IL17RA activates MAPK (ERK, p38), NF-κB, and JNK pathways upon IL-17 binding, driving inflammation and tumor progression . The recombinant partial protein mimics native receptor behavior:
In vitro: Stimulates ERK and p38 phosphorylation in transformed intestinal organoids .
Binding kinetics:
The recombinant protein is produced under stringent conditions:
Expression: Codon-optimized vectors in mammalian systems ensure proper folding .
Purification: Affinity chromatography (e.g., Protein A for Fc-tagged proteins) .
Storage: Lyophilized at -80°C; reconstituted in PBS for short-term use .
IL17RA is a therapeutic target in autoimmune diseases and cancers. Preclinical studies highlight:
IL-17RA possesses a complex extracellular domain (ECD) architecture that critically influences its ligand binding properties. The ECD consists of multiple fibronectin-type III-like domains organized in a specific spatial arrangement. Crystal structure analysis of human IL-17RA in the unliganded state reveals a conserved dimerization interface that facilitates receptor-receptor interactions even in the absence of ligand binding. This pre-existing dimerization interface becomes particularly important when IL-17 cytokines bind to the receptor . The ECD can be subdivided into domains D1-D2 that directly interact with IL-17 cytokines, while the D3-D4 domains serve as structural elements that position the receptor properly relative to the cell membrane .
Upon ligand binding, IL-17RA undergoes significant conformational changes, including an "elbow motion" that allows optimal positioning of the D2 domain for interactions with IL-17 family members. The C-terminal tail of IL-17 cytokines adopts an extended conformation when binding to IL-17RA, forming β-strand/β-strand interactions with the D2 domain of the receptor . This flexibility in both the receptor and ligand structures ensures optimal interaction surfaces between IL-17RA and its various ligands.
IL-17RA serves as a shared co-receptor for several IL-17 family members, including IL-17A, IL-17C, IL-17E, IL-17F, and the IL-17A/F heterodimer . Signaling through IL-17RA requires the formation of heteromeric receptor complexes with specific partner receptors depending on the cytokine:
IL-17A, IL-17F, and IL-17A/F heterodimer: IL-17RA pairs with IL-17RC
IL-17E: IL-17RA pairs with IL-17RB
IL-17C: IL-17RA pairs with IL-17RE
The structural arrangement of these receptor complexes determines signaling specificity. For example, IL-17A binding induces IL-17RA dimerization, which then drives the formation of a 2:2:2 hexameric signaling assembly (signalosome) with IL-17RC . This hexameric complex has a specific geometry where the juxtamembrane regions of the two IL-17RC subunits are positioned approximately 90Å apart, which is crucial for proper intracellular signal transduction .
An interesting species-specific interaction has been documented: human IL-17RA does not appear to form productive complexes with mouse IL-17RC, highlighting the importance of considering species compatibility in experimental design .
IL-17RA signaling produces distinct cellular responses depending on cell type and microenvironment:
In keratinocytes:
Potentiates expression of IL-36γ and CXCL1 mRNA
Synergizes with TNF-α to induce inflammatory mediators
In mesenchymal stem cells (MSCs):
Regulates immunosuppressive properties affecting Th17 cell proliferation and differentiation
Controls expression of VCAM1, ICAM1, and PD-L1, which mediate MSC interactions with immune cells
Influences MSC therapeutic potential in experimental autoimmune encephalomyelitis (EAE)
In multiple cell types:
Promotes T cell activation
Induces production of IL-6, G-CSF, and SCF
Stimulates production of pro-inflammatory chemokines
Enhances neutrophil generation and recruitment to inflammation sites
These diverse effects make IL-17RA a central player in both protective immunity and pathological inflammation.
The study of IL-17RA dimerization requires specialized approaches to capture both structural arrangements and functional consequences:
Structural methods:
X-ray crystallography has successfully revealed the dimerization interface of IL-17RA, showing critical residues like alanine 104 located within the α-helical region of the BC loop at the heart of the IL-17RA-IL-17RA protein-protein interface
Solution-based biophysical techniques including size-exclusion chromatography, analytical ultracentrifugation, and multi-angle light scattering can confirm the dimerization status of recombinant IL-17RA
FRET/BRET (Förster/Bioluminescence Resonance Energy Transfer) assays using differentially tagged IL-17RA constructs can monitor dimerization in living cells
Functional approaches:
Site-directed mutagenesis targeting key residues at the dimerization interface (e.g., alanine 104) can generate dimerization-defective variants for comparative studies
Co-immunoprecipitation experiments using differently tagged versions of IL-17RA can biochemically detect receptor oligomerization
Signaling assays comparing wild-type IL-17RA and dimerization-defective mutants can reveal the functional importance of dimerization in downstream effects
When studying IL-17RA dimerization, researchers should consider that IL-17RA exists as a multimer on the cell surface even without ligand binding, suggesting pre-formed receptor complexes may be important for signaling initiation .
