IL5RA (Interleukin-5 Receptor Alpha) is a critical subunit of the IL-5 receptor complex, essential for eosinophil survival, differentiation, and chemotaxis . Mouse models involving IL5RA have been instrumental in studying its role in immunology, oncology, and inflammatory diseases. These models include recombinant IL5RA proteins (engineered for research applications) and transgenic or knockout mice designed to mimic human pathologies or study receptor function.
Recombinant mouse IL5RA proteins are widely used as tools in bioassays, signaling studies, and therapeutic antibody development. Below are key specifications and applications:
These proteins enable precise studies of IL5RA interactions with IL-5 and downstream signaling components like JAK2 and CSF2RB .
A humanized B-hIL5/hIL5RA mouse model was developed to evaluate human IL-5/IL5RA-targeted therapies (e.g., mepolizumab, benralizumab) . Key features:
Validation: Human IL-5 administration increases eosinophil counts in blood and tissues.
Therapeutic Testing: Demonstrated efficacy in reducing eosinophils in asthma-like models .
Mice with disrupted Il5ra genes exhibit:
Normal Phenotype: No major developmental defects.
Immune Deficiencies: Reduced eosinophil responses to IL-5, impaired B-cell differentiation, and altered basophil function .
Unexpectedly, IL5RA is expressed on neutrophils in mice and humans, particularly in inflamed lungs :
Observation | Implication |
---|---|
Neutrophil IL5RA in Inflamed Lungs | IL-5 may modulate neutrophil ROS production and survival . |
Human BALF Neutrophils | IL5RA expression linked to severe asthma . |
Antibody Engineering: Murine antibodies against human IL5RA (e.g., 5R65.7) demonstrate high affinity and neutralizing capacity .
Immunogenic Cell Death (ICD): IL5RA expression in myeloma is associated with ICD-related genes, suggesting its role in immunotherapy response .
IL5RA interacts with multiple signaling partners in mice:
IL5RA, also known as interleukin 5 receptor alpha or CD125, is a specific subunit of a heterodimeric cytokine receptor in mice. It functions by pairing with a signal transducing beta subunit that is shared with IL-3 and GM-CSF receptors to form a functional IL-5 receptor complex. In mice, IL5RA is predominantly expressed on eosinophils and basophils, serving as a cell surface receptor that plays crucial roles in the survival, differentiation, and chemotaxis of eosinophils .
The IL5RA protein is a membrane-localized receptor with a canonical length of 420 amino acid residues and a mass of approximately 47.7 kDa, subject to post-translational modifications including glycosylation . As a member of the Type I cytokine receptor family, IL5RA functions as a marker for identifying eosinophils, basophils, and mast cells in research contexts . Recent studies have also revealed its importance in B1 cell development and maintenance, suggesting broader immunological functions beyond eosinophil regulation .
IL5RA expression in mice shows distinct tissue and cell-type specificity patterns:
Hematopoietic tissues: Highest expression occurs in bone marrow (particularly in eosinophil progenitors) and specific immune cell subsets in peripheral blood and spleen
Immune cells: Predominant expression on eosinophils and basophils, with additional expression on certain B cell populations, particularly B1 cells
Brain tissue: Differential expression across brain regions according to Allen Brain Atlas data, with specific regional patterns
Cell clusters: Single-cell RNA sequencing has identified specific B cell clusters (clusters 0 and 2) with enriched IL5RA expression in mice
Recent research using transgenic IL5ra reporter mouse models has enabled more precise tracking of IL5RA expression patterns in vivo . Interestingly, some studies have found that certain cell populations detectible with anti-IL5RA antibodies do not express the IL5ra reporter transgene, suggesting potential differences in protein isoform expression or detection sensitivity across methodologies .
IL5RA signaling in mice regulates multiple important cellular processes through several interconnected pathways:
Eosinophil development and function: Controls terminal differentiation of eosinophils from progenitor cells, mediates survival signals, and regulates eosinophil activation
B1 cell maintenance: Supports the development and function of B1 cells through regulation of specific gene sets
Downstream gene regulation: Activates expression of target genes including Ass1, Anxa2, Ccnd1, and Zcwpw1 in B1 cells as demonstrated by qPCR studies
JAK/STAT signaling: Primarily signals through JAK2 and STAT5 activation
Tissue protection mechanisms: Recent studies indicate IL5RA-dependent eosinophils provide hepatoprotective functions during liver ischemia-reperfusion injury
Alarmin response integration: Functions downstream of alarmin-initiated pathways, as IL-33 signaling triggers IL-5 production by ILC2s, which then acts through IL5RA
The complex signaling network associated with IL5RA positions it as a critical mediator between environmental signals and cellular responses in both homeostatic and inflammatory conditions.
