IL3RA Human

Interleukin-3 Receptor Subunit Alpha Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure and Characteristics

IL3RA is a single-pass transmembrane protein encoded by the IL3RA gene located on the X-Y pseudoautosomal regions (Xp22.3 and Yp11.3) . Its structural features include:

ParameterDescriptionSource
Molecular WeightCalculated: 34.5 kDa; Observed: 50–60 kDa (due to glycosylation)
Key MotifsWSXWS (critical for folding), Box 1 (JAK interaction/activation)
Expression HostHEK293 cells (commonly used for recombinant protein production)
Subcellular LocationPlasma membrane (type I cytokine receptor family, type 5 subfamily)

The protein’s extracellular domain binds IL-3, while the intracellular domain interacts with signaling molecules like JAK2 and STAT5 .

Functional Roles and Signaling Pathways

IL3RA forms a heterodimer with the β-subunit (IL3RB) to mediate IL-3 signaling, which regulates:

  • Hematopoiesis: Proliferation and differentiation of progenitor cells into lineage-specific cells .

  • Immune Response: Activation of monocytes, B-lymphocytes, and mast cells .

Key Signaling Pathways

PathwayKey ComponentsBiological EffectSource
JAK-STATJAK2 → STAT5 phosphorylationTranscriptional activation of target genes
PI3K/AKTPI3K → AKT activationCell survival and proliferation
MAPKERK1/2 phosphorylationRegulation of apoptosis and differentiation

IL3RA’s signaling is tightly regulated by post-translational modifications, such as ubiquitination-mediated degradation. RNFT2 (a RING finger E3 ligase) targets IL3RA for proteasomal degradation, modulating IL-3-dependent responses .

Regulation of IL3RA Stability

  • RNFT2/IL-3Rα Axis: RNFT2 ubiquitinates IL3RA at Lys357, promoting its degradation. IL-3 stimulation enhances this process, while LPS priming reduces RNFT2 abundance, stabilizing IL3RA and amplifying proinflammatory signaling .

  • Proteasomal Degradation: IL3RA half-life is ~6 hours in murine cells, and degradation is MG132-sensitive (proteasome-dependent) .

Role in Disease Pathogenesis

DiseaseIL3RA’s RoleClinical CorrelationSource
Acute Myeloid Leukemia (AML)Marker for leukemic stem cells; overexpression correlates with tumor loadTarget for therapies (e.g., anti-CD123 antibodies)
SepsisAmplifies cytokine storm via IL-3 signalingNeutralizing IL3RA reduces lung injury
PreeclampsiaElevated placental IL3RA linked to peripartum cardiomyopathy and tachycardiaPredictive biomarker for cardiovascular risks

Biomarker in Hematologic Cancers

  • Leukemic Stem Cells: IL3RA is highly expressed on AML stem cells, making it a therapeutic target. Anti-CD123 antibodies (e.g., CSL362) show promise in clinical trials .

  • Minimal Residual Disease (MRD): IL3RA expression levels monitor treatment response in leukemias .

Placental IL3RA and Cardiovascular Outcomes

  • Severe Preeclampsia (sPE): Elevated placental IL3RA (e.g., 4.5-fold in African ancestry patients) correlates with postpartum cardiomyopathy and tachycardia .

  • Ethnic Variability: African ancestry sPE placentas show higher IL3RA expression compared to Asian/European groups .

Recombinant IL3RA Applications

Recombinant human IL3RA (e.g., HEK293-derived) is used in:

  • Bioassays: Binds IL-3 with high affinity (EC₅₀: 0.32–1.6 μg/mL) .

  • Research Tools: ELISA, receptor binding studies, and drug development .

Genetic and Chromosomal Context

  • Pseudoautosomal Localization: IL3RA and CSF2RA (GM-CSF receptor α) are clustered in the X-Y pseudoautosomal region, enabling dosage compensation .

  • Genetic Variants: Missense mutations in IL3RA have been linked to spinal stenosis, suggesting roles in extracellular matrix maintenance .

