Parameter | Value | Source |
---|---|---|
Expression Host | Sf9 Baculovirus cells | |
Purity | >90% (SDS-PAGE) | |
Formulation | 0.5 mg/ml in PBS (pH 7.4) + 10% glycerol | |
Storage | 4°C (short-term), -20°C (long-term) with 0.1% HSA/BSA carrier |
IL-9 Human, Sf9 Active modulates multiple immune cell subsets:
T Cells:
Mast Cells:
ILC2/Eosinophils:
Cell Proliferation: ED₅₀ ≤ 0.3 ng/ml in MO7e human megakaryocytic leukemic cells .
Cytokine Production: Induces IL-5, IL-13, and IL-9R upregulation in mast cells/ILC2 .
Interleukin-9 (IL-9) is a protein that plays a role in regulating hematopoiesis. It promotes the growth of certain blood cells, including megakaryoblastic leukemic cells. IL-9 is produced by various immune cells, such as mast cells, Treg cells, NKT cells, Th17 cells, Th2 cells, ILC2 cells, and primarily by Th9 cells, a subset of CD4+ T cells.
Recombinant human IL-9, expressed in Sf9 insect cells using a baculovirus system, is a single, glycosylated polypeptide chain. It consists of 132 amino acids (residues 19-144), with a molecular weight of 14.9 kDa. The IL-9 protein includes a 6-amino acid Histidine tag fused at the C-terminus and is purified using proprietary chromatographic techniques.
The IL-9 solution is provided at a concentration of 0.5 mg/ml in a buffer containing 10% glycerol and Phosphate-Buffered Saline (pH 7.4).
For short-term storage (2-4 weeks), the product can be stored at 4°C. For longer periods, it is recommended to store the product frozen at -20°C. To ensure stability during long-term storage, adding a carrier protein (0.1% HSA or BSA) is recommended. Avoid repeated freezing and thawing cycles.
The purity of the IL-9 protein is greater than 85% as determined by SDS-PAGE analysis.
The biological activity of IL-9 is assessed through a cell proliferation assay using MO7e human megakaryocytic leukemic cells. The ED50 value, representing the effective concentration for half-maximal response, is less than or equal to 0.3 ng/ml.
Interleukin 9, Protein Il9, Il9, HP40, Cytokine P40, T-cell growth factor P40
QGCPTLAGIL DINFLINKMQ EDPASKCHCS ANVTSCLCLG IPSDNCTRPC FSERLSQMTN
TTMQTRYPLI FSRVKKSVEV LKNNKCPYFS CEQPCNQTTA GNALTFLKSL LEIFQKEKMR GMRGKIHHHH HH
Human IL-9 is a glycosylated cytokine of approximately 30-40 kDa, with the mature protein spanning amino acids Gln19-Ile144 . When expressed in Spodoptera frugiperda (Sf21) cells using a baculovirus expression system, the recombinant protein maintains the biological activity of native IL-9 but may exhibit differences in glycosylation patterns.
The baculovirus-derived IL-9 typically migrates as 33-37 kDa under reducing conditions during SDS-PAGE due to glycosylation, despite having a calculated molecular weight of approximately 16 kDa . This glycosylation profile is important for researchers to consider when comparing experimental results between different sources of IL-9.
IL-9 signaling is primarily mediated through interaction with the IL-9 receptor (IL-9R), which forms a functional complex with the common gamma chain (γc) . This receptor engagement activates the JAK/STAT pathway, particularly:
The activation of these pathways leads to various cellular responses depending on the target cell type, including proliferation, survival enhancement, and altered gene expression profiles. In macrophages, IL-9 stimulation leads to M1 polarization in an IFNγ-dependent manner .
For optimal stability and activity:
Long-term storage: Maintain lyophilized protein at -20°C or lower
Reconstitution: Follow specific instructions provided in the Certificate of Analysis, typically using a neutral buffer such as PBS (pH 7.4)
Avoid repeated freeze-thaw cycles to maintain protein integrity and activity
Once reconstituted, aliquot and store at appropriate temperatures based on intended time to use
For experimental reproducibility, researchers should document reconstitution methods, buffer composition, and storage conditions in their protocols.
Effective IL-9 concentrations vary significantly based on the experimental system and desired biological response:
When designing dose-response experiments, researchers should include concentrations above and below these reference points to establish system-specific optimal dosing.
A multi-parameter approach is recommended for validating IL-9 activity:
Bioassay validation: Measure proliferative responses in IL-9-responsive cell lines
Receptor engagement: Confirm IL-9R expression on target cells via flow cytometry or western blot
Signaling verification: Assess phosphorylation of downstream STAT proteins (particularly STAT1, STAT3, STAT4, and STAT5)
Functional readouts: Depending on cell type:
For macrophages: M1 marker expression (CD80, CD86, TNF-α, IL-12)
For T cells: Proliferation, cytokine production, stemness markers
For mast cells: Degranulation and cytokine production
Control experiments should include IL-9 neutralizing antibodies or receptor blocking antibodies to confirm specificity of observed effects .
