IL 17 Human

Interleukin-17 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Biological Functions

IL-17A drives both protective and pathological immune responses:

Host Defense

  • Antimicrobial Peptide Induction: Stimulates β-defensins and S100A8 in epithelial cells to combat pathogens like Candida albicans .

  • Neutrophil Recruitment: Upregulates CXCL1, CXCL8, and G-CSF to mobilize neutrophils .

  • Barrier Integrity: Enhances tight junction proteins (e.g., claudins) in intestinal and mucosal epithelia .

Inflammatory Regulation

  • Cytokine Amplification: Synergizes with TNF-α and IL-1β to amplify IL-6, IL-8, and matrix metalloproteinases .

  • Autoimmunity: Promotes tissue damage in rheumatoid arthritis (via osteoclastogenesis) , psoriasis (keratinocyte activation) , and type 1 diabetes (islet cell apoptosis) .

Cellular Sources of IL-17A

While Th17 cells are the primary producers, IL-17A is also secreted by:

  • γδ T cells and CD8+ T cells during acute infections .

  • Innate Lymphoid Cells (ILC3s) and Neutrophils in mucosal tissues .

  • Paneth Cells in the gut, maintaining microbial homeostasis .

Autoimmune Disorders

  • Rheumatoid Arthritis: Synovial fibroblasts exposed to IL-17A overproduce IL-6 and MMPs, driving joint destruction .

  • Type 1 Diabetes: IL-17A exacerbates inflammatory and apoptotic responses in pancreatic islets .

  • Psoriasis: IL-17A/IL-23 axis promotes keratinocyte hyperproliferation and neutrophil infiltration .

Cancer

  • Pro-Tumor Effects: Enhances angiogenesis and immunosuppression in melanoma and colorectal cancer .

  • Anti-Tumor Effects: Activates CD8+ T cells and NK cells in certain contexts .

Research Tools and Assays

HEK-Blue™ IL-17 cells are widely used to quantify bioactive IL-17 isoforms :

ParameterSpecification
Detectable IsoformsHuman IL-17A, IL-17F, IL-17E (IL-25)
Detection Range1–100 ng/mL (IL-17A), 3–100 ng/mL (IL-17F)
Pathway ReadoutNF-κB/AP-1 activation via SEAP reporter .

Therapeutic Targeting

Clinical trials focus on IL-17A neutralization:

  • Secukinumab (anti-IL-17A): Approved for psoriasis and ankylosing spondylitis .

  • Brodalumab (IL-17RA blocker): Effective in moderate-to-severe plaque psoriasis .

  • Challenges: IL-17 blockade may increase susceptibility to mucocutaneous infections .

Recent Advances (2023–2025)

  • Dual IL-17A/F Inhibitors: Bimekizumab shows superior efficacy in psoriatic arthritis .

  • Microbiome Modulation: Gut microbiota-derived metabolites regulate Th17 differentiation, offering novel therapeutic avenues .

