IL-17E, also known as IL-25, is a distinct member of the interleukin-17 cytokine family that plays crucial roles in host defense and inflammatory responses. Unlike IL-17A and IL-17F which primarily promote responses against extracellular bacteria and fungi, IL-17E in rats predominantly drives type 2 immune responses associated with parasitic infections and allergic inflammation . IL-17E shares structural homology with other IL-17 family members through five spatially conserved cysteine residues that mediate dimerization, but it has unique receptor binding properties and downstream signaling pathways .
IL-17E signals through a heterodimeric receptor complex consisting of IL-17RA and IL-17RB, whereas other family members utilize different receptor combinations . In rat models, IL-17E expression has been detected in various tissues including the gastrointestinal tract, lymph nodes, and spleen, with particular relevance in immune responses against parasitic infections like Trichuris muris and N. brasiliensis .
In rat models, IL-17E is produced by diverse cell populations including both immune and non-immune cells. Primary sources include specialized subset of T helper cells, mast cells, eosinophils, and epithelial cells at barrier surfaces such as the intestine and respiratory tract . Recent studies have identified a critical population of ckit+lin− cells that respond to IL-17E and are essential for mounting effective Th2 immune responses in rodent models .
Unlike IL-17A which is predominantly produced by Th17 cells, IL-17E production in rats shows a more heterogeneous cellular origin, reflecting its distinct role in coordinating type 2 immune responses. Following stimulation with specific antigens or pathogens, these IL-17E-producing cells are activated and secrete this cytokine to initiate downstream immunological cascades that differ substantially from those triggered by other IL-17 family members .
Several complementary methodologies can be employed for accurate quantification of IL-17E in rat biological samples. Enzyme-linked immunosorbent assay (ELISA) remains the gold standard for measuring IL-17E protein concentration in serum and tissue homogenates, offering good sensitivity and specificity when using validated rat-specific antibodies . Flow cytometry with intracellular cytokine staining provides valuable insights into cellular sources of IL-17E, allowing researchers to identify specific IL-17E-producing cell populations within rat tissues .
For gene expression analysis, quantitative real-time PCR (qRT-PCR) effectively measures IL-17E mRNA levels, though protein measurements should accompany transcriptional data since post-transcriptional regulation may influence final protein levels . Western blotting can confirm IL-17E protein expression in tissue lysates, while immunohistochemistry allows visualization of IL-17E distribution within tissue architecture. For comprehensive analysis, researchers should employ multiple techniques, as studies have shown that measurement methodology can significantly influence detected cytokine levels in rodent models .
For IL-17E neutralization studies in rats, several approaches exist with varying degrees of specificity and efficacy. Administration of anti-IL-17E neutralizing antibodies represents the most direct approach, allowing temporal control of IL-17E blockade without permanently altering the animal's genetics . These antibodies can be delivered systemically or locally depending on the research question, with dosing regimens carefully optimized to maintain effective neutralization throughout the experimental period.
For genetic approaches, CRISPR-Cas9 technology can be employed to generate IL-17E knockout rats, providing a complete absence of functional IL-17E throughout development and adulthood. Alternatively, siRNA or antisense oligonucleotides can be used for transient IL-17E knockdown in specific tissues. For overexpression models, researchers can utilize adenoviral or lentiviral vectors carrying the IL-17E gene under constitutive or inducible promoters, or generate transgenic rat lines expressing IL-17E under tissue-specific promoters, similar to approaches that have shown IL-17E overexpression leads to eosinophilia, neutrophilia, and elevated serum IgE levels in mouse models .
In rat models of neuroinflammatory conditions, IL-17E expression undergoes dynamic changes that vary significantly with disease progression and developmental stage. Studies examining rodent models of autism spectrum disorder (ASD) have revealed that IL-17E levels are generally increased compared to control animals, suggesting a potential role for this cytokine in neuroinflammatory processes underlying neurodevelopmental disorders . The temporal pattern of IL-17E expression appears critical, with significant alterations observed at specific developmental timepoints that may correspond to critical windows in neurodevelopment.
For instance, studies have demonstrated that maternal immune activation (MIA) can trigger elevated IL-17E levels in pregnant rats and their offspring, with these alterations potentially contributing to ASD-like behavioral phenotypes . Importantly, the relationship between IL-17E and neuroinflammation appears to be age-dependent, with one study showing significant alterations only at postnatal day 30, suggesting developmental regulation of this cytokine's expression . Research into experimental autoimmune encephalomyelitis (EAE) models has further indicated that IL-17E may actually exert protective effects in some neuroinflammatory contexts, with administration of IL-17E ameliorating disease severity through suppression of IL-23 production and subsequent inhibition of pathogenic Th17 responses .
