IL-17A Recombinant Monoclonal Antibodies are engineered therapeutic agents designed to neutralize interleukin-17A (IL-17A), a proinflammatory cytokine critical in autoimmune diseases and inflammatory conditions. These antibodies are produced via recombinant DNA technology, ensuring high specificity and consistency. They have emerged as pivotal tools in both research and clinical settings, particularly for treating psoriasis, rheumatoid arthritis, and inflammatory bowel disease .
IL-17A antibodies enable precise analysis of cytokine function:
Recombinant antibodies are central to treating IL-17A-driven diseases:
Bimekizumab, a dual-specific antibody, shows enhanced efficacy by neutralizing both IL-17A and IL-17F:
Autoimmune Diseases: IL-17A promotes neutrophil recruitment and proinflammatory cytokine release (e.g., IL-6, COX-2) . Antibodies like ixekizumab disrupt this cascade, reducing disease severity .
Cancer: IL-17A exhibits dual roles—promoting tumor invasion via EMT (e.g., MTA1 induction) or inducing apoptosis in ovarian cancer via STAT3 inhibition . Recombinant antibodies are being explored to modulate these pathways .
Dual-Specific Antibodies: Expanding bimekizumab-like strategies to target IL-17A/F in inflammatory bowel disease and uveitis .
Cancer Therapeutics: Exploring IL-17A inhibition to suppress tumor-promoting pathways while mitigating off-target effects .
Manufacturing Optimization: Improving thermal stability and reducing aggregation to enhance antibody shelf life .
The IL17A monoclonal antibody was produced using recombinant human IL17A protein as an immunogen. The cDNA encoding the IL17A monoclonal antibody was sequenced and cloned into a plasmid vector. This vector was then transfected into a host cell using a suitable method. The resulting IL17A recombinant monoclonal antibody was purified using affinity chromatography. Its specificity was confirmed using ELISA, demonstrating reactivity only with human IL17A protein. The antibody exhibited specific binding to recombinant human IL17A (CSB-BP624104HU(M)) with an EC50 range of 1.818-2.170 ng/mL.
IL-17A is a cytokine protein produced by various immune cells, including T helper 17 (Th17) cells, γδ T cells, natural killer (NK) T cells, and certain innate lymphoid cells. Its primary function is to induce inflammatory responses, particularly in the context of defending against bacterial and fungal infections. IL-17A promotes the recruitment and activation of neutrophils and other immune cells to sites of infection, as well as the production of pro-inflammatory cytokines and chemokines. Additionally, IL-17A has been implicated in the pathogenesis of several autoimmune and inflammatory diseases, including rheumatoid arthritis, multiple sclerosis, and psoriasis.
IL-17A is an effector cytokine involved in both innate and adaptive immune responses, playing a crucial role in antimicrobial host defense and maintaining tissue integrity. It signals through the IL17RA-IL17RC heterodimeric receptor complex, triggering a homotypic interaction between IL17RA and IL17RC chains with the TRAF3IP2 adapter. This interaction initiates a downstream cascade involving TRAF6-mediated activation of the NF-κB and MAPkinase pathways, ultimately leading to the transcriptional activation of various genes. These genes include those encoding cytokines, chemokines, antimicrobial peptides, and matrix metalloproteinases, resulting in a potential for strong immune inflammation.
IL-17A acts as a critical link between T cell-mediated adaptive immunity and acute inflammatory responses, facilitating the destruction of extracellular bacteria and fungi. As a signature effector cytokine of T-helper 17 cells (Th17), it primarily induces neutrophil activation and recruitment to infection and inflammatory sites. In airway epithelium, IL-17A mediates neutrophil chemotaxis by inducing the production of CXCL1 and CXCL5 chemokines. In secondary lymphoid organs, it contributes to germinal center formation by regulating the chemotactic response of B cells to CXCL12 and CXCL13, enhancing B cell retention within germinal centers, somatic hypermutation rates, and selection towards plasma cells.
IL-17A is also an effector cytokine of a subset of γδ T cells, functioning as part of an inflammatory circuit downstream of IL1B, TLR2, and IL23A-IL12B to promote neutrophil recruitment for efficient bacterial clearance. It is also an effector cytokine of innate immune cells, including invariant natural killer (iNKT) cells and group 3 innate lymphoid cells, mediating initial neutrophilic inflammation. IL-17A contributes to the maintenance of epithelial barrier integrity during homeostasis and pathogen infection. Upon acute injury, it directly participates 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.