Product Specs

Introduction
IL-17, also known as interleukin-17A, is a signaling protein that plays a role in immune responses, particularly in the context of inflammation. It is produced by a subset of T helper cells called Th17 cells, as well as other immune cells. IL-17 acts on various cell types, including epithelial cells, fibroblasts, and immune cells, to promote the production of inflammatory mediators such as cytokines, chemokines, and matrix metalloproteinases. These mediators contribute to the recruitment of immune cells to the site of inflammation and the development of inflammatory responses. Dysregulation of IL-17 signaling has been implicated in the pathogenesis of various autoimmune and inflammatory diseases, making it a target for therapeutic intervention in conditions such as rheumatoid arthritis, psoriasis, and inflammatory bowel disease.
Description
Recombinant human interleukin-17A, expressed in E. coli, is a non-glycosylated polypeptide chain with a molecular weight of approximately 31 kDa. It comprises two identical subunits, each containing 132 amino acids, forming a homodimeric structure. The purification process involves proprietary chromatographic techniques to ensure high purity. This product is intended for research purposes only.
Physical Appearance
Sterile, white, lyophilized (freeze-dried) powder.
Formulation
Provided as a sterile, lyophilized powder in a buffer composed of 20 mM citrate, 0.1 M NaCl, at pH 4.5.
Solubility
To reconstitute, it is recommended to dissolve the lyophilized interleukin-17A in sterile, 18 MΩ-cm H2O to a concentration of at least 100 µg/ml. This solution can be further diluted in other aqueous solutions as needed.
Stability
Lyophilized interleukin-17A, when stored desiccated at or below -18°C, is stable for 3 weeks at room temperature. Once reconstituted, it should be stored at 4°C for up to 7 days. For long-term storage, it is advisable to add a carrier protein, such as 0.1% HSA or BSA, and store at or below -18°C. Avoid repeated freeze-thaw cycles.
Purity
The purity is determined to be greater than 95.0% as assessed by SDS-PAGE.
Biological Activity
The biological activity, as measured by the dose-dependent induction of IL-6 in the Hs68 cell line, exhibits an ED50 of approximately 2 ng/ml, corresponding to a specific activity of 500,000 IU/mg.
Protein Content
Protein quantification was conducted using two independent methods: 1. UV spectroscopy at 280 nm, utilizing an absorbance value of 1.13 as the extinction coefficient for a 0.1% (1 mg/ml) solution, calculated using the PC GENE computer analysis program (IntelliGenetics). 2. Analysis by RP-HPLC, employing a standard solution of IL-17 as a reference standard.
Synonyms
CTLA-8, IL-17, IL-17A, Cytotoxic T-lymphocyte-associated antigen 8.
Source
Escherichia Coli.

Q&A

What is the IL-17 family and how does IL-17A differ from other family members?

The IL-17 family consists of six structurally related cytokines (IL-17A through IL-17F). IL-17A is the prototypical and most well-studied member, often simply referred to as "IL-17." While IL-17F shares the most similarities with IL-17A in terms of cellular sources and function, other family members like IL-17C have distinct roles. IL-17A and IL-17F are co-expressed on linked genes and typically co-produced by Type 17 cells. They can exist as homodimers or as an IL-17A/F heterodimer. All these forms signal through the same receptor complex (IL-17RA/IL-17RC heterodimer), but IL-17A homodimers induce a much stronger signal compared to IL-17F homodimers, with the IL-17A/F heterodimer producing intermediate signaling strength .

Unlike IL-17A, which is produced by immune cells, IL-17C is primarily produced by epithelial cells and acts in an autocrine manner to promote anti-microbial responses and barrier maintenance in the skin and intestine . This distinction highlights the tissue-specific and context-dependent functions of different IL-17 family members.

Which cell types produce IL-17A in humans?

IL-17A is produced by multiple cell populations in humans. The primary sources include:

  • CD4+ T helper 17 (Th17) cells, which are characterized by expression of the "master" transcription factor RORγt

  • CD8+ T cells (Tc17)

  • Various innate lymphocytes including:

    • γδ T cells

    • Natural killer T (NKT) cells

    • Group 3 innate lymphoid cells (ILC3)

    • 'Natural' Th17 cells

Some reports also suggest that myeloid-lineage cells such as neutrophils and microglia can produce IL-17A, though this remains somewhat controversial and less well understood . In experimental settings, human peripheral blood mononuclear cells (PBMCs) treated with PMA and calcium ionomycin for 16 hours show detectable IL-17 production that can be measured by flow cytometry .

How is IL-17A signaling initiated and regulated in human cells?

IL-17A signaling in humans begins when the cytokine binds to a heterodimeric receptor complex consisting of IL-17RA paired with IL-17RC. Crystal structure studies of the human IL-17RA extracellular domain complexed to IL-17F homodimers have revealed an unusual fibronectin III domain conformation .

The binding process occurs in a stepwise manner: after either IL-17RA or IL-17RC binds to IL-17A, the affinity for the reciprocal subunit increases, which is consistent with studies showing ligand-inducible association of IL-17RA and IL-17RC on the cell surface . In humans, IL-17RA has a much higher affinity for IL-17A than for IL-17F, while IL-17RC binds both ligands with similar affinity .

