IL5RA (Interleukin 5 receptor alpha subunit), also known as CD125, is a type I transmembrane protein belonging to the type I cytokine receptor family. It serves as a critical component of the high-affinity IL-5 receptor complex. This receptor complex consists of the alpha chain (IL5RA) that specifically binds to IL-5 and the common beta chain (CD131) that is shared with receptors for IL-3 and GM-CSF .
The IL-5/IL-5RA signaling axis plays crucial roles in:
Differentiation, proliferation, recruitment, and activation of eosinophils
Pathological eosinophilia development
Mechanistically, IL5RA acts by forming a heterodimeric receptor with the CSF2RB subunit and subsequently binding to interleukin-5. In unstimulated conditions, IL5RA interacts constitutively with JAK2. When the heterodimeric receptor is activated, it leads to JAK2 stimulation and subsequent activation of the JAK-STAT pathway, triggering downstream signaling cascades .
IL-5RA expression is predominantly found in:
Eosinophils (highest expression)
Basophils
CD34+ progenitor cells
T helper 2 (TH2) cells
Mast cells
Natural killer T cells (NK T cells)
When designing experiments involving IL5RA:
Select appropriate cell models (TF-1/IL-5Rα cells are commonly used as they express the receptor and can demonstrate IL-5–dependent proliferation)
Consider using primary human eosinophils from patients with eosinophilic conditions for more clinically relevant studies
Account for variable expression levels across different cell types when interpreting results
Include appropriate controls to verify receptor expression in your experimental system
Several types of IL5RA antibodies are available for research applications:
| Antibody Type | Characteristics | Common Applications |
|---|---|---|
| Polyclonal | Recognize multiple epitopes; derived from immunized animals | Western blot, ELISA; broader detection capabilities |
| Monoclonal | Recognize single epitope; high specificity | Western blot, IHC-P, flow cytometry; reproducible results |
| Recombinant | Engineered in vitro; consistent production | All applications; higher batch-to-batch consistency |
| Humanized | Contain human framework regions with murine CDRs | Therapeutic applications, in vivo studies |
| Therapeutic (e.g., benralizumab) | Designed for specific clinical effects | ADCC assays, receptor blocking studies |
Examples include polyclonal antibodies like Proteintech's 12655-1-AP and recombinant monoclonal antibodies like Abcam's EPR5450(2) and CAL40 .
ADCC is a key mechanism by which therapeutic anti-IL5RA antibodies like benralizumab deplete eosinophils. The process occurs through these steps:
The Fc portion of anti-IL5RA antibodies binds to FcγRIIIa receptors on natural killer (NK) cells
This binding triggers NK cells to release cytotoxic granules containing perforin and granzymes
These cytotoxic molecules induce apoptosis in the target cells (eosinophils and basophils expressing IL5RA)
For example, benralizumab induces apoptosis through ADCC, where NK cells target eosinophils and basophils expressing IL5RA and induce their cytotoxic action . Benralizumab has higher affinity to human FcγRIIIa, resulting in enhanced ADCC action compared to other anti-IL-5 monoclonal antibodies .
Engineering approaches can enhance ADCC activity, as demonstrated with the antibody 5R65.7, which showed more potent biological activities than benralizumab analogue in ex vivo assays with peripheral eosinophils from patients with severe eosinophilic asthma (SEA) .
Several methodologies can distinguish epitope binding differences:
Domain-swapped variants: Creating domain-swapped IL-5Rα extracellular-domain variants where human IL-5Rα domains (D1, D2, D3) are replaced with corresponding murine IL-5Rα sequences. For example, research showed benralizumab binds to human D1 (hD1), while antibody 5R65.7 recognizes epitopes within human D3 (hD3) .
Yeast Surface Display (YSD) system: This facilitates expression of stable functional protein domains and allows mapping of antibody-binding regions to individual variants .
Competition assays: These determine if different antibodies compete for the same binding site. In one study, a competition ELISA revealed that affinity-matured antibodies blocked the binding of soluble IL-5Rα to human IL-5 with different IC50 values .