IL-17A, in synergy with IL17F, mediates the production of antimicrobial β-defensins DEFB1, DEFB103A, and DEFB104A by mucosal epithelial cells, limiting microbial entry through epithelial barriers. It is also involved in antiviral host defense through various mechanisms. IL-17A enhances immunity against West Nile virus by promoting T cell cytotoxicity. It may play a beneficial role in influenza A virus (H5N1) infection by enhancing B cell recruitment and immune response in the lung. IL-17A contributes to influenza A virus (H1N1) clearance by driving the differentiation of B-1a B cells, facilitating the production of virus-specific IgM antibodies at the first line of host defense.
IL-17A (also known as CTLA-8) is a 32 kDa disulfide-linked homodimer consisting of 136 amino acids that functions as a CD4+ T cell-derived cytokine. It promotes inflammatory responses and is elevated in numerous pathological conditions including rheumatoid arthritis, asthma, multiple sclerosis, psoriasis, and transplant rejection. IL-17A belongs to a family of six related proteins (IL-17A through IL-17F) and signals through the IL-17 receptor (IL-17R/CDw217). Its central role in mediating inflammatory responses makes it a crucial target for immunological research, particularly for developing therapeutic approaches for autoimmune and inflammatory disorders .
Recombinant monoclonal antibodies are produced using in vitro expression systems rather than hybridoma technology. These antibodies are generated by cloning specific genes that code for the desired antibody into an expression vector, which is then expressed in vitro. The key advantages include significantly better specificity and superior lot-to-lot consistency compared to traditional monoclonal antibodies. This manufacturing process allows for precise genetic manipulation to enhance binding affinity, species cross-reactivity, or introduce specific conjugation sites. Additionally, the recombinant approach eliminates concerns related to hybridoma instability or animal welfare considerations associated with traditional methods .
IL17A recombinant monoclonal antibodies are utilized across multiple research applications including:
Western Blot (WB) for protein detection and quantification
Immunohistochemistry (IHC) for tissue localization
Flow Cytometry for cellular analysis, particularly intracellular staining
Enzyme-Linked Immunosorbent Assay (ELISA) for quantitative detection
Immunofluorescence (IF) for subcellular localization
Each application requires specific optimization protocols, with particular attention to antibody concentration and buffer conditions. For intracellular staining applications, appropriate fixation and permeabilization methods are crucial for accessing the target protein while maintaining cellular architecture .
When working with IL17A antibodies across different species models, researchers must carefully validate cross-reactivity profiles. Based on available data, human IL17A antibodies may show variable cross-reactivity with mouse and rat orthologs. For instance, certain clones show 100% cross-reactivity with recombinant human (rh) IL-17A/IL-17F heterodimer but no cross-reactivity with rhIL-17F, recombinant mouse IL-17, or recombinant rat IL-17A .
To address this challenge:
Perform initial validation experiments with positive controls from each species
Consider using species-specific antibodies when possible
For cross-species studies, select antibodies raised against conserved epitopes
Document exact clone numbers and validation data in your research protocols
Utilize Western blotting with recombinant proteins to confirm specificity before proceeding to more complex applications
This approach prevents misinterpretation of data due to unexpected cross-reactivity patterns or lack of reactivity in certain species .
Detecting low levels of IL17A expression in immune cells requires careful optimization of experimental conditions:
Cell Stimulation Protocol: Treat peripheral blood mononuclear cells (PBMCs) with 50 ng/mL PMA and 250 ng/mL calcium ionomycin for 16 hours to upregulate IL-17A expression before antibody staining .
Fixation and Permeabilization: Use specialized fixation buffers followed by permeabilization solutions designed for intracellular cytokine detection. Standard protocols often fail to adequately expose intracellular epitopes of IL-17A.
Antibody Titration: Determine the optimal antibody concentration through titration experiments (typically starting at 1 μg/mL for Western blot applications) .
Signal Amplification: For very low expression levels, consider employing signal amplification systems such as tyramide signal amplification or polymer-based detection systems.
Controls: Always include appropriate positive controls (stimulated T cells) and negative controls (isotype antibodies matched to your primary antibody) .