Once activated, the IL-17 receptor complex triggers multiple signaling pathways that ultimately lead to the induction of pro-inflammatory genes, including chemokines (CXCL1, CXCL2, CXCL8/IL-8), cytokines (IL-6, G-CSF), and antimicrobial peptides (β-defensins, S100A8, lipocalin 2) . These molecular responses collectively promote neutrophil recruitment and activation at sites of inflammation or infection.

What are the primary methods for detecting IL-17A in human samples?

Several validated methods are available for detecting IL-17A in human samples:

  • Western Blot: Can be used to detect IL-17A in cell lysates, such as from human primary naïve CD4+ T cells or differentiated Th17 cells. Under reducing conditions, human IL-17A appears as a band at approximately 15 kDa .

  • Immunohistochemistry (IHC): IL-17A can be detected in tissue sections using specific antibodies. For example, IL-17A has been detected in paraffin-embedded sections of human tonsil, where staining is localized to lymphocytes . IHC has also been used to quantify IL-17A+ cells in skin lesions from psoriasis patients .

  • Flow Cytometry: Intracellular IL-17A can be detected in human PBMCs, especially after stimulation with PMA and calcium ionomycin. This requires fixation with paraformaldehyde and permeabilization with saponin before staining with anti-IL-17A antibodies .

  • Immunoprecipitation: IL-17A can be immunoprecipitated from human cell lysates (e.g., differentiated Th17 cells) using specific antibodies and protein G sepharose, followed by detection by Western blot .

  • Functional Assays: The biological activity of IL-17A can be assessed by measuring IL-6 secretion from responsive cells (like NIH/3T3 fibroblasts) after stimulation with recombinant human IL-17A .

How do Th17 cells develop in humans, and what factors influence IL-17A production?

Human Th17 cell development represents a complex process involving multiple cytokines and transcription factors. Naïve CD4+ T cells can differentiate into Th17 cells in response to several differentiation triggers:

  • TGFβ

  • IL-6

  • IL-1β

  • IL-21

The master transcription factor for Th17 development is RORγt. While IL-23 is not required for initial Th17 differentiation, it is essential for maintenance and functional activity of these cells in vivo .

A detailed analysis of the human IL-17A promoter has revealed multiple transcription factors that can positively or negatively regulate IL-17A expression . The development pathway differs somewhat from mouse models, which has important implications for translational research.

Experimentally, human Th17 cells can be generated in vitro by stimulating naïve CD4+ T cells with the appropriate cytokine cocktail. The resulting cells produce IL-17A that can be detected in both the cell lysate and culture supernatant, as demonstrated by Western blot analyses .

What are the dual roles of IL-17A in human disease and homeostasis?

IL-17A exhibits a fascinating dichotomy in human health:

Protective Roles:

  • Crucial for defense against extracellular fungal and bacterial pathogens

  • Important for barrier surface protection and repair

  • Induces antimicrobial peptides that provide immediate protection against microbial invasion

  • Promotes neutrophil recruitment through chemokine induction (CXCL1, CXCL2, CXCL8/IL-8)

  • Induces IL-6 and G-CSF, cytokines that promote myeloid-driven innate inflammation

Pathogenic Roles:

  • Drives inflammation in autoimmune diseases, particularly rheumatoid arthritis (RA), psoriasis, and multiple sclerosis

  • Contributes to immunopathology in inflammatory syndromes

  • Implicated in both promotion and inhibition of various cancers, depending on context

This duality reflects the evolutionary conservation of IL-17A - its inflammatory activities have been, on balance, beneficial for human survival, but these same properties can become problematic under certain conditions. The context- and tissue-dependent functions of IL-17A highlight the nuanced balance between its protective and pathogenic roles .

How do IL-17A antagonists show efficacy in human autoimmune disorders, and what biomarkers predict response?