IL-17E plays a pivotal role in orchestrating effective immune responses against parasitic infections in rat models, particularly against helminth parasites. Studies have demonstrated that IL-17E is essential for the clearance of intestinal nematodes such as Trichuris muris and Nippostrongylus brasiliensis in rodent models . Mechanistically, IL-17E initiates and amplifies type 2 immune responses characterized by the production of IL-4, IL-5, and IL-13, which collectively promote goblet cell hyperplasia, mucus production, and smooth muscle contractility in the gastrointestinal tract—all critical mechanisms for worm expulsion.
Experimental approaches utilizing genetic deletion of IL-17E or its receptors (IL-17RA and IL-17RB) in rats have confirmed the necessity of this signaling pathway for effective anti-helminth immunity . Upon infection, IL-17E activates specific innate lymphoid cell populations, particularly the recently characterized ckit+lin− cells, which respond to IL-17E stimulation by producing large amounts of type 2 cytokines . Administration of recombinant IL-17E has been shown to accelerate parasite clearance in experimental models, further confirming its therapeutic potential in parasitic diseases. These findings highlight IL-17E as a critical regulator of anti-parasitic immunity in rats, positioning it as a potential therapeutic target for helminthic infections.
The relationship between IL-17E and other inflammatory markers in rat models reveals complex, context-dependent interactions that merit careful experimental analysis. Unlike IL-17A and IL-17F, which typically positively correlate with pro-inflammatory markers like IL-6 and TNF-α, IL-17E often demonstrates inverse correlations with these classical inflammatory mediators . In rat models of inflammatory bowel disease, increased IL-17E levels correlate with decreased expression of IL-12 and IL-23, two cytokines crucial for Th1 and Th17 differentiation respectively, suggesting an immunoregulatory function .
Analysis of correlation data from multiple rodent studies indicates that IL-17E positively correlates with Th2-associated markers including IL-4, IL-5, IL-13, IgE levels, and eosinophil counts . This pattern reflects IL-17E's role in promoting type 2 immune responses rather than classical inflammatory cascades. Interestingly, studies in neuroinflammatory conditions have shown that IL-17E levels correlate with behavioral changes in ASD rat models, suggesting potential links between this cytokine and behavioral phenotypes . When designing correlation studies, researchers should account for tissue-specific expression patterns and temporal dynamics, as IL-17E correlations with other inflammatory markers can change significantly across developmental stages and disease progression .
Investigating IL-17E receptor signaling in rat models requires careful consideration of several critical experimental factors. Foremost, researchers must recognize that IL-17E signals through a heterodimeric receptor complex comprising IL-17RA and IL-17RB, with IL-17RA serving as a common receptor chain for multiple IL-17 family members . This shared receptor usage necessitates careful experimental design to distinguish IL-17E-specific signaling from other IL-17 family member effects.
When designing receptor blockade studies, researchers should consider targeting IL-17RB rather than IL-17RA to achieve IL-17E-specific inhibition, as IL-17RA blockade would affect multiple IL-17 cytokines . Cell-specific receptor expression analysis is essential, as IL-17RB expression patterns vary substantially among different tissues and cell types in rats. Methods for studying receptor signaling should include phosphorylation studies of downstream mediators like ACT1 and analysis of target gene expression .
Receptor desensitization phenomena should be accounted for in chronic stimulation models, as persistent IL-17E exposure may lead to receptor downregulation and altered signaling kinetics . Time-course experiments are crucial since IL-17E receptor signaling dynamics change with developmental stage and disease progression . Finally, researchers should consider potential species differences between rats and mice when translating findings, as subtle variations in receptor distribution and signaling pathways exist between rodent species.
Addressing strain-specific variability in IL-17E expression requires systematic methodological approaches and careful data interpretation. Studies have demonstrated significant baseline differences in IL-17E levels among common laboratory rat strains, similar to the elevated IL-17 concentrations observed in BTBR mice compared to C57BL/6 controls in autism models . This inherent variability necessitates strain-matched controls for all experiments and potentially larger sample sizes to account for increased data dispersion.
For comparative studies, researchers should conduct preliminary experiments to establish strain-specific reference ranges for IL-17E expression in relevant tissues. Mixed-effects statistical models incorporating strain as a random effect can help partition variance attributable to strain differences from experimental treatment effects. When pooling data from multiple studies, meta-analytic approaches should include strain as a moderator variable to account for systematic differences. Additionally, researchers should consider genetic mechanisms underlying strain differences, as polymorphisms in IL-17E or its regulatory elements may contribute to observed variability. Understanding these strain-specific differences is not merely a technical consideration but may provide valuable insights into genetic determinants of IL-17E regulation relevant to human disease heterogeneity.