For accurate quantification of IL-9:
ELISA: Using commercial kits (e.g., ELISA MAX Deluxe Set Mouse IL-9 kit) for measuring IL-9 in culture supernatants or biological fluids
Multiplex bead arrays: For simultaneous measurement of IL-9 alongside other cytokines
Western blotting: For semi-quantitative analysis in cell lysates
qPCR: For measuring IL-9 expression at the mRNA level
When analyzing IL-9 in complex biological samples, researchers should be aware of potential matrix effects and consider appropriate sample dilution and spike recovery experiments to validate quantification accuracy.
IL-9 has significant effects on macrophage polarization with implications for tumor immunology:
M1 polarization mechanism: IL-9 polarizes macrophages toward the proinflammatory M1 phenotype through an IFNγ-dependent pathway
Reprogramming capabilities: IL-9 can reeducate M2 macrophages and tumor-associated macrophages (TAMs) toward the M1 phenotype both in vitro and in vivo
Chemokine induction: IL-9-polarized macrophages release:
IL-9 receptor (IL-9R) engineering represents an emerging approach in T cell immunotherapy:
Natural orthogonality: Due to limited natural expression of IL-9R, this receptor system offers near-orthogonal qualities that can be exploited in engineered T cells
Signaling advantages: Compared to synthetic orthogonal IL-9 receptors (o9R), T cells engineered with natural IL-9R exhibit:
STAT signaling profile: IL-9R activates STAT1, STAT3, STAT5, and uniquely STAT4, creating an optimal signaling balance for T cell function
Importantly, IL-9R-engineered T cells demonstrate exquisite sensitivity to signaling perturbations, with STAT1 functioning as a critical rheostat between:
These findings suggest that IL-9/IL-9R signaling may offer unique advantages for engineered T cell therapies compared to other cytokine/receptor systems.
Production of functional Sf9-derived IL-9 involves several technical considerations:
Expression system optimization:
Purification challenges:
Glycosylation heterogeneity: Results in mixed populations requiring careful chromatography
Protein aggregation: May occur during concentration steps
Endotoxin removal: Critical for in vivo applications
Quality control assessments:
Researchers should consider these factors when either producing IL-9 in-house or selecting commercial sources for their experiments.
Inconsistent responses to IL-9 can be addressed through systematic investigation:
Receptor expression analysis:
Quantify IL-9R and γc levels on target cells via flow cytometry
Verify receptor functionality through phospho-STAT assays
Consider cell-specific receptor isoforms or splice variants
Contextual signaling considerations:
Pre-existing activation state of cells (naive vs. activated)
Presence of competing or synergistic cytokines
Cell-specific JAK/STAT expression profiles
Standardization approaches:
Use internal standards between experiments
Include positive control cell lines with known IL-9 responses
Normalize data to account for baseline variations
When inconsistencies persist, researchers should consider sequencing their IL-9 and IL-9R to verify the absence of mutations that might affect binding or signaling.
When translating findings between mouse models and human systems:
Sequence homology: Mature human IL-9 shares only 57% amino acid sequence identity with mouse IL-9 , which can impact:
Cross-species reactivity
Antibody recognition
Receptor binding kinetics
Functional differences:
Cell type-specific responses may vary between species
Concentration requirements for bioactivity may differ
Downstream gene targets may show species-specific patterns
Experimental design considerations:
Use species-matched cytokines and reagents when possible
Consider humanized mouse models for more translational relevance
Validate key findings in both systems independently
These differences underscore the importance of careful experimental design when conducting translational research involving IL-9.
IL-9 exhibits context-dependent functions that can lead to apparently contradictory results:
Disease context specificity:
Analytical framework for resolving conflicts:
Examine timing of IL-9 exposure (acute vs. chronic)
Consider tissue microenvironment (cytokine milieu, oxygen tension)
Analyze cell populations present (mast cells, T cells, macrophages)
Evaluate route of administration and local concentrations
Technical considerations:
Differences in recombinant protein sources and activity
Variations in neutralizing antibody specificity and efficacy
Genetic background effects in animal models
When faced with conflicting data, researchers should systematically evaluate these variables and consider designing experiments that directly address the apparent contradictions.
Structure-guided engineering approaches offer promising avenues for IL-9-based therapeutics:
Receptor signal modulation:
Stability and half-life enhancements:
PEGylation or fusion to Fc domains to extend circulating half-life
Disulfide engineering to improve thermal stability
Glycosylation site modifications to reduce immunogenicity
Targeting strategies:
Bifunctional fusion proteins combining IL-9 with targeting domains
Cell-specific delivery using nanoparticle formulations
Conditionally active IL-9 variants responsive to tumor microenvironment signals
These approaches could enhance the therapeutic index of IL-9-based interventions while minimizing off-target effects.
IL-9's ability to polarize macrophages has several potential therapeutic applications:
TAM reprogramming strategies:
Local delivery of IL-9 to reshape the tumor microenvironment
Combination with checkpoint inhibitors to overcome immunosuppression
Sequential therapy to first polarize TAMs before activating T cells
Delivery optimization:
Biomarker development:
Identification of patients likely to benefit from IL-9-based therapies
Monitoring markers of macrophage polarization during treatment
Correlation of M1/M2 ratios with clinical outcomes
These approaches could be particularly relevant for "