Multiple complementary techniques can be employed to detect IL5RA expression in mouse tissues, each with specific advantages:
Most widely used for quantitative single-cell analysis
Requires validated anti-IL5RA (CD125) antibodies
Allows for multiparameter analysis and sorting of IL5RA-positive cells
Optimal for analyzing expression across heterogeneous cell populations
Real-time quantitative PCR with validated primers for mouse Il5ra
PrimePCR assays are available specifically designed following MIQE guidelines
Allows precise quantification of transcript levels across experimental conditions
Can detect multiple transcript variants
IL5RA reporter mice express fluorescent proteins under Il5ra promoter control
Enable direct visualization and isolation of IL5RA-expressing cells
Particularly valuable for developmental studies and lineage tracing
Can reveal regulatory elements controlling Il5ra expression
Provides comprehensive transcriptional profiling at single-cell resolution
Has identified previously unknown IL5RA-expressing cell populations
Enables correlation of IL5RA expression with global gene expression patterns
Recent studies used scRNA-seq to identify IL5RA enrichment in specific B cell clusters
When selecting methods, researchers should consider that different detection approaches may yield slightly different results. For example, studies have observed populations detectable with anti-IL5RA antibodies that do not express IL5ra reporter transgenes , suggesting the importance of validating findings across multiple detection platforms.
Assessing IL5RA functionality requires approaches that evaluate receptor-mediated signaling and biological outcomes:
Measures phosphorylation of downstream molecules (JAK2, STAT5)
Provides single-cell resolution of pathway activation
Can be performed ex vivo after IL-5 stimulation
Directly links receptor expression to signaling activity
Compare phenotypes between wild-type and Il5ra-deficient mice
Use conditional knockout models to assess tissue-specific functions
Employ IL5RA reporter mice to track receptor-expressing cells
Recent studies have shown that IL5RA-deficient mice exhibit exacerbated liver injury, demonstrating a protective role for IL5RA signaling
Measure eosinophil numbers in blood and tissues
Assess IL-5-responsive gene expression (Ass1, Anxa2, Ccnd1, Zcwpw1)
Evaluate B1 cell frequencies and function
Compare tissue pathology in disease models (e.g., liver IR injury models have revealed protective functions of IL5RA-expressing eosinophils)
Anti-IL-5 or anti-IL5RA neutralizing antibodies to block signaling
Recombinant IL-5 administration to enhance signaling
Comparison of effects across different genetic backgrounds
Studies have shown that recombinant IL-13 administration can reduce hepatic IR injury, suggesting downstream effects of IL5RA signaling pathways
A comprehensive assessment of IL5RA functionality should combine multiple approaches to establish both the signaling capacity of the receptor and its biological consequences in relevant cell populations and disease contexts.
Selecting appropriate anti-IL5RA antibodies requires careful consideration of several factors:
Use IL5ra knockout mice as negative controls
Compare multiple antibody clones in parallel
Validate with orthogonal methods (e.g., reporter mice, mRNA expression)
Test across relevant tissue types and experimental conditions
Flow cytometry applications require antibodies recognizing extracellular domains
Western blot applications typically require antibodies against denatured epitopes
Functional studies need antibodies that either block or don't interfere with IL-5 binding
Multiple conjugates (FITC, biotin, HRP, Alexa dyes) are available for different applications
Different clones may have varying abilities to detect post-translationally modified IL5RA
Some clones may recognize specific isoforms of IL5RA (up to 5 different isoforms have been reported)
Epitope accessibility can differ in various fixation and permeabilization protocols
Consider species cross-reactivity if comparing mouse and human systems
Flow cytometry: Pre-conjugated antibodies with optimized fluorophores
Immunohistochemistry: Antibodies validated for paraffin-embedded tissues
Western blotting: Antibodies optimized for denatured protein detection
Functional blocking: Antibodies that interfere with IL-5 binding
Research has revealed that some IL5RA-expressing populations may be detectable with certain monoclonal antibodies but not with IL5RA reporter transgenes , suggesting heterogeneity in epitope expression or differences in detection sensitivity. Researchers should carefully validate antibodies for their specific application and experimental system.