Anti-IL-17A antibodies have shown significant efficacy in treating several human autoimmune disorders, particularly psoriasis, rheumatoid arthritis, and autoimmune uveitis. Clinical trials have reported promising results with these therapies . The fundamental mechanism involves blocking the pro-inflammatory effects of IL-17A at tissue sites.

Interestingly, a single nucleotide polymorphism (SNP) in the IL-23R gene (R381Q) is associated with reduced susceptibility to multiple autoimmune disorders and has been linked to reduced IL-17A production in human T cells . This genetic marker represents one potential biomarker that might predict response to IL-17A-targeted therapies.

Research has also identified tissue-specific biomarkers in conditions like psoriasis. Immunohistochemistry analyses of skin lesions show characteristic patterns of IL-17A+ cells, which can be quantitatively compared between classical psoriasis and paradoxical psoriasiform reactions. These analyses reveal that paradoxical skin lesions may show similar values of IL-17A+ cells but different patterns of other cytokines such as IFN-gamma, IL-22, and IL-36 gamma .

What are the key differences between human and mouse IL-17 receptor systems, and how does this impact translational research?

Several important differences exist between human and mouse IL-17 receptor systems that can significantly impact translational research:

  • Ligand-Receptor Affinity: Human IL-17RA has much higher affinity for IL-17A than for IL-17F, whereas human IL-17RC binds both ligands with similar affinity. In contrast, murine IL-17RC primarily binds to IL-17F .

  • Expression Patterns: There may be differences in tissue-specific expression patterns of IL-17 receptors between humans and mice.

  • Downstream Signaling: While the core signaling pathways are conserved, there can be species-specific differences in signaling intensity or regulation.

These differences highlight the challenges in translating findings from mouse models to human disease. In designing preclinical studies, researchers must account for these species-specific variations to maximize translational relevance. Human in vitro systems, humanized mouse models, or direct studies on human samples may provide more reliable data for predicting human responses to IL-17-targeted therapies.

What methodological advances have improved detection and functional analysis of IL-17A in human samples?

Recent methodological advances have significantly enhanced our ability to detect and analyze IL-17A in human samples:

  • Single-cell Analysis: Single-cell RNA sequencing and mass cytometry (CyTOF) now allow researchers to identify IL-17A-producing cells with unprecedented resolution, enabling the discovery of previously unrecognized cellular sources.

  • Improved Antibodies: Highly specific antibodies against human IL-17A, such as those validated for multiple applications (Western blot, IHC, flow cytometry, and immunoprecipitation) , have improved detection sensitivity and specificity.

  • Functional Assays: Neutralization assays measuring IL-6 secretion induced by IL-17A in reporter cell lines provide quantitative assessment of IL-17A biological activity and antibody efficacy. The neutralization dose (ND50) for typical anti-IL-17A antibodies ranges from 0.02-0.12 μg/mL in the presence of 15 ng/mL recombinant human IL-17A .

  • Tissue Imaging: Advanced immunohistochemistry techniques allow precise quantification of IL-17A+ cells in tissues. For example, researchers have used this approach to compare IL-17A expression in different types of psoriatic skin lesions .

  • Ex vivo Models: Development of ex vivo human tissue models has allowed for more physiologically relevant studies of IL-17A functions in specific tissue microenvironments.

How should researchers design experiments to study IL-17A production in human primary cells?

When designing experiments to study IL-17A production in human primary cells, researchers should consider the following recommendations:

  • Cell Source Selection:

    • Peripheral blood mononuclear cells (PBMCs) are readily accessible but contain relatively few IL-17A-producing cells under baseline conditions

    • Consider tissue-specific sources for certain studies (e.g., synovial fluid cells for rheumatoid arthritis research)

    • Use flow cytometry sorting to isolate specific T cell subsets for more focused analyses

  • Stimulation Protocols:

    • For acute stimulation: PMA (50 ng/mL) and calcium ionomycin (250 ng/mL) for 16 hours is an established protocol to induce IL-17A production