Product Specs

Introduction
IL3RA, also known as Interleukin 3 Receptor Subunit Alpha, is a transmembrane protein belonging to the type 1 cytokine receptor family, specifically the type 5 subfamily. This protein serves as a receptor for interleukin 3, a cytokine primarily secreted by activated T cells and mast cells. IL3RA plays a crucial role in the development and function of various blood cells. Notably, its expression is elevated in certain leukemia cells, making it a potential therapeutic target for leukemia treatment.
Description
Recombinant IL3RA protein, expressed in Sf9 insect cells using a baculovirus expression system, is a single polypeptide chain with glycosylation. It encompasses amino acids 20-305 of the human IL3RA protein, with a C-terminal 6-histidine tag. The molecular weight of this recombinant protein is approximately 34.1 kDa. Purification is achieved through proprietary chromatographic techniques.
Physical Appearance
Clear, colorless solution, sterile-filtered.
Formulation
The IL3RA protein is provided as a solution at a concentration of 0.25 mg/mL in a buffer consisting of 10% glycerol and phosphate-buffered saline (pH 7.4).
Stability
For short-term storage (up to 4 weeks), the protein can be stored at 4°C. For extended storage, it is recommended to store the protein at -20°C. To ensure optimal stability during long-term storage, the addition of a carrier protein such as albumin (HSA or BSA) to a final concentration of 0.1% is advisable. Repeated freezing and thawing of the protein should be avoided.
Purity
The purity of the IL3RA protein is greater than 90%, as determined by SDS-polyacrylamide gel electrophoresis (SDS-PAGE) analysis.
Synonyms

Interleukin 3 Receptor Subunit Alpha, Interleukin 3 Receptor, Alpha (Low Affinity), IL-3 Receptor Subunit Alpha, IL-3R Subunit Alpha, CD123 Antigen, IL-3R-Alpha, IL-3RA, IL3R, Interleukin-3 Receptor Subunit Alpha, IL-3 Receptor Alpha SP2 Isoform, HIL-3Ra, IL3RAY, CD123, IL3RX, IL3RY, IL3RA.

Source
Sf9, Baculovirus cells.
Amino Acid Sequence

ADLKEDPNPP ITNLRMKAKA QQLTWDLNRN VTDIECVKDA DYSMPAVNNS YCQFGAISLC EVTNYTVRVA NPPFSTWILF PENSGKPWAG AENLTCWIHD VDFLSCSWAV GPGAPADVQY DLYLNVANRR QQYECLHYKT DAQGTRIGCR FDDISRLSSG SQSSHILVRG RSAAFGIPCT
DKFVVFSQIE ILTPPNMTAK CNKTHSFMHW KMRSHFNRKF RYELQIQKRM QPVITEQVRD RTSFQLLNPG TYTVQIRARE RVYEFLSAWS TPQRFECDQE EGANTRAWRH HHHHH.

Q&A

What is IL3RA and what is its primary function in human biology?

IL3RA, also known as CD123, is the alpha subunit of the interleukin-3 receptor and forms a heterodimeric cytokine receptor complex with a beta subunit (IL3RB/CD131). This receptor specifically recognizes interleukin-3 (IL-3) and initiates signal transduction processes critical for hematopoietic cell development. The IL3RA gene is located in a pseudoautosomal region on chromosomes X or Y . Functionally, IL3RA serves as a cell surface receptor expressed on hematopoietic progenitor cells, monocytes, and B-lymphocytes that controls the production and differentiation of hematopoietic progenitor cells into lineage-restricted cells . When IL-3 binds to the receptor, it induces heterodimerization with IL3RB, triggering phosphorylation and activation of downstream signaling molecules.

The signaling cascade initiated by IL3RA activation involves several critical effector proteins, including JAK2 and PI3K, which play essential roles in cell proliferation and differentiation pathways . Specifically, the activation of JAK2 leads to STAT5-mediated transcriptional programming that regulates numerous genes involved in hematopoietic development . This signaling network is tightly regulated and plays a crucial role in normal blood cell development and function.

How do different IL3RA isoforms impact research methodology and therapeutic development?

Recent research has revealed the existence of multiple IL3RA isoforms that significantly impact both research approaches and therapeutic development. A study utilizing long-read transcriptomics on the PacBio platform augmented with short-read RNA sequencing identified novel IL3RA isoforms in pediatric acute myeloid leukemia (AML) samples . This discovery has profound implications for research methodology and therapeutic targeting.

The presence of different isoforms explains the observed discrepancies when various monoclonal antibodies are used to quantify CD123 on AML patient samples . These isoforms likely exhibit structural variations that affect epitope presentation on the cell surface, potentially modulating antibody binding efficiency and therapeutic responses. For researchers, this necessitates careful consideration of antibody selection for detection and quantification experiments, as different antibodies may recognize distinct epitopes that are variably present across isoforms.