These methodological refinements can significantly improve the signal-to-noise ratio when detecting limited IL17A expression in primary immune cells or tissue samples.
Differentiating between IL17A homodimers and IL17A/F heterodimers requires strategic experimental design:
Antibody Selection: Choose antibodies with documented specificity profiles. For example, certain IL17A antibodies show 100% cross-reactivity with IL-17A/IL-17F heterodimers while showing no cross-reactivity with IL-17F homodimers .
Sequential Immunoprecipitation: Perform initial immunoprecipitation with anti-IL17A, followed by Western blotting with anti-IL17F (and vice versa) to identify heterodimers.
Molecular Weight Analysis: Under reducing conditions, IL17A appears at approximately 17 kDa on Western blots, while IL17A/F heterodimers may show distinct banding patterns between 14-19 kDa .
Recombinant Protein Controls: Include purified recombinant IL17A, IL17F, and IL17A/F heterodimers as controls in your experiments to establish distinct detection patterns.
Mass Spectrometry: For definitive differentiation, consider targeted mass spectrometry approaches after immunoprecipitation to identify specific peptide sequences unique to each dimer configuration.
This multi-faceted approach enables researchers to accurately distinguish between these structurally similar but functionally distinct cytokine configurations .
Successful Western blot analysis with IL17A antibodies requires several critical optimization steps:
Sample Preparation: For primary human samples, CD4+ T cells must be properly activated to express detectable IL17A. For recombinant systems, transfection efficiency should be verified.
Reduction Conditions: Use reducing conditions (typically including β-mercaptoethanol or DTT) as IL17A contains disulfide bonds that affect epitope accessibility. Documented protocols confirm detection under reducing conditions using Immunoblot Buffer Group 1 .
Antibody Concentration: Start with 1 μg/mL of anti-IL17A monoclonal antibody, followed by appropriate HRP-conjugated secondary antibody .
Membrane Selection: PVDF membranes have demonstrated superior results compared to nitrocellulose for IL17A detection .
Expected Molecular Weight: Look for specific bands at approximately 17 kDa for native IL17A, though in transfection systems bands may appear between 14-19 kDa due to variations in post-translational modifications .
Controls: Include positive controls (stimulated CD4+ T cells) and negative controls (mock transfected cells) to verify specificity .
These methodological refinements significantly improve reproducibility and sensitivity when detecting IL17A via Western blot.
Intracellular staining for IL17A requires attention to several critical parameters:
Cell Stimulation: Stimulate PBMCs with 50 ng/mL PMA and 250 ng/mL calcium ionomycin for 16 hours to upregulate IL17A expression .
Fixation Protocol: Use specialized flow cytometry fixation buffers that maintain cellular architecture while enabling antibody access to intracellular targets .
Permeabilization Strategy: Apply permeabilization/wash buffers specifically designed for intracellular cytokine detection rather than general permeabilization reagents .
Antibody Selection: Choose antibodies validated specifically for flow cytometry applications, as not all IL17A antibodies perform equally across different applications .
Controls: Include unstimulated cells as negative controls, isotype controls to assess non-specific binding, and fluorescence-minus-one (FMO) controls for accurate gating strategies .
Multicolor Panel Design: When incorporating IL17A detection into multicolor panels, consider spectral overlap and compensation requirements, particularly when using bright fluorophores.
Adherence to these methodological guidelines enables reliable detection of IL17A-producing cells in complex samples like PBMCs or tissue digests.
Comprehensive validation of IL17A recombinant monoclonal antibodies should follow this multi-step approach:
ELISA Validation: Perform direct ELISAs against recombinant IL17A, IL17F, and IL17A/F heterodimers to establish specificity profiles. Document cross-reactivity percentages with related proteins .
Western Blot Verification: Test antibodies against both recombinant proteins and biological samples (e.g., activated T cells known to express IL17A) .
Knockout/Knockdown Controls: When possible, include IL17A knockout cells or knockdown systems as negative controls to confirm specificity.
Epitope Mapping: Determine the specific epitope recognized by the antibody to predict potential cross-reactivity with related proteins.
Blocking Experiments: Perform pre-adsorption experiments with recombinant IL17A to demonstrate that binding can be specifically blocked.
Species Cross-Reactivity Assessment: Systematically test against IL17A from different species if cross-species applications are intended .
