Reducing expression of proinflammatory cytokines and chemokines.

    • Decreasing adhesion molecule expression (ICAM-1).

    • Limiting monocyte adhesion to activated endothelium .

  • Inhibitors of TRAF3IP2 adapter to disrupt IL-17R-mediated signaling .

  • NF-κB pathway inhibitors to block downstream transcriptional responses.

Experimental Design Considerations:

  • Preventive versus therapeutic intervention timing.

  • Local versus systemic administration.

  • Combination with other inflammatory pathway inhibitors.

  • Assessment of both intended target inhibition and off-target effects.

  • Verification of target engagement using phospho-specific antibodies for p38 MAPK or other pathway components .

For cardiovascular disease models, the p38 MAPK pathway represents a specific therapeutic target for IL-17A-mediated endothelial activation. Inhibition of this pathway ameliorates the expression of inflammatory cytokines, chemokines, and adhesion molecules, potentially reducing atherosclerotic progression .

How can transcriptomic and proteomic approaches enhance understanding of IL-17A signaling networks?

High-throughput molecular profiling technologies offer powerful tools for dissecting IL-17A signaling networks:

Transcriptomic Approaches:

  • RNA-seq time-course experiments:

    • Capture early (1-3 hours) and late (6-24 hours) transcriptional responses.

    • Identify primary versus secondary response genes.

    • Compare profiles from different cell types (endothelial cells, fibroblasts, epithelial cells).

  • Single-cell RNA-seq:

    • Resolve heterogeneous responses within cell populations.

    • Identify IL-17A-responsive cell subsets in complex tissues.

    • Map transcriptional trajectories during IL-17A stimulation.

  • Comparative analysis workflows:

    • IL-17A vs. IL-17F vs. IL-17A/F heterodimer responses.

    • IL-17A alone vs. IL-17A combined with TNF-α or IL-1β.

    • Wild-type vs. signaling-deficient cells (p38 MAPK inhibited) .

Proteomic Methods:

  • Phosphoproteomics to map IL-17A-induced signaling events:

    • Early phosphorylation events in p38 MAPK cascade .

    • Temporal dynamics of pathway activation.

    • Identification of novel signaling nodes.

  • Secretome analysis:

    • Quantify changes in secreted inflammatory mediators.

    • Identify novel IL-17A-regulated factors.

  • Proximity labeling approaches:

    • Map IL-17 receptor complex components.

    • Identify transient protein-protein interactions.

Integrated Multi-omics:

  • Combined transcriptome-proteome correlation analysis.

  • Network modeling of IL-17A signaling:

    • Identify critical nodes and potential feedback loops.

    • Map crosstalk with other inflammatory pathways.

    • Predict potential therapeutic targets.

  • Validation strategies for omics findings:

    • CRISPR screening of candidate regulators.

    • Targeted inhibition of newly identified pathways.

    • In vivo confirmation in disease models.

Computational Analysis:

  • Pathway enrichment analysis to identify IL-17A-regulated biological processes.

  • Transcription factor binding site analysis to map transcriptional regulators.

  • Interactome mapping to visualize protein interaction networks.

  • Cross-species conservation analysis to identify evolutionarily conserved IL-17A responses.

Advanced transcriptomic and proteomic approaches can reveal how IL-17A specifically activates the p38 MAPK pathway in endothelial cells, both transcriptionally and post-translationally, leading to vascular inflammation and potential atherosclerotic progression .

What are the emerging research directions in IL-17A biology and experimental applications?

Current research on IL-17A is expanding in several promising directions that require sophisticated experimental approaches:

Tissue Microenvironment Interactions:

  • Investigation of IL-17A's role in tissue-specific immune responses using organoid models and tissue-on-chip technologies.

  • Analysis of how tissue metabolic state influences IL-17A signaling, particularly in hyperlipidemic environments where IL-17 receptor expression is upregulated .

  • Development of spatial transcriptomics approaches to map IL-17A-responsive cellular niches within complex tissues.

Precision Targeting Strategies:

  • Development of heterodimer-specific inhibitors that selectively target IL-17A/A homodimers versus IL-17A/F heterodimers.

  • Cell type-selective delivery systems for IL-17A pathway modulators.

  • Temporal control of IL-17A signaling using optogenetic or chemically-inducible systems.

  • Selective inhibition of downstream pathways (e.g., p38 MAPK) in specific cell populations .

Systems Biology Approaches:

  • Multi-scale modeling of IL-17A signaling from molecular interactions to tissue-level responses.

  • Machine learning applications to predict IL-17A-dependent disease progression.

  • Network analysis to identify critical regulatory nodes in IL-17A signaling.

  • Quantitative systems pharmacology to optimize therapeutic targeting strategies.

Translational Applications:

  • Development of more physiologically relevant assays to predict in vivo IL-17A function.

  • Biomarker discovery for IL-17A pathway activation in clinical samples.

  • Patient-derived models to assess personalized responses to IL-17A inhibition.

  • Combinatorial targeting approaches addressing IL-17A along with synergistic pathways.

The burgeoning understanding of IL-17A's role in cardiovascular inflammation through specific activation of p38 MAPK in endothelial cells opens new avenues for targeting this pathway in atherosclerosis and related disorders. Future research will likely focus on integrating these molecular insights into therapeutic strategies that selectively modulate IL-17A signaling in disease-relevant tissues while preserving its protective functions in antimicrobial immunity .

How can researchers optimize experimental design when studying IL-17A in complex disease models?

Optimizing experimental design for IL-17A studies in complex disease models requires systematic consideration of multiple variables:

Model Selection and Refinement:

  • Choose animal models that recapitulate human disease-relevant IL-17A pathways:

    • ApoE−/−/IL-17A−/− double knockout mice for atherosclerosis studies .

    • Tissue-specific IL-17 receptor conditional knockout models.

    • Humanized mouse models expressing human IL-17 receptors.

  • Consider temporal aspects of disease progression:

    • Early inflammatory phase versus chronic disease.

    • Preventive versus therapeutic intervention timing.

  • Account for sex differences in IL-17A responses and disease susceptibility.

Intervention Design:

  • Dose optimization strategies:

    • Perform detailed dose-response studies (0.1-100 ng/mL range for in vitro, scaled appropriately for in vivo).

    • Consider pharmacokinetics and tissue distribution for in vivo studies.

  • Targeting approaches:

    • Compare direct IL-17A neutralization versus receptor blockade.

    • Evaluate pathway-specific inhibition (e.g., p38 MAPK inhibitors for vascular studies) .

    • Test combination approaches targeting multiple inflammatory mediators.

Readout Selection:

  • Multi-parameter assessment:

    • Molecular markers (phospho-p38 MAPK, cytokine/chemokine profiles) .

    • Cellular responses (adhesion molecule expression, leukocyte adhesion) .

    • Tissue pathology (inflammatory infiltration, tissue damage).

    • Functional outcomes (vascular function, tissue repair).

  • Temporal analysis:

    • Early signaling events (minutes to hours).

    • Intermediate transcriptional responses (hours).

    • Late functional consequences (days to weeks).

Statistical and Experimental Design Considerations:

  • Power analysis for appropriate sample sizing.

  • Include both positive controls (known IL-17A-dependent models) and negative controls.

  • Randomization and blinding procedures for in vivo studies.

  • Consider factorial experimental designs to assess multiple variables simultaneously.

  • Include genetic background controls for knockout models.

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.