Methodologically, researchers studying IL3RA should employ multiple complementary approaches:

  • Long-read sequencing technologies to identify full-length isoforms

  • Isoform-specific PCR primers for targeted quantification

  • Multiple antibody clones recognizing different epitopes for protein detection

  • Functional assays to determine isoform-specific activities

For therapeutic development, the heterogeneity in IL3RA isoform expression may partially explain the observed variability in patient responses to anti-CD123 therapeutics . This suggests that comprehensive isoform profiling might be valuable for patient stratification and therapeutic selection, potentially improving clinical outcomes in diseases where IL3RA-targeted approaches are employed.

What experimental methods provide the most reliable quantification of IL3RA expression?

Reliable quantification of IL3RA expression is crucial for both research and clinical applications. Several methodological approaches can be employed, each with distinct advantages depending on the specific research question.

For protein-level detection and quantification:

For transcript-level analysis:

  • Combined short-read and long-read RNA sequencing approaches provide the most comprehensive assessment of IL3RA expression, including identification of all expressed isoforms . The long-read approach using platforms like PacBio gives full isoform sequences that can be reliably translated into open reading frames, offering advantages over traditional short-read sequencing alone.

  • Quantitative PCR with carefully designed primers can provide targeted quantification of total IL3RA expression or specific isoforms. This approach is particularly valuable for analyzing large sample cohorts where sequencing might be cost-prohibitive.

For optimal results, researchers should consider combining multiple methodologies and including appropriate controls. Correlation between transcript and protein levels should also be established, as post-transcriptional regulation may result in discrepancies between mRNA and surface protein expression.

How does IL3RA signaling integrate with other cytokine receptor pathways?

IL3RA participates in a complex signaling network that intersects with multiple cytokine receptor pathways, particularly those sharing the common beta chain (IL3RB/CD131). This beta subunit is also utilized by receptors for granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-5 (IL-5) . Understanding these interconnections is crucial for interpreting experimental results and developing targeted interventions.

The signaling cascade initiated by IL3RA activation begins with heterodimerization with IL3RB following ligand binding . This receptor complex formation triggers the phosphorylation and activation of several downstream effector proteins, particularly JAK2 and PI3K . These activated kinases then initiate separate but interconnected signaling branches that collectively regulate cell proliferation, differentiation, and survival. The JAK2 pathway leads to STAT5 activation and nuclear translocation, resulting in transcriptional regulation of target genes .

Experimentally, researchers can distinguish IL3RA-specific signals from those of related receptors through several approaches:

  • Selective stimulation with recombinant IL-3

  • Use of receptor-specific blocking antibodies

  • Genetic approaches (siRNA, CRISPR) targeting IL3RA specifically

  • Pharmacological inhibitors targeting specific nodes in the signaling cascade

Since IL3RA shares downstream signaling components with other cytokine receptors, cross-talk between these pathways can influence cellular responses. This can lead to redundancy or synergy in biological effects, which should be considered when interpreting experimental results. Additionally, in pathological conditions like leukemia, aberrant activation of these shared pathways may contribute to disease progression and therapeutic resistance.

What are the structural characteristics of IL3RA that influence antibody binding and therapeutic targeting?

The structural features of IL3RA significantly impact antibody recognition and therapeutic targeting strategies. While detailed three-dimensional structural information is not provided in the search results, several key aspects of IL3RA structure are critical for researchers to consider.

IL3RA contains specific structural domains that form the binding interface with IL-3 and mediate interactions with the beta subunit (IL3RB). The presence of multiple IL3RA isoforms, as identified through long-read transcriptomics , suggests that alternative splicing or other post-transcriptional modifications may alter the protein structure. These structural variations likely affect epitope presentation, explaining the observed discrepancies when different monoclonal antibodies are used to quantify CD123 on patient samples .

For therapeutic development, understanding these structural nuances is essential. Different antibody-based therapeutics (including antibody-drug conjugates and CAR-T cells) target specific epitopes on the CD123 molecule . If these epitopes are altered or absent in certain isoforms, therapeutic efficacy may be compromised in patients expressing those variants. This structural heterogeneity may contribute to the variability observed in patient responses to anti-CD123 therapeutics .