    • For Th17 differentiation: Culture naïve CD4+ T cells with a combination of TGFβ, IL-6, IL-1β, and IL-21, followed by IL-23 for maintenance

    • Include appropriate positive controls (e.g., established Th17 cell lines) and negative controls

  • Detection Methods:

    • Intracellular cytokine staining with flow cytometry provides single-cell resolution

    • ELISA or multiplex assays for secreted IL-17A in culture supernatants

    • qRT-PCR for IL17A mRNA expression

    • Western blot can detect IL-17A protein in cell lysates and supernatants

  • Experimental Validation:

    • Confirm specificity with neutralizing antibodies

    • Include functional readouts (e.g., downstream target gene expression)

    • Consider co-staining for other Th17-associated molecules (RORγt, IL-22, CCR6)

What are the optimal sample preparation techniques for analyzing IL-17A in human tissue specimens?

Optimal sample preparation for IL-17A analysis in human tissues depends on the analytical method and tissue type:

  • For Immunohistochemistry:

    • Fixation: Immersion fixation with paraformaldehyde or formalin followed by paraffin embedding works well for most tissues

    • Antigen Retrieval: Often necessary for formalin-fixed tissues; methods include heat-induced epitope retrieval in citrate buffer (pH 6.0)

    • Blocking: Use appropriate blocking solutions to minimize background staining

    • Primary Antibody: Validate concentration (e.g., 1 μg/mL) and incubation time (typically 1 hour at room temperature)

    • Detection: HRP-polymer systems offer good sensitivity with low background

    • Counterstaining: Hematoxylin provides good nuclear contrast

    • Controls: Include isotype controls and known positive tissues (e.g., tonsil sections)

  • For Flow Cytometry:

    • Tissue Disaggregation: Optimize enzymatic digestion protocols to maintain cell viability and surface markers

    • Fixation/Permeabilization: Paraformaldehyde fixation followed by saponin permeabilization is effective for IL-17A staining

    • Stimulation: Consider a brief stimulation period (e.g., 4-6 hours) with PMA/ionomycin plus a protein transport inhibitor

    • Antibody Selection: Use fluorophores appropriate for the tissue's autofluorescence profile

  • For Protein Extraction and Western Blot:

    • Extraction Buffers: Use buffers containing appropriate protease inhibitors

    • Sample Storage: Flash-freeze tissues and store at -80°C until processing

    • Protein Quantification: Standardize loading based on total protein concentration

    • Electrophoresis Conditions: Run under reducing conditions to detect IL-17A at approximately 15 kDa

How can researchers differentiate between the various forms of IL-17 (IL-17A homodimers, IL-17F homodimers, and IL-17A/F heterodimers) in experimental settings?

Differentiating between IL-17A homodimers, IL-17F homodimers, and IL-17A/F heterodimers requires specialized techniques:

  • Specific Antibodies:

    • Use antibodies that exclusively recognize IL-17A or IL-17F epitopes

    • For heterodimer detection, employ antibody pairs that capture one subunit and detect the other

  • Surface Plasmon Resonance (SPR):

    • SPR can distinguish between these forms based on their differential binding kinetics to IL-17RA and IL-17RC

    • Human IL-17RA has much higher affinity for IL-17A than for IL-17F, while human IL-17RC binds both ligands with similar affinity

  • Non-denaturing Electrophoresis:

    • Native PAGE followed by Western blotting can separate the different dimers based on size and charge

    • Follow with specific antibody detection

  • Functional Assays:

    • Compare biological activities, as IL-17A homodimers induce stronger signaling than IL-17F homodimers, with IL-17A/F heterodimers showing intermediate potency

    • Use reporter cell lines expressing human IL-17RA/RC complexes and measure downstream responses

  • Recombinant Standards:

    • Include purified recombinant IL-17A homodimers, IL-17F homodimers, and IL-17A/F heterodimers as reference standards

What are the most reliable bioassays for measuring IL-17A functional activity in human samples?