Several engineered mouse models have been developed to study IL5RA biology, each with specific research applications:
Complete deletion of functional IL5RA expression
Show impaired eosinophil development and function
Exhibit increased susceptibility to certain tissue injuries
Studies have demonstrated exacerbated liver ischemia-reperfusion injury in these mice
Express fluorescent proteins under Il5ra promoter control
Allow direct visualization and tracking of IL5RA-expressing cells
Enable isolation of IL5RA-expressing populations by FACS
Recent studies report development of new IL5RA reporter mouse strains that have expanded research capabilities
Allow cell-type specific deletion when crossed with appropriate Cre lines
Examples include eosinophil-specific knockout models
Particularly valuable for distinguishing direct from indirect effects
Can be combined with inducible systems for temporal control
IL5RA models can be crossed with other relevant gene knockouts
Studies have utilized mice with eosinophil-specific deletion of IL-4/IL-13 (IL-4/IL-13ΔEOS) to examine downstream effector mechanisms
Combinations with ST2−/− or IL-33−/− models have revealed upstream regulators
ILC2-deficient models (Nmur1iCre-eGFP Id2fl/fl) have demonstrated the role of ILC2s in maintaining IL5RA+ B1 cells
Each model provides distinct advantages for investigating specific aspects of IL5RA biology. Selection should be based on the particular research question, with consideration of potential developmental effects, compensation mechanisms, and genetic background influences.
IL5RA deficiency leads to several distinctive phenotypes across different disease models:
Profound reduction in eosinophil numbers in blood, bone marrow, and tissues
Impaired eosinophil recruitment to inflammatory sites
Reduced eosinophil development from progenitors
Altered tissue distribution patterns
Exacerbated liver ischemia-reperfusion injury
Increased serum ALT and AST levels indicating greater hepatocyte damage
Reduced numbers of IL5RA+ B1 cells
Downregulation of IL-5 target genes (Ass1, Anxa2, Ccnd1, Zcwpw1) in B cells
Altered antibody production profiles
These findings suggest IL5RA signaling is important for B1 cell maintenance
Modified responses to parasitic infections
Altered allergic inflammation
Changes in ILC2-dependent immune pathways
Modified mucosal immune responses
The diverse phenotypes observed in IL5RA-deficient mice highlight the complex roles of this receptor across multiple immune processes and disease contexts. These findings have expanded our understanding of IL5RA beyond its canonical role in eosinophil development to include tissue protection and B cell regulation.
IL5RA reporter mice present unique methodological considerations that researchers should address:
Essential to confirm correlation between reporter expression and endogenous IL5RA protein
Validate across multiple tissues and cell types
Test under different activation conditions
Compare with anti-IL5RA antibody staining
Determine optimal flow cytometry parameters for reporter detection
Establish imaging protocols that preserve reporter fluorescence
Consider tissue autofluorescence and implement appropriate controls
Optimize fixation methods that maintain reporter signal
Distinguish between current and historical expression (reporter protein may persist after transcription ceases)
Consider threshold detection differences between reporter and endogenous protein
Interpret quantitative data in the context of reporter sensitivity
Be cautious about extrapolating from reporter brightness to expression level
Include appropriate wild-type controls lacking the reporter
Consider using heterozygous reporter mice if homozygosity affects function
Account for potential developmental effects of the reporter construct
Use reporter expression in conjunction with other markers for comprehensive analysis
Addressing these challenges requires careful experimental design and appropriate controls to maximize the utility of IL5RA reporter mice as research tools.
IL5RA signaling operates within complex cytokine networks with several key interactions:
Research demonstrates that IL-33 signaling through ST2 induces IL-5 production by ILC2s
IL-33−/− and ST2−/− mice show impaired eosinophil accumulation in tissues
This establishes a sequential pathway: IL-33 → ILC2 activation → IL-5 production → IL5RA signaling
In liver ischemia-reperfusion models, this pathway is critical for eosinophil-mediated tissue protection
IL5RA signaling functions within the broader Type 2 cytokine network (IL-4, IL-13, TSLP)
IL-13 administration to wild-type mice reduces hepatic IR injury
Mice with eosinophil-specific deletion of IL-4/IL-13 show increased tissue injury
This suggests IL5RA-dependent eosinophils mediate protection partly through IL-4/IL-13 production
Group 2 innate lymphoid cells (ILC2s) are a critical non-redundant source of IL-5
ILC2-deficient mice show reduced IL5RA+ B1 cells and downregulation of IL-5 target genes in B1 cells
This positions ILC2s as essential upstream regulators of IL5RA-dependent processes
Alarmin-responsive ILC2s link tissue damage to IL5RA signaling through IL-5 production
IL5RA uses a common beta chain shared with IL-3 and GM-CSF receptors
This creates potential for competitive binding and coordinated regulation
Understanding this shared signaling machinery helps explain redundancies and specificities
The beta subunit is required for the biological activities of IL-5
IL5RA signaling induces specific gene expression programs
These may overlap with or diverge from programs induced by other cytokines
qPCR analysis has identified specific IL-5 target genes downregulated in IL-5-deficient conditions
This transcriptional integration determines the ultimate cellular responses
Understanding these interactions requires sophisticated experimental approaches, including conditional deletion models, temporal control of cytokine availability, and systems biology approaches to model network behavior.