This comprehensive validation strategy ensures experimental reliability and facilitates accurate interpretation of results when using these antibodies in research applications.
IL17A has a distinct molecular structure that influences antibody targeting strategies:
Structural Feature | Details | Relevance to Antibody Development |
---|---|---|
Molecular Weight | 32 kDa (homodimer) | Affects migration patterns in Western blots and purification strategies |
Primary Structure | 136 amino acids | Provides multiple potential epitopes for antibody recognition |
Active Form | Disulfide-linked homodimer | Structural epitopes may be lost in reducing conditions |
Key Domain | IL17 domain (Ile20-Ala155) | Primary target region for most antibodies |
Signal Peptide | Present (removed in mature protein) | Antibodies targeting this region won't recognize mature IL17A |
Glycosylation | N-linked glycosylation sites | May affect antibody accessibility to certain epitopes |
Understanding these structural features is essential for selecting appropriate antibodies and interpreting experimental results. Most validated IL17A recombinant monoclonal antibodies target epitopes within the IL17 domain (Ile20-Ala155), which contains the functionally important receptor-binding regions .
Different IL17A recombinant monoclonal antibody clones exhibit distinct characteristics important for experimental design:
Clone Designation | Host | Applications | Species Reactivity | Cross-Reactivity Profile | Notable Features |
---|---|---|---|---|---|
eBio64DEC17 | Mouse | Multiple | Human, Mouse, Rat | Broad species reactivity | Validated for multiple applications |
41802R | Mouse | WB, Flow Cytometry | Human | 100% with IL17A/F heterodimer, 0% with IL17F | Highly specific for human IL17A |
Various polyclonal | Rabbit | WB, IF, IHC | Mouse, Rat | Variable | Broader epitope recognition |
Anti-IL17A(secukinumab biosimilar) | - | ELISA, FCM | Human | Highly specific | Therapeutic antibody analog |
This comparative analysis highlights the importance of selecting the appropriate clone based on your specific experimental requirements, including species of interest, application, and need for distinguishing between IL17A homodimers and IL17A/F heterodimers .
IL17A regulates multiple signal transduction pathways that can serve as functional readouts for antibody efficacy:
Signaling Pathway | IL17A-Induced Effect | Potential Readout Assays | Relevance to Antibody Testing |
---|---|---|---|
NF-κB | Activation | Luciferase reporter assays, p65 nuclear translocation | Primary pathway for validating neutralizing activity |
MAPK Pathways | Activation | Phospho-flow cytometry, Western blot for phospho-ERK/JNK/p38 | Rapid response suitable for acute neutralization assays |
IL-6 Expression | Upregulation | ELISA, qPCR | Well-documented downstream effect of IL17A signaling |
Cyclooxygenase-2 (PTGS2/COX-2) | Enhanced expression | Western blot, enzyme activity assays | Important inflammatory mediator regulated by IL17A |
Nitric Oxide (NO) Production | Enhanced production | Griess assay, DAF-FM diacetate staining | Functional readout in various cell types |
ICAM-1 Expression | Enhanced expression | Flow cytometry, cell adhesion assays | Important for fibroblast studies |
These pathways provide functional validation of antibody efficacy beyond simple binding assays. When testing neutralizing antibodies against IL17A, researchers should monitor at least two independent signaling outputs to confirm complete blockade of biological activity .
Researchers selecting IL17A recombinant monoclonal antibodies should consider several critical factors to ensure experimental success:
Application Suitability: Different antibody clones perform optimally in specific applications. Verify that the antibody has been validated for your intended use (Western blot, flow cytometry, IHC, etc.) .
Species Reactivity: Confirm that the antibody recognizes IL17A from your species of interest. Many antibodies exhibit restricted species reactivity profiles .
Specificity Profile: Determine whether the antibody can distinguish between IL17A homodimers and IL17A/F heterodimers if this distinction is relevant to your research question .
Clone Characteristics: Recombinant monoclonal antibodies offer superior lot-to-lot consistency and specificity compared to other antibody types, making them preferable for longitudinal studies or quantitative applications .
Validation Data: Review available validation data, including Western blot images, ELISA cross-reactivity profiles, and flow cytometry histograms to ensure the antibody performs as expected .
Titration Requirements: All antibodies require careful titration for optimal performance. Budget time for this optimization step in your experimental planning .