The most reliable bioassays for measuring IL-17A functional activity in human samples include:

  • IL-6 Induction Assay:

    • Fibroblast cell lines (such as NIH/3T3) respond to IL-17A by secreting IL-6

    • Quantify IL-6 production using ELISA following stimulation with test samples

    • Include a standard curve with recombinant human IL-17A

    • Confirm specificity by including neutralizing anti-IL-17A antibodies

    • The typical neutralization dose (ND50) range is 0.02-0.12 μg/mL in the presence of 15 ng/mL recombinant human IL-17A

  • Chemokine Production Assay:

    • Measure CXCL8 (IL-8) production from epithelial or endothelial cells following IL-17A stimulation

    • This assay captures a key biological function of IL-17A in promoting neutrophil recruitment

  • Reporter Cell Lines:

    • Engineered cell lines expressing IL-17RA/RC and a reporter construct (e.g., luciferase) driven by IL-17A-responsive promoters

    • Provide rapid, sensitive, and quantitative readout of IL-17A bioactivity

  • Gene Expression Analysis:

    • qRT-PCR measurement of IL-17A-induced genes (CXCL1, CXCL2, CXCL8, S100A8, β-defensins)

    • Can be performed on stimulated primary cells or cell lines

  • Neutrophil Migration Assay:

    • Measure neutrophil chemotaxis in response to conditioned media from IL-17A-stimulated cells

    • Provides functional assessment of IL-17A's role in neutrophil recruitment

How should researchers address inconsistent IL-17A detection in human samples?

Inconsistent IL-17A detection is a common challenge in human research. To address this issue:

  • Sample Handling and Processing:

    • Standardize collection procedures, processing times, and storage conditions

    • Consider that IL-17A may be unstable in certain preservation media or during freeze-thaw cycles

    • For blood samples, process within 2-4 hours of collection to prevent ex vivo changes

  • Stimulation Conditions:

    • Baseline IL-17A production is often low; consider ex vivo stimulation with PMA/ionomycin

    • Standardize stimulation protocols (duration, concentrations, media composition)

    • Include positive controls (known IL-17A producers) to verify assay functionality

  • Detection Method Optimization:

    • Optimize antibody concentrations and incubation conditions

    • For IHC, test different antigen retrieval methods and detection systems

    • For flow cytometry, ensure proper fixation/permeabilization protocols

    • For ELISA, consider high-sensitivity kits and optimize sample dilutions

  • Technical Validation:

    • Use multiple detection methods to cross-validate findings

    • Include spike-in recovery experiments to assess matrix effects

    • Consider blocking potentially interfering factors in complex samples

  • Biological Factors:

    • Account for circadian variations in cytokine production

    • Consider patient heterogeneity (age, sex, comorbidities, medications)

    • Assess potential confounding effects of other inflammatory mediators

What are the critical controls needed when analyzing IL-17A expression and function in experimental settings?

Critical controls for IL-17A research include:

  • Positive Controls:

    • Recombinant human IL-17A at known concentrations

    • Differentiated Th17 cells from healthy donors

    • Human tonsil tissue sections for IHC (known to contain IL-17A+ lymphocytes)

    • PMA/ionomycin-stimulated PBMCs for flow cytometry

  • Negative Controls:

    • Isotype-matched control antibodies for immunostaining and neutralization studies

    • Unstimulated cells or tissues

    • Samples from relevant IL-17A-deficient systems (when available)

    • Samples with confirmed absence of IL-17A expression

  • Specificity Controls:

    • Pre-absorption of antibodies with recombinant IL-17A

    • siRNA knockdown of IL-17 receptors in target cells

    • Neutralizing antibodies to confirm observed effects are IL-17A-specific

    • Competitive binding assays

  • Technical Controls:

    • Standard curves for quantitative assays

    • Housekeeping genes/proteins for normalization in expression studies

    • Vehicle controls for all treatments

    • Multiple biological and technical replicates

  • Cross-Validation:

    • Confirm findings using multiple detection methods

    • Validate biological activity with functional assays

    • Correlate protein detection with mRNA expression

How do researchers interpret conflicting data regarding IL-17A's role in specific human diseases?