Analyzing developmental changes in IL-17E expression requires rigorous experimental design and specialized analytical approaches. Longitudinal sampling strategies should be prioritized whenever possible, with careful consideration of sampling intervals based on critical developmental windows in rats—particularly during early postnatal periods when immune system maturation occurs rapidly . When longitudinal sampling is not feasible, cross-sectional designs should include sufficient animals at each timepoint to capture individual variability.
Statistical approaches should employ models specifically designed for longitudinal data, such as repeated measures ANOVA, mixed-effects models, or functional data analysis methods that can accommodate autocorrelation within subjects. Studies have demonstrated that IL-17E expression can change significantly with developmental stage, with some alterations only becoming apparent at specific timepoints such as postnatal day 30 . Age-specific reference ranges should be established for proper interpretation of experimental interventions, and researchers should be cautious about extrapolating findings across developmental stages.
The table below summarizes recommended sampling timepoints for developmental studies of IL-17E in rat models:
Developmental Period | Age Range | Sampling Frequency | Key Biological Processes |
---|---|---|---|
Prenatal | E10-E20 | Daily or every other day | Neurogenesis, organogenesis |
Early postnatal | P0-P14 | Every 2-3 days | Blood-brain barrier formation, microglial maturation |
Juvenile | P15-P28 | Weekly | Synaptic pruning, immune system maturation |
Adolescent | P29-P42 | Weekly | Hormonal changes, behavior development |
Adult | P43+ | Monthly | Stable immunological phenotype |
Translating IL-17E findings from rat models to human applications requires careful consideration of cross-species similarities and differences. While the fundamental biology of IL-17E appears conserved between rats and humans—including receptor composition (IL-17RA/IL-17RB heterodimers), downstream signaling pathways, and its role in type 2 immune responses—important species-specific differences exist . The receptor distribution patterns show subtle variations between species, potentially affecting tissue responsiveness to IL-17E. Additionally, the dynamics of IL-17E production during immune responses may differ in timing and magnitude between rats and humans.
Emerging technologies are revolutionizing our ability to study IL-17E function in rat neural tissues with unprecedented precision. Single-cell RNA sequencing now enables comprehensive mapping of IL-17E and IL-17RB expression across neural cell populations, revealing previously unrecognized cellular targets in the central nervous system . This approach has identified heterogeneous expression patterns of IL-17 receptors among neuronal subtypes, astrocytes, microglia, and oligodendrocytes in rodent models.
Spatially-resolved transcriptomics techniques like Visium and MERFISH allow researchers to analyze IL-17E signaling within the preserved architectural context of neural tissues, providing insights into region-specific effects that may contribute to neuroinflammatory phenotypes . For functional studies, chemogenetic and optogenetic approaches can be combined with IL-17E administration to examine how this cytokine modulates neural circuit activity in behaving animals. CRISPR-Cas9-mediated cell-type-specific deletion of IL-17 receptors in rat neural tissues now enables precise dissection of the cellular targets mediating IL-17E effects on brain function and behavior.
Additionally, new in vitro models such as rat brain organoids provide systems for studying IL-17E effects on neurodevelopmental processes under controlled conditions. As interest in neuroimmune interactions grows, these methodologies offer promising approaches to elucidate IL-17E's roles in neural function and neuroinflammatory conditions, complementing existing research on its contributions to autism spectrum disorder and other neurological conditions .
Interleukin-17E (IL-17E), also known as IL-25, is a member of the IL-17 cytokine family, which includes six structurally related cytokines: IL-17A, IL-17B, IL-17C, IL-17D, IL-17E, and IL-17F . These cytokines play crucial roles in immune regulation and inflammation. IL-17E is unique among its family members due to its ability to promote Th2-biased immune responses, contrasting with the Th1- and Th17-biased inflammation promoted by other IL-17 family members .
IL-17E is primarily involved in promoting Th2-type immune responses. It stimulates the production of cytokines such as IL-4, IL-5, and IL-13, which are associated with allergic reactions and helminth infections . This cytokine also plays a role in enhancing the production of IgE, a key antibody in allergic responses .
IL-17E has been implicated in various diseases, particularly those involving allergic inflammation and asthma. Its ability to promote Th2 responses makes it a potential therapeutic target for conditions characterized by excessive Th2 activity . Additionally, IL-17E has been studied for its role in autoimmune diseases and cancer, although its functions in these contexts are less well understood compared to other IL-17 family members .
Recombinant IL-17E (Rat) is a laboratory-produced version of the natural cytokine, used in research to study its biological functions and potential therapeutic applications. By using recombinant IL-17E, researchers can investigate its effects on immune cells and its role in various disease models, providing valuable insights into its mechanisms of action and potential clinical uses.