Research has revealed significant roles for IL5RA in B cell biology beyond its canonical function in eosinophils:
IL5RA expression is enriched in specific B cell subsets, particularly B1 cells
Single-cell RNA sequencing has identified specific B cell clusters (clusters 0 and 2) with enriched IL5RA expression
IL5RA+ B1 cells are significantly reduced in ILC2-deficient mice
This indicates ILC2-derived IL-5 is essential for maintaining IL5RA+ B1 cells
Multiple IL-5-dependent genes are downregulated in B1 cells from ILC2-deficient mice
These genes (Ass1, Anxa2, Ccnd1, Zcwpw1) are not expressed in B2 cells
qPCR confirmation shows reduced expression of these genes in sort-purified B1 cells from IL-5-deficient mice
This reveals a specific transcriptional program activated by IL5RA signaling in B1 cells
The reduction in IL5RA+ B1 cells and IL-5 target genes in B1 cells from ILC2-deficient mice suggests ILC2s as an essential source of IL-5
This establishes an ILC2-IL-5-IL5RA-B1 cell axis
The axis may be critical for maintaining certain B1 cell populations and their functions
This network connects innate lymphoid cell activation to B1 cell regulation
IL5RA in B cells is thought to signal through similar pathways as in eosinophils
The receptor may interact with syndecan binding protein (syntenin), which is required for IL5-mediated activation of the transcription factor SOX4
Several alternatively spliced transcript variants of IL5RA have been reported , which may have distinct functions in B cells
These signaling mechanisms link IL5RA to specific B cell transcriptional programs
The specific role of IL5RA in B cells represents an important area for further research, particularly in understanding how the IL-5/IL5RA axis connects different branches of the immune system and regulates B cell subpopulations with distinct functions.
Recent research has uncovered unexpected tissue-protective functions of IL5RA-expressing eosinophils:
Eosinophils demonstrate critical hepatoprotective function during liver ischemia-reperfusion (IR) injury
IL5RA-deficient mice develop exacerbated hepatic IR injury with increased serum ALT and AST levels
Histological analysis confirms greater hepatocyte necrosis in these models
This provides compelling evidence for a critical hepatoprotective function of eosinophils
ST2−/− and IL-33−/− mice show impaired eosinophil accumulation in the liver during injury
These mice develop more severe hepatic IR injury similar to IL5RA-deficient mice
This establishes the IL-33/ST2 axis as an upstream regulator of eosinophil recruitment
The pathway connects tissue damage (IL-33 release) to protective eosinophil responses
IL-13 administration to wild-type mice reduces hepatic IR injury
Mice with eosinophil-specific deletion of IL-4/IL-13 show increased hepatic IR injury
This indicates eosinophil-derived IL-4/IL-13 contribute to tissue protection
IL5RA signaling may thus regulate tissue protection partly through modulating eosinophil cytokine production
IL5RA-deficient mice show increased neutrophil accumulation in injured tissues
This suggests eosinophils may limit neutrophil-mediated tissue damage
The mechanism may involve direct inhibition or competitive recruitment
Neutrophil-eosinophil cross-regulation represents an important area for further study
These findings challenge traditional views of eosinophils as primarily pathogenic cells and highlight the context-dependent nature of IL5RA signaling in tissue homeostasis and injury. The protective role of eosinophils has important implications for therapeutic approaches targeting the IL-5/IL5RA pathway in various disease contexts.
Experimental variability in IL5RA detection requires systematic approaches to ensure reliable results:
Develop consistent protocols for sample preparation, antibody staining, and analysis
Document detailed methods including buffer compositions, incubation times, and temperatures
Maintain consistent instrument settings for flow cytometry and imaging
Use standardized positive and negative controls across experiments
Compare results from multiple detection methods (antibodies, reporter mice, PCR)
Research has shown that some cell populations detectible with anti-IL5RA antibodies do not express IL5ra reporter transgenes
These discrepancies may reflect biological differences or technical limitations
Use multiple complementary approaches for comprehensive analysis
Validate antibodies using IL5RA knockout tissues as negative controls
Compare multiple antibody clones and conjugates
Test under different fixation and permeabilization conditions
Consider the detection of specific IL5RA isoforms (up to 5 different isoforms have been reported)
Use validated primer sets designed following MIQE guidelines
Consider primer design to detect all relevant transcript variants
Include appropriate housekeeping genes for normalization
Implement standard curves to ensure quantitative accuracy
Include biological and technical replicates
Blind analysis when possible to reduce observer bias
Use appropriate statistical methods for analysis
Power calculations to determine adequate sample sizes
By systematically addressing these factors, researchers can improve the reliability and reproducibility of IL5RA detection across different experimental conditions and research settings.
Discrepancies between protein and transcript measurements are common in IL5RA research and require specific resolution strategies:
Protein expression typically lags behind transcript expression
Design time-course experiments to capture both mRNA and protein dynamics
Consider protein half-life and mRNA stability
Account for potential delays between transcription and translation
Multiple IL5RA transcript variants and protein isoforms exist
Ensure detection methods capture all relevant isoforms
Use isoform-specific primers or antibodies when appropriate
Consider alternative splicing regulation in different cell types
Investigate microRNA-mediated regulation of IL5RA translation
Assess RNA-binding proteins that may affect translation efficiency
Consider nonsense-mediated decay of certain transcript variants
Evaluate translational efficiency in different cellular contexts
IL5RA undergoes glycosylation and potentially other modifications
These modifications may affect antibody binding and protein function
Consider using methods to detect specific modifications
Evaluate modification patterns across different cell types
Combine RNA-seq with proteomics data
Use single-cell multi-omics where feasible
Apply computational methods to integrate data types
Develop models that account for regulatory relationships
By systematically investigating these factors, researchers can better understand the relationship between IL5RA transcript and protein levels, improving interpretation of experimental results and resolving apparent discrepancies between different measurement approaches.
Determining causality in complex systems requires specialized experimental approaches:
Use IL5RA conditional knockout mice crossed with cell-type specific Cre lines
Compare phenotypes between global and conditional knockouts
This approach has been used with eosinophil-specific deletion of IL-4/IL-13 to examine downstream mediators
Target multiple cell types individually to dissect complex phenotypes
Create mixed bone marrow chimeras with wild-type and IL5RA-deficient cells
Allows identification of cell-intrinsic versus environmental effects
Can reveal non-hematopoietic contributions to phenotypes
Enables assessment of competitive fitness of IL5RA-sufficient versus deficient populations
Isolate specific cell populations to test direct IL-5 responsiveness
Compare responses between cells from wild-type and IL5RA-deficient mice
Use pharmacological inhibitors with appropriate controls
Recent studies have used sort-purified cells for qPCR confirmation of IL-5 target genes
Use neutralizing antibodies at different disease stages
Compare prophylactic versus therapeutic intervention
Studies have shown protective effects of recombinant IL-13 administration in liver injury models
Monitor disease kinetics to identify primary versus secondary effects
Apply single-cell transcriptomics to identify direct IL-5 responsive cells
Recent research has used single-cell RNA sequencing to identify IL5RA-enriched B cell clusters
Use computational approaches to infer causal relationships
Integrate multiple data types to build comprehensive models
By combining these approaches, researchers can build strong cases for direct versus indirect effects of IL5RA signaling and develop more precise models of how IL5RA contributes to complex disease phenotypes and protective immune responses.
Interleukin-5 receptor alpha (IL-5Rα) is a critical component in the immune system, particularly in the regulation and function of eosinophils. This receptor is a part of the cytokine receptor family and plays a significant role in various immune responses, including allergic reactions and asthma.
The IL-5 receptor is a heterodimer composed of an alpha (α) and a beta (βc) subunit. The alpha subunit is specific to IL-5, while the beta subunit is shared with receptors for interleukin-3 (IL-3) and granulocyte-macrophage colony-stimulating factor (GM-CSF) . The glycosylation of the Asn 196 residue of the Rα subunit is essential for the binding of IL-5 .
IL-5Rα is primarily involved in the growth, differentiation, and activation of eosinophils. Eosinophils are white blood cells that play a crucial role in the body’s immune response, particularly in combating parasitic infections and contributing to allergic reactions . The receptor’s activation by IL-5 triggers a cascade of intracellular events that lead to the proliferation and survival of eosinophils .
Recombinant IL-5Rα, particularly from mouse models, is extensively used in research to study its function and therapeutic potential. These recombinant proteins are produced using various expression systems and are crucial for in vitro and in vivo studies . They help in understanding the receptor’s role in immune responses and in developing targeted therapies.