Interpreting conflicting data on IL-17A's role in human diseases requires a systematic approach:

  • Contextual Analysis:

    • Consider disease stage and activity - IL-17A may have different roles in initiation versus progression

    • Evaluate tissue specificity - IL-17A functions can vary dramatically across tissue microenvironments

    • Assess patient heterogeneity - genetic backgrounds may influence IL-17A responses

  • Methodological Evaluation:

    • Compare sample types (blood versus affected tissue) and processing methods

    • Consider sensitivity and specificity of detection methods

    • Evaluate whether studies measured IL-17A expression, activity, or downstream effects

    • Assess whether appropriate controls were included

  • Biological Complexity:

    • Recognize that IL-17A operates within a complex cytokine network

    • Consider compensatory mechanisms when IL-17A is blocked

    • Evaluate potential redundancy with other IL-17 family members (especially IL-17F)

    • Assess the balance between IL-17A's protective and pathogenic functions

  • Translational Discrepancies:

    • Account for differences between human and mouse IL-17 receptor systems

    • Consider limitations of animal models in recapitulating human disease

    • Evaluate whether in vitro findings translate to in vivo contexts

  • Clinical Correlation:

    • Compare observational data with interventional studies using IL-17A blockers

    • Consider treatment responses in different patient subgroups

    • Assess whether clinical trial outcomes align with mechanistic predictions

What statistical approaches are most appropriate for analyzing IL-17A data in clinical and experimental studies?

The most appropriate statistical approaches for IL-17A data analysis depend on the study design and data characteristics:

  • For Continuous IL-17A Measurements:

    • Assess normality using Shapiro-Wilk or Kolmogorov-Smirnov tests

    • For normally distributed data: t-tests, ANOVA, or linear regression

    • For non-normally distributed data (common with cytokines): non-parametric tests (Mann-Whitney U, Kruskal-Wallis) or log transformation

    • Consider repeated measures approaches for longitudinal data

  • For Categorical IL-17A Data:

    • Chi-square or Fisher's exact test for comparing proportions of IL-17A+ cells

    • Binary logistic regression for predictive models

    • Receiver operating characteristic (ROC) analysis for determining optimal cut-off values

  • For Correlation Analyses:

    • Pearson correlation for normally distributed data

    • Spearman rank correlation for non-normally distributed data

    • Multiple regression for controlling confounding factors

    • Path analysis for examining indirect relationships

  • For Clinical Studies:

    • Sample size calculations should account for expected variability in IL-17A measures

    • Consider stratified analyses based on relevant clinical or genetic factors

    • Use multivariate methods to adjust for potential confounders

    • Time-to-event analyses (Kaplan-Meier, Cox regression) for outcome studies

  • For High-Dimensional Data:

    • Principal component analysis or t-SNE for dimensionality reduction

    • Hierarchical clustering to identify IL-17A-related patient subgroups

    • Machine learning approaches for complex datasets

    • Network analysis to understand IL-17A's position within cytokine networks

What emerging technologies might advance understanding of IL-17A biology in human health and disease?

Several emerging technologies hold promise for advancing IL-17A research:

  • Single-Cell Multi-Omics:

    • Integration of single-cell transcriptomics, proteomics, and epigenomics to comprehensively characterize IL-17A-producing cells

    • Spatial transcriptomics to map IL-17A expression within complex tissue architectures

    • Single-cell secretome analysis to link cellular phenotypes with IL-17A production capacity

  • Advanced Imaging Technologies:

    • Multiplex immunofluorescence to simultaneously visualize IL-17A with multiple markers

    • Intravital microscopy to track IL-17A-producing cells in real-time

    • Mass cytometry imaging to quantify IL-17A in tissue contexts with high multiplexing capacity

  • CRISPR-Based Approaches:

    • Precision gene editing to study IL-17A regulation in primary human cells

    • CRISPR activation/interference systems to modulate IL-17A expression

    • CRISPR screens to identify novel regulators of IL-17A production or signaling

  • Organoid and Microphysiological Systems:

    • Human tissue-specific organoids to study IL-17A effects in physiologically relevant contexts

    • Organ-on-chip platforms to examine IL-17A functions at tissue interfaces

    • 3D bioprinting to create complex tissue models for IL-17A research

  • Systems Biology Approaches:

    • Multi-scale computational modeling to predict IL-17A network responses

    • Machine learning algorithms to identify IL-17A-associated biomarkers

    • Network pharmacology to discover novel modulators of IL-17A pathways

How might researchers better distinguish beneficial versus pathogenic IL-17A responses in human tissues?

Distinguishing beneficial versus pathogenic IL-17A responses requires sophisticated approaches:

  • Temporal Analysis:

    • Compare acute versus chronic IL-17A exposure effects on tissues

    • Develop time-resolved techniques to track the evolution of IL-17A responses

    • Employ inducible systems to control the timing of IL-17A signaling

  • Microenvironmental Context:

    • Analyze how tissue-specific factors modify IL-17A responses

    • Assess the influence of the local cytokine milieu on IL-17A function

    • Evaluate how different cell types in a tissue respond to IL-17A

  • Receptor and Signaling Dynamics:

    • Investigate differential engagement of signaling pathways downstream of IL-17 receptors

    • Examine how receptor expression levels affect response outcomes

    • Study post-translational modifications that might switch IL-17A function

  • Cellular Source Analysis:

    • Determine if IL-17A from different cellular sources (Th17 cells versus γδ T cells, etc.) has distinct functional effects

    • Develop methods to selectively modulate IL-17A from specific cell populations

  • Comparative Disease Studies:

    • Compare IL-17A signatures in protective scenarios (e.g., infection defense) versus pathogenic conditions (e.g., autoimmunity)

    • Identify biomarkers that distinguish protective versus harmful IL-17A responses

    • Develop methods to selectively inhibit pathogenic IL-17A signaling while preserving beneficial functions

Product Science Overview

Introduction

Interleukin-17 (IL-17) is a family of pro-inflammatory cytokines that play a crucial role in the immune response. The IL-17 family consists of six members: IL-17A, IL-17B, IL-17C, IL-17D, IL-17E (also known as IL-25), and IL-17F . These cytokines are produced by a subset of T helper cells known as Th17 cells, which are stimulated by IL-23 .

Discovery and Structure

IL-17A, the first member of the IL-17 family, was discovered in 1993 by Rouvier et al., who isolated the IL-17A transcript from a rodent T-cell hybridoma . The protein encoded by IL-17A exhibits high homology with a viral IL-17-like protein found in the genome of Herpesvirus saimiri . The IL-17 family members share a conserved cysteine-rich region, which is crucial for their structure and function .

Biological Functions

IL-17 cytokines are key players in the immune response, particularly in promoting inflammation. They act by binding to their respective receptors, which include IL-17RA, IL-17RB, and IL-17RC . Upon binding, IL-17 activates several signaling cascades that lead to the induction of chemokines. These chemokines recruit immune cells, such as monocytes and neutrophils, to the site of inflammation . IL-17 works in concert with other cytokines, such as tumor necrosis factor (TNF) and interleukin-1 (IL-1), to amplify the inflammatory response .

Clinical Significance

IL-17A is recognized as a hallmark molecule of Th17 cells and plays a pivotal role in various infectious diseases, inflammatory and autoimmune disorders, and cancer . Recent studies have indicated that IL-17A is a biomarker and a therapeutic target in sepsis . Dysregulated expression of IL-17 has been associated with several autoimmune disorders, such as psoriasis .

Recombinant IL-17

Recombinant IL-17 refers to the IL-17 protein that is produced using recombinant DNA technology. This technology allows for the production of large quantities of IL-17 for research and therapeutic purposes. Recombinant IL-17 is used in various studies to understand its role in the immune response and to develop potential therapies for diseases associated with IL-17 dysregulation .

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.