Crystallography/structural analysis: While not explicitly mentioned in the search results, this technique can provide atomic-level insights into antibody-antigen interactions.
Engineering improved anti-IL5RA antibodies involves several sophisticated techniques:
Humanization of murine antibodies:
Affinity maturation using yeast surface display:
Fc engineering for enhanced ADCC:
Modify the Fc region to increase binding to FcγRIIIa receptors on NK cells
Optimize glycosylation patterns to enhance ADCC activity
A successful example is antibody 5R65.7, which achieved stronger affinity (KD ≈ 4.64 nM) than a benralizumab analogue (KD ≈ 26.8 nM) and demonstrated improved neutralizing activity toward IL-5–dependent cell proliferation .
Proper validation of IL5RA antibody specificity requires multiple approaches:
Cross-reactivity testing:
Test against structurally related receptors (IL-1 RI, IL-2 R, IL-3 R, IL-4 R, etc.)
Example: One IL5RA antibody showed less than 5% cross-reactivity with recombinant human IL-1 RII, IL-2 R beta, IL-2 R gamma, and IL-3 R, and less than 1% cross-reactivity with IL-1 RI, IL-4 R, IL-6 R, IL-7 R, IL-9 R, and IL-10 R
Multi-antigen non-specificity ELISA:
Species cross-reactivity assessment:
Positive and negative cell lines:
Use known IL5RA-expressing cells (TF-1/IL-5Rα, eosinophils) and non-expressing controls
Validate with techniques such as flow cytometry or Western blot
For optimal Western blot results with IL5RA antibodies:
Use appropriate cell lysates known to express IL5RA (human platelets, Jurkat cells)
Include positive controls (recombinant IL5RA protein)
SDS-PAGE: Use 12% Tris-HCl polyacrylamide gels
Transfer: Use CN membrane and block with 5% skim milk for at least one hour
Primary antibody: Dilute to manufacturer-recommended concentration (e.g., 1:500-1:1000 for Proteintech 12655-1-AP)
Secondary antibody: Use appropriate HRP-conjugated antibody (e.g., anti-rabbit or anti-human IgG)
Detection: Perform chemiluminescent detection
Observed molecular weights: 70-75 kDa and 43 kDa (may vary due to glycosylation)
When using reduced conditions, expect heavy chain and light chain bands at approximately 50 kDa and 25 kDa, respectively
Several methodologies are available for measuring antibody-antigen interactions:
Bio-layer interferometry:
Using instruments like Octet QKe (ForteBio)
Protocol: Dilute purified antibody to 1 μg/ml in kinetics buffer and immobilize on anti-human IgG Fc capture biosensors
Monitor binding isotherms by exposing sensors to different concentrations of soluble IL-5Rα
Measure association for 300s and dissociation for 600s
Use reference sensors without antigen to account for non-specific binding
Calculate association and dissociation rate constants by fitting to sensorgrams
Competitive ELISA:
Coat plates with IL-5-mFc protein (100 ng/well)
Add various concentrations of anti-IL-5Rα antibodies (0-1 μM) with soluble IL-5Rα (50 nM)
Detect residual binding of sIL-5Rα using HRP-conjugated anti-His antibody
Present data as percentage of sIL-5Rα bound relative to no antibody competition
Calculate IC50 by fitting normalized dose-response data to nonlinear sigmoidal curve
Surface Plasmon Resonance (SPR):
While not explicitly mentioned in the search results, this is another common method for determining binding kinetics
Several functional assays can evaluate IL5RA antibody activity:
Cell proliferation inhibition assay:
Use cell lines stably expressing IL-5Rα (e.g., TF-1/IL-5Rα cells)
Stimulate with recombinant human IL-5 to induce proliferation
Add various concentrations of anti-IL5RA antibodies
Measure cell proliferation and calculate IC50 values
Example: Antibody 5R65 showed comparable antiproliferative activity to benralizumab analogue in this assay
ADCC assay:
Isolate primary human eosinophils as target cells
Use NK cells as effector cells
Co-culture with anti-IL5RA antibodies
Measure target cell depletion or apoptosis
Example: 5R65.7 manifested more potent biological activities than benralizumab analogue in ex vivo assays with peripheral eosinophils from patients with SEA
Receptor binding inhibition:
Test the ability of antibodies to block IL-5 binding to IL-5Rα