Successful expression and purification of functional recombinant IL-17RA requires careful consideration of several factors:
Expression systems:
Mammalian expression systems (HEK293, CHO cells) are preferred for human IL-17RA as they provide appropriate post-translational modifications and protein folding machinery
For the extracellular domain only, insect cell expression (Sf9, Hi5) can offer higher yields while maintaining proper folding
Bacterial systems generally yield improperly folded IL-17RA due to the complexity of its structure and need for glycosylation
Construct design:
Including fusion tags (Fc, His, Avi-tag) can facilitate purification and detection
For structural studies, it's often advantageous to express only the extracellular domain (ECD)
The Avi-tag approach allows precise biotinylation at a single site, ensuring uniform protein orientation when bound to streptavidin-coated surfaces without interfering with bioactivity
Purification strategy:
Multi-step purification typically includes affinity chromatography (based on the fusion tag), followed by ion-exchange and size-exclusion chromatography
Buffer optimization is crucial for maintaining stability of the purified protein
For crystallography, additional steps to remove flexible regions or heterogeneity may be necessary
Quality control:
Biophysical techniques (circular dichroism, thermal shift assays) should be employed to confirm proper folding
Functional binding assays with IL-17 family cytokines verify biological activity
Mass spectrometry can confirm protein identity and assess post-translational modifications
Several complementary approaches can robustly measure IL-17RA-dependent cellular responses:
Signaling activation:
Phospho-specific Western blotting to detect activation of downstream signaling molecules (e.g., NF-κB pathway components)
Reporter gene assays using promoters of IL-17RA-responsive genes driving luciferase expression
Flow cytometry-based phospho-flow analysis for single-cell resolution of signaling events
Gene expression:
Quantitative PCR for measuring induction of known downstream genes like IL-36γ and CXCL1, which are potentiated by IL-17RA dimerization
RNA-seq to comprehensively profile the IL-17RA-dependent transcriptome
ELISA or multiplex cytokine assays to quantify secreted proteins induced by IL-17RA signaling
Cell-type specific functional assays:
In keratinocytes: antimicrobial peptide production, barrier function assays
In MSCs: T-cell proliferation suppression assays, analysis of immunoregulatory molecule expression (VCAM1, ICAM1, PD-L1)
In immune cells: cytokine production profiles, cell differentiation and activation markers
For rigorous analysis, using IL-17RA knockout cells reconstituted with either wild-type or dimerization-defective IL-17RA provides a clean system to assess specific receptor functions. This approach has successfully demonstrated that IL-17RA dimerization lowers the threshold for IL-17-induced expression of downstream effector molecules .
IL-17RA dimerization represents a critical molecular switch that precisely controls signal transduction through several mechanisms:
Spatial organization of signaling components:
IL-17-induced IL-17RA dimerization drives formation of a 2:2:2 hexameric signalosome with IL-17RC
This architectural arrangement positions the juxtamembrane regions of IL-17RC subunits approximately 90Å apart
The precise geometry of the complex determines the spatial organization of intracellular signaling components
Signal amplification:
Dimerization of IL-17RA creates an optimal platform for recruiting adapter proteins and signaling enzymes
The formation of the complete hexameric complex with IL-17RC enhances signaling efficiency
Studies with dimerization-defective IL-17RA mutants show reduced expression of downstream targets like IL-36γ and CXCL1, indicating that dimerization amplifies signal strength
Pathway specificity:
Different ligand-induced conformational changes in the IL-17RA dimer may differentially affect recruitment of various adaptor proteins
The specific architecture of the IL-17RA-containing complex may determine which downstream pathways are preferentially activated
IL-17RA dimerization may create unique binding surfaces for specific intracellular signaling molecules
Studies using dimerization-defective IL-17RA variants have revealed that while IL-17RC appears to play a key structural role in driving the spatial organization of intracellular signaling components, IL-17RA dimerization plays a more indirect but equally important role in defining the precise geometry of the signaling complex .