Methodological approaches to address these challenges include:

  • Epitope mapping studies to identify conserved regions across isoforms

  • Structural biology techniques to characterize binding interfaces

  • Functional assays to assess the impact of structural variations on ligand binding and signaling

  • Development of therapeutics targeting multiple epitopes or conserved regions

For researchers developing IL3RA-targeted agents, considering these structural aspects and validating binding across different cellular contexts and isoform expression patterns is crucial for maximizing therapeutic potential.

How does the tissue and cell type-specific expression of IL3RA inform experimental design?

IL3RA exhibits distinct expression patterns across various tissues and cell types, which has significant implications for experimental design and interpretation. Understanding these patterns is essential for selecting appropriate experimental models and controls.

Primarily, IL3RA is expressed on hematopoietic progenitor cells, monocytes, and B-lymphocytes . This expression profile makes it particularly relevant for hematological studies and explains its importance as a therapeutic target in hematological malignancies such as AML . Additionally, IL3RA expression has been documented in brain tissue, although its function in neural contexts is less well characterized .

When designing experiments to study IL3RA, researchers should consider:

  • Cell model selection: Choose models that naturally express IL3RA or can be engineered to express specific isoforms. The THP-1 cell line has been documented to express detectable levels of IL3RA and can serve as a positive control for expression studies .

  • Control selection: Include appropriate positive and negative controls for expression analysis. When using antibody-based detection methods, be aware that different epitopes may be variably represented across isoforms .

  • Context-specific function: Design functional assays that reflect the physiological context of the cells being studied. For instance, in hematopoietic progenitors, assessments of proliferation and differentiation are particularly relevant.

  • Pathological relevance: When studying disease states, consider how IL3RA expression may differ from normal tissue. In AML, for example, CD123 is expressed on most blast cells , making it a valuable therapeutic target.

  • Species considerations: While human and murine IL3RA share functional similarities, there are important differences that should be considered when using animal models. Validation in human samples or humanized models is crucial for translational research.

For quantitative expression analysis, researchers may employ The Human Protein Atlas data or similar resources to guide their experimental design and interpretation of results in various tissue contexts.

What are the optimal methods for analyzing IL3RA isoform expression and function?

Analyzing IL3RA isoform expression and function requires a multi-faceted approach that combines advanced molecular techniques with functional assessments. Based on current research methodologies, several complementary strategies are recommended.

For comprehensive isoform identification and characterization:

  • Long-read RNA sequencing on platforms such as PacBio provides full-length isoform sequences that can be reliably translated into open reading frames . This approach overcomes the limitations of short-read sequencing in resolving complex splicing patterns and identifying novel isoforms.

  • Combine long-read data with short-read RNA sequencing for accurate quantification of relative isoform abundance across samples . This integrated approach leverages the depth of short-read sequencing with the length advantages of long-read technologies.

For targeted isoform detection and quantification:

  • Design isoform-specific PCR primers targeting unique exon junctions or sequence variants

  • Develop isoform-specific antibodies that recognize epitopes unique to particular variants

  • Consider targeted proteomics approaches to identify isoform-specific peptides

For functional characterization:

  • Express individual isoforms in appropriate cellular contexts using overexpression systems

  • Use CRISPR/Cas9 genome editing to create isoform-specific knockouts or modifications

  • Assess key functional parameters including:

    • Ligand binding affinity for IL-3

    • Heterodimerization with IL3RB

    • Activation of downstream signaling pathways (JAK2, STAT5, PI3K)

    • Biological outcomes (proliferation, differentiation, survival)

A robust experimental workflow might include:

  • Initial characterization of all expressed isoforms using long-read sequencing

  • Quantification of isoform expression across different samples or conditions

  • Correlation of isoform expression with clinical parameters or experimental outcomes

  • Functional studies of selected isoforms to determine their specific contributions to biology or disease

This comprehensive approach enables researchers to move beyond simple expression analysis to understand the functional significance of IL3RA isoform diversity in normal physiology and disease.

How should researchers design experiments to investigate IL3RA-mediated signaling pathways?

Designing experiments to investigate IL3RA-mediated signaling requires careful consideration of cellular contexts, stimulation parameters, and appropriate readouts. A systematic approach ensures reliable and reproducible results that accurately reflect the biology of this receptor system.