Can be measured by competitive binding assays
Important for therapeutic antibodies that aim to block IL-5 signaling
When designing experiments to study IL5RA in eosinophilic diseases:
Patient sample selection:
Include patients with confirmed eosinophilic conditions (e.g., SEA)
Consider disease severity, treatment history, and eosinophil counts
Include appropriate healthy controls
Cell isolation methods:
Experimental controls:
Include isotype controls for antibodies
Use established therapeutic antibodies (e.g., benralizumab) as positive controls
Include unstimulated and IL-5 stimulated conditions
Translational relevance:
Correlate in vitro findings with clinical parameters
Consider ex vivo assays with patient-derived cells
Evaluate potential biomarkers of response
Ethical considerations:
Obtain proper ethical approval for human sample collection
Follow institutional guidelines for patient consent and data protection
By following these systematic approaches, researchers can generate robust and clinically relevant data on IL5RA biology in eosinophilic diseases.
Several technical challenges may arise when working with IL5RA antibodies:
IL5RA can have multiple observed molecular weights (43 kDa, 70-75 kDa) compared to calculated weights (38 kDa, 48 kDa)
Solution: Use deglycosylation enzymes before Western blot; include recombinant protein controls; use antibodies targeting different epitopes
Solution: Enrich for IL5RA-expressing cells; use more sensitive detection methods; consider transfection to increase expression
Solution: Perform thorough validation with known positive and negative controls; use antibodies with demonstrated specificity (<5% cross-reactivity with related receptors)
Solution: Standardize experimental conditions; use multiple donor samples; include positive controls like benralizumab
When faced with contradictory results using different IL5RA antibodies:
Consider epitope differences:
Evaluate antibody formats and properties:
Assess experimental conditions:
Validate with multiple techniques:
Confirm findings using orthogonal methods (e.g., flow cytometry, Western blot, and functional assays)
Use genetic approaches (siRNA knockdown) to confirm specificity
Critical quality control parameters to assess include:
Researchers should request validation data and technical specifications from manufacturers to ensure antibodies meet these quality parameters for their specific applications.
Several emerging technologies may transform IL5RA antibody research:
Single B cell sequencing and antibody discovery:
Enables faster identification of novel anti-IL5RA antibodies with unique properties
Allows mining of the immune repertoire of patients with eosinophilic conditions
CRISPR/Cas9 gene editing:
Creates precise IL5RA knockout or knock-in cell lines for antibody validation
Enables engineering of reporter cell lines with modified IL5RA signaling components
Structural biology advances:
Cryo-EM and X-ray crystallography to determine precise antibody-IL5RA binding interfaces
Structure-guided design of antibodies with improved properties
Bispecific antibody platforms:
Development of bispecific antibodies targeting IL5RA and additional targets
Potential for enhanced therapeutic effects in eosinophilic diseases
Advanced functional screening assays:
High-throughput systems to screen antibodies for specific functional properties
Microfluidic systems for single-cell analysis of antibody effects
Several key questions remain in IL5RA biology that could be addressed with antibody tools:
Receptor complex dynamics:
How does IL5RA interact with the common beta chain in different cellular contexts?
What are the precise conformational changes upon IL-5 binding?
Signaling pathway specificity:
Role in non-eosinophil cells:
Tissue-specific functions:
How does IL5RA function differ between circulating and tissue-resident eosinophils?
Are there tissue-specific cofactors that modify IL5RA signaling?
Therapeutic resistance mechanisms:
Why do some patients with eosinophilic diseases not respond to anti-IL5RA therapy?
Are there compensatory pathways that bypass IL-5/IL5RA signaling?