Genetic variations in IL-17RA can substantially alter receptor function through multiple mechanisms:
Structural impacts:
Mutations in the dimerization interface (such as at alanine 104) can disrupt receptor dimerization, reducing signaling capacity
Variants in ligand-binding domains may alter affinity for specific IL-17 family members
Mutations in intracellular domains can affect recruitment of signaling adaptors
Physiological consequences:
IL-17RA deficiency in MSCs significantly impairs their immunosuppressive function and therapeutic potential in EAE, demonstrating the receptor's importance in stem cell-mediated immunomodulation
Complete loss of IL-17RA function compromises host defense against certain microbial infections
IL-17RA mutations may contribute to autoimmune disease susceptibility or protection
Experimental applications:
Dimerization-defective IL-17RA variants serve as valuable tools to study the specific role of receptor dimerization in signaling
Domain-specific mutations help identify critical regions for different IL-17RA functions
Studying naturally occurring IL-17RA variants can reveal novel aspects of receptor biology
When designing IL-17RA mutants for research, several considerations are important:
The location of the mutation relative to functional domains
Whether the mutation affects only one function (e.g., dimerization) or has pleiotropic effects
The potential impact on protein stability and expression
Recombinant IL-17RA serves as both a research tool and template for therapeutic development:
As a research tool:
Soluble IL-17RA-Fc fusion proteins can act as cytokine traps in experimental systems
Structure-based design of IL-17RA variants with altered binding properties can help dissect pathway-specific effects
Avi-tagged IL-17RA allows uniform orientation on streptavidin surfaces for binding studies and drug screening
Therapeutic development strategies:
Decoy receptors based on IL-17RA extracellular domain can neutralize IL-17 family cytokines
Small molecule inhibitors targeting the IL-17RA dimerization interface could prevent signalosome formation
Monoclonal antibodies against specific epitopes of IL-17RA can block particular ligand interactions while preserving others
Experimental models for testing interventions:
IL-17RA knockout cell lines reconstituted with various receptor constructs provide clean systems for evaluating targeted therapeutics
EAE animal models are valuable for testing IL-17RA-targeting strategies in neuroinflammation
Human keratinocyte models can assess effects on skin inflammation pathways
When developing IL-17RA-targeting therapeutics, it's important to consider the receptor's involvement in both pathological inflammation and beneficial host defense, necessitating careful specificity in intervention approaches.
IL-17RA signaling exhibits complex interactions with other inflammatory pathways, creating integrated networks that shape immune responses:
Synergy with TNF-α:
IL-17A and IL-17F synergize with TNF-α in inducing CXCL1, G-CSF, and IL-6
This synergistic effect requires both TNF receptor I and TNF receptor II
The molecular mechanisms involve cooperative effects on transcription factor recruitment and chromatin remodeling
Inhibitory interactions:
IL-17/IL-17RA interactions can inhibit TNF-α-induced upregulation of fibroblast CCL5 and VCAM-1
Different IL-17RA-dependent responses show differential sensitivity to blocking antibodies, suggesting divergent intracellular signaling pathways
Effects on MSC immunomodulation:
The IL-17/IL-17RA axis is critical for MSC "licensing" - the process that enhances their immunosuppressive properties
IL-17RA-dependent signals in MSCs regulate their ability to inhibit pathogenic Th17 cells and promote regulatory T cell generation
Pre-treatment of MSCs with IL-17A enhances their therapeutic effect in EAE, demonstrating practical applications of this pathway interaction
These complex interactions highlight the context-dependent nature of IL-17RA signaling and suggest that targeted modulation could have pathway-specific effects.
IL-17RA has been implicated in various disease models with significant therapeutic implications:
In autoimmune diseases:
In the EAE model of multiple sclerosis, wild-type MSCs significantly reduced disease severity, while IL-17RA-deficient MSCs worsened disease progression
IL-17RA expression on MSCs was required for reducing Th17 cell frequency in draining lymph nodes and for generating CD4+CD25+Foxp3+ regulatory T cells
IL-17RA-dependent signals are critical for MSC immunosuppressive functions in autoimmune contexts
In inflammatory conditions:
IL-17RA activity contributes to neutrophil generation and recruitment to inflammation sites
The receptor is required for host defense against microbial infections
IL-17RA signaling influences the progression of arthritis from inflammation to destructive joint erosion
Therapeutic implications:
Enhancement of IL-17RA signaling in MSCs could potentially improve their therapeutic efficacy in multiple sclerosis treatment
Modulation of the IL-17/IL-17RA axis represents a promising approach for controlling inflammatory diseases
Targeted interventions affecting specific IL-17RA-dependent pathways could provide selective immunomodulation
These findings highlight the dual role of IL-17RA in both pathological and beneficial immune responses, necessitating careful consideration in therapeutic targeting.