Cell system selection is critical:

  • Use cell types that naturally express both IL3RA and IL3RB to study physiologically relevant signaling

  • Consider primary cells (hematopoietic progenitors, monocytes) for maximum biological relevance

  • Cell lines like THP-1 can provide consistent experimental systems with detectable IL3RA expression

  • Engineering systems with controlled expression of IL3RA variants allows comparison of isoform-specific signaling

Stimulation protocols should be carefully optimized:

  • Use recombinant human IL-3 at physiologically relevant concentrations

  • Include appropriate controls (unstimulated, isotype controls, irrelevant cytokines)

  • Perform time-course experiments to capture both immediate-early signaling events and delayed responses

  • Consider pre-treatment with inhibitors targeting specific pathway components to dissect signaling networks

For signaling analysis, multiple complementary readouts provide the most comprehensive assessment:

  • Phosphorylation analysis of key nodes including JAK2 and STAT5 using phospho-specific antibodies

  • Transcriptional profiling to identify target genes activated by IL3RA signaling

  • Functional assays measuring biological outcomes (proliferation, differentiation, survival)

  • Protein-protein interaction studies to map the dynamic assembly of signaling complexes

Advanced techniques for in-depth signaling analysis include:

  • Proximity ligation assays to visualize protein interactions in situ

  • Live-cell imaging with fluorescent reporters to track signaling dynamics

  • Mass spectrometry-based phosphoproteomics for unbiased analysis of signaling networks

  • CRISPR screens to identify novel components of the IL3RA signaling pathway

When interpreting results, researchers should consider the potential impact of IL3RA isoform heterogeneity on signaling outputs . Different isoforms may activate distinct signaling pathways or exhibit quantitative differences in pathway activation, contributing to the functional diversity of IL3RA in different cellular contexts.

What controls and validation steps are essential when studying IL3RA in disease models?

Robust experimental design for studying IL3RA in disease models requires careful consideration of controls and validation steps to ensure reproducible and translatable findings. These considerations are particularly important given the complexity of IL3RA biology, including isoform diversity and context-specific functions.

Essential controls for expression analysis:

  • Positive controls: Include cell types known to express IL3RA, such as THP-1 cells , to validate detection methods

  • Negative controls: Cell lines lacking IL3RA expression or IL3RA knockout models

  • Isotype controls: For antibody-based detection, include appropriate isotype controls to assess non-specific binding

  • Multiple detection methods: Validate expression using orthogonal techniques (flow cytometry, Western blot, qPCR)

  • Multiple antibody clones: Use different antibodies recognizing distinct epitopes to account for isoform variations

For functional studies, critical controls include:

  • Receptor blocking: Use IL3RA-specific blocking antibodies to confirm specificity of observed effects

  • Ligand specificity: Compare responses to IL-3 versus other cytokines that signal through shared pathways

  • Genetic manipulation: Include IL3RA knockdown/knockout conditions to establish causality

  • Rescue experiments: Reconstitute IL3RA expression in knockout models to confirm specificity

Validation across experimental systems enhances reliability:

  • In vitro to in vivo translation: Confirm key findings from cell lines in primary cells and animal models

  • Cross-species validation: Validate findings across different species when using animal models

  • Patient-derived materials: When possible, validate findings using primary patient samples

  • Independent cohorts: Replicate findings across independent patient cohorts or experimental series

Disease-specific considerations for IL3RA studies:

  • In hematological malignancies: Compare findings between normal hematopoietic cells and malignant counterparts

  • Patient heterogeneity: Account for variability in IL3RA expression and isoform distribution among patients

  • Treatment effects: Consider how therapeutic interventions might alter IL3RA expression or function

  • Microenvironmental factors: Assess how the disease microenvironment affects IL3RA biology

Implementing these controls and validation steps ensures that research findings on IL3RA are robust, reproducible, and clinically relevant, ultimately advancing our understanding of its role in disease pathogenesis and therapeutic targeting.

How does IL3RA expression correlate with clinical outcomes in hematological malignancies?

IL3RA (CD123) expression has significant implications for clinical outcomes in hematological malignancies, particularly in acute myeloid leukemia (AML). Understanding these correlations is essential for both prognostic assessment and therapeutic decision-making.

CD123 is expressed on the surface of most AML blasts, making it a valuable therapeutic target for clinical intervention . While the search results don't provide specific prognostic correlations, the widespread expression of CD123 on leukemic cells has led to substantial interest in targeting this receptor therapeutically.

The clinical significance of IL3RA expression extends beyond simple presence/absence determination. Research indicates that variability in IL3RA mRNA isoform expression may induce epitope variation on the cell surface, potentially modulating antibody and therapeutic responses . This heterogeneity might explain the observed variability in patient responses to anti-CD123 therapeutics and suggests that more nuanced assessment of IL3RA expression patterns could improve patient stratification.

Methodologically, researchers investigating clinical correlations should consider:

  • Comprehensive isoform profiling using long-read transcriptomics combined with short-read RNA sequencing

  • Multiple antibody detection systems to account for epitope variations

  • Correlation of expression patterns with response to specific therapeutic modalities

  • Longitudinal assessment to track changes in expression during disease progression or treatment

Additionally, researchers should be aware that different monoclonal antibodies directed against CD123 show sizable discrepancies when used to quantify this antigen on AML patient samples . This methodological challenge necessitates standardized approaches for clinical assessment and careful interpretation of historical data using different detection methods.

For future clinical studies, integrating IL3RA isoform analysis with other molecular and clinical parameters may provide more precise prognostic and predictive information, ultimately enabling more personalized therapeutic approaches for patients with hematological malignancies.

What are the current approaches for targeting IL3RA in therapeutic development?

IL3RA (CD123) has emerged as an important therapeutic target, particularly in hematological malignancies like acute myeloid leukemia (AML) and blastic plasmacytoid dendritic cell neoplasm (BPDCN). Several approaches have been developed to target this receptor for therapeutic benefit.

Antibody-drug conjugates (ADCs) represent one major strategy for IL3RA targeting. These therapies combine the specificity of anti-CD123 antibodies with the cytotoxic potential of conjugated payloads, enabling selective delivery of toxic compounds to CD123-expressing cells . This approach aims to maximize therapeutic efficacy while minimizing systemic toxicity.

Chimeric antigen receptor (CAR) T cells directed against CD123 represent another promising approach . These engineered T cells specifically recognize and eliminate CD123-expressing cells, leveraging the power of adaptive immunity against malignant cells. The development of CD123-directed CAR-T cells has shown promise in preclinical studies and early clinical trials.

Protein therapeutics targeting IL3RA include tagraxofusp-erzs, an IL3-diphtheria toxin fusion protein that has received regulatory approval for BPDCN . This agent combines the targeting specificity of IL-3 with the potent cytotoxicity of diphtheria toxin, resulting in selective killing of CD123-expressing cells.

Despite these advances, therapeutic targeting of IL3RA faces several challenges. The heterogeneity in IL3RA isoform expression may induce epitope variation on the cell surface, potentially affecting antibody binding and therapeutic response . Different monoclonal antibodies directed against CD123 show sizable discrepancies when used to quantify this antigen on patient samples, highlighting the complexity of targeting this receptor .

For researchers and clinicians developing IL3RA-targeted therapies, methodological considerations include:

  • Comprehensive characterization of target expression using multiple detection methods

  • Evaluation of therapeutic agents against cells expressing different IL3RA isoforms

  • Development of companion diagnostics that accurately predict therapeutic response

  • Combination strategies to address potential resistance mechanisms

These approaches collectively aim to improve the efficacy and precision of IL3RA-targeted therapies while overcoming the challenges posed by biological heterogeneity.

How might IL3RA research inform therapeutic approaches beyond hematological malignancies?

While IL3RA (CD123) has been extensively studied in the context of hematological malignancies, emerging research suggests potential implications for therapeutic approaches in other disease areas. Understanding these broader applications requires consideration of IL3RA biology across different physiological and pathological contexts.

One notable area of research extends to inflammatory and allergic conditions. Studies have revealed connections between the IL-3 signaling pathway and asthma pathophysiology. IL-3 downregulation has been associated with more severe allergic asthma in pre-school children, and IL-3-deficient mice showed increased asthmatic traits in experimental models . These findings suggest that modulating the IL-3/IL3RA axis might have therapeutic potential in allergic disorders.

The expression of IL3RA in brain tissue, as documented in The Human Protein Atlas , raises intriguing questions about its potential roles in neurological conditions. While the specific functions of IL3RA in neural cells remain to be fully elucidated, this expression pattern suggests possible involvement in neuroimmune interactions or other brain-specific processes that could be therapeutically relevant.

Additionally, the IL3RA signaling pathway intersects with fundamental cellular processes including proliferation, differentiation, and survival. These pathways are dysregulated in numerous pathological conditions beyond hematological malignancies, suggesting potential applications in other disease contexts where these processes contribute to pathogenesis.

From a methodological perspective, researchers exploring IL3RA in non-hematological contexts should consider:

  • Comprehensive characterization of expression patterns in relevant tissues and cell types

  • Functional studies to determine context-specific roles of IL3RA signaling

  • Careful validation of targeting strategies in appropriate disease models

  • Consideration of isoform diversity and its implications for tissue-specific functions

By expanding research beyond traditional applications in hematological malignancies, investigators may uncover novel therapeutic opportunities for modulating IL3RA signaling across a broader spectrum of human diseases.

What are the most pressing unanswered questions about IL3RA biology?

Despite significant advances in understanding IL3RA biology, several critical questions remain unanswered. These knowledge gaps represent important opportunities for future research that could significantly impact both basic understanding and therapeutic applications.

The functional significance of IL3RA isoform diversity represents one of the most pressing unanswered questions. Recent work demonstrated the existence of novel IL3RA isoforms in pediatric AML samples , but the specific biological roles of these variants remain largely unexplored. Key questions include whether different isoforms activate distinct signaling pathways, exhibit varying affinities for IL-3, or contribute differently to normal and malignant hematopoiesis. Understanding these functional distinctions could have profound implications for therapeutic targeting and patient stratification.

The regulatory mechanisms controlling IL3RA expression and isoform selection also remain incompletely understood. While IL3RA is known to be expressed on hematopoietic progenitor cells, monocytes, and B-lymphocytes , the transcriptional and post-transcriptional mechanisms that regulate this expression pattern are not fully characterized. Elucidating these regulatory pathways could potentially identify novel approaches to modulate IL3RA expression therapeutically.

The structural basis for IL3RA function, particularly the conformational changes that occur during receptor activation and the structural features that differentiate isoforms, represents another important area for investigation. Detailed structural studies would inform structure-based drug design and potentially enable the development of more selective therapeutic agents.

Additionally, the potential roles of IL3RA in non-hematopoietic tissues, such as brain , remain largely unexplored. Understanding the function of IL3RA in these contexts could reveal unexpected biological roles and potentially identify novel therapeutic applications.

Methodologically, addressing these questions will require integration of advanced technologies including:

  • Cryo-electron microscopy for structural studies

  • CRISPR-based functional genomics for isoform-specific manipulation

  • Single-cell multi-omics for comprehensive characterization of expression patterns

  • Advanced animal models to study tissue-specific functions in vivo

How might emerging technologies advance IL3RA research in the next five years?

The landscape of IL3RA research is poised for transformation through the application of several emerging technologies that promise to deepen our understanding of this important receptor and improve therapeutic targeting approaches.

Long-read sequencing technologies have already demonstrated value in identifying novel IL3RA isoforms . In the coming years, advances in these platforms will likely enable more comprehensive characterization of transcript diversity at higher throughput and lower cost. Integration with single-cell approaches will allow researchers to map isoform expression patterns at unprecedented resolution, potentially revealing cell type-specific expression programs and regulatory mechanisms.

Advanced CRISPR-based genome editing tools will facilitate more precise functional studies of IL3RA. Beyond simple knockout approaches, base editing and prime editing technologies will enable introduction of specific mutations to study structure-function relationships. CRISPR activation/interference systems will allow temporal control of IL3RA expression, helping to dissect its roles in different developmental and disease contexts. High-throughput CRISPR screens targeting regulatory elements will elucidate the complex control mechanisms governing IL3RA expression.

Structural biology approaches, particularly advances in cryo-electron microscopy, will likely provide higher-resolution views of the IL3RA protein, potentially including complexes with IL-3 and the beta subunit. These structural insights will inform the rational design of therapeutics with improved specificity and potency. Computational approaches integrating structural data with molecular dynamics simulations will enhance our understanding of receptor activation mechanisms.

Spatial transcriptomics and proteomics technologies will enable researchers to study IL3RA expression and function within the native tissue architecture, providing crucial insights into its roles in complex cellular environments like the bone marrow niche or inflammatory tissues. These approaches will be particularly valuable for understanding how IL3RA-expressing cells interact with their microenvironment in both normal and pathological contexts.

Advanced protein engineering platforms will accelerate the development of next-generation therapeutics targeting IL3RA. These may include multi-specific antibodies targeting multiple epitopes, conditionally active biologics that function only in specific microenvironments, and engineered cell therapies with enhanced safety and efficacy profiles.

The integration of these technologies with computational approaches and machine learning will enable more comprehensive analysis of complex datasets, potentially revealing patterns and relationships that would be difficult to discern through traditional methods alone.

What interdisciplinary approaches might yield new insights into IL3RA function and therapeutic applications?

Advancing our understanding of IL3RA biology and its therapeutic applications will increasingly require interdisciplinary approaches that integrate expertise and methodologies from diverse scientific fields. These collaborative efforts promise to yield insights that might not emerge from traditional disciplinary approaches alone.

Integrating structural biology with computational approaches represents one promising interdisciplinary direction. Advanced structural characterization techniques combined with molecular dynamics simulations and computational drug design could accelerate the development of small molecules or biologics targeting specific functional domains of IL3RA. This integration could help address the challenges posed by isoform heterogeneity by identifying conserved structural features or designing agents capable of recognizing multiple isoforms.

Systems biology approaches combining transcriptomics, proteomics, and functional genomics can provide comprehensive views of IL3RA within broader signaling networks. This holistic perspective is crucial for understanding how IL3RA interacts with other pathways in both normal physiology and disease states. Such analyses might reveal unexpected connections and potential combination therapy approaches for diseases where IL3RA plays a role.

Immuno-engineering represents another powerful interdisciplinary approach. Combining immunology expertise with bioengineering principles has already yielded CAR-T cell therapies targeting CD123 . Future advances might include engineered cells with enhanced targeting precision, controlled persistence, or conditional activation mechanisms that improve safety and efficacy profiles. Similarly, protein engineering approaches could yield novel IL3RA-targeted biologics with optimized properties.

Clinical and translational research integration is essential for advancing IL3RA-targeted therapies. Combining basic research findings with clinical observations can identify biomarkers predictive of therapeutic response and resistance mechanisms. The observed variability in patient responses to anti-CD123 therapeutics highlights the importance of this translational perspective. Collaborative efforts between laboratory scientists and clinicians will be crucial for designing rational clinical trials and interpreting their results.

Data science and artificial intelligence approaches applied to large, integrated datasets spanning genomics, transcriptomics, proteomics, and clinical outcomes could reveal patterns not obvious through conventional analysis. These computational approaches might identify patient subgroups most likely to benefit from IL3RA-targeted therapies or suggest novel combination strategies.

By fostering these interdisciplinary collaborations and integrating diverse methodological approaches, researchers can accelerate progress in understanding IL3RA biology and developing more effective therapeutic strategies for diseases where this receptor plays a significant role.

Product Science Overview

Introduction

Interleukin-3 Receptor Subunit Alpha, also known as CD123, is a specific subunit of the interleukin-3 receptor. This receptor is a heterodimeric cytokine receptor composed of a ligand-specific alpha subunit and a signal-transducing beta subunit. The alpha subunit is crucial for binding interleukin-3 (IL-3), while the beta subunit is necessary for the biological activities of IL-3. The receptor is involved in the regulation of hematopoietic progenitor cells, monocytes, and B-lymphocytes, controlling their production and differentiation into lineage-restricted cells .

Gene and Protein Structure

The gene encoding Interleukin-3 Receptor Subunit Alpha (IL3RA) is located in a cytokine receptor gene cluster in the X-Y pseudoautosomal region on chromosomes X or Y. The protein encoded by this gene is a cell surface receptor expressed on hematopoietic progenitor cells, monocytes, and B-lymphocytes . The receptor’s structure allows it to interact with IL-3, leading to the activation of various signaling pathways, including the JAK2-STAT5 pathway, which plays a role in cell proliferation and differentiation .

Biological Function

The primary function of Interleukin-3 Receptor Subunit Alpha is to mediate the effects of IL-3. Upon binding to IL-3, the receptor undergoes heterodimerization with the beta subunit, leading to the phosphorylation and activation of effector proteins such as JAK2 and PI3K. This activation triggers a cascade of signaling events that result in the proliferation and differentiation of hematopoietic progenitor cells .

Clinical Significance

Interleukin-3 Receptor Subunit Alpha is strongly expressed in various leukemic blasts and leukemic stem cells, making it an excellent target for leukemia therapy . The receptor’s role in hematopoiesis and its expression in leukemic cells have made it a focus of research for developing targeted therapies for hematologic cancers .

Recombinant Production

Recombinant human Interleukin-3 Receptor Subunit Alpha is produced using recombinant DNA technology. This involves inserting the gene encoding IL3RA into a suitable expression system, such as bacteria, yeast, or mammalian cells, to produce the protein in large quantities. The recombinant protein is then purified and used for various research and therapeutic applications .

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 2024 Thebiotek. All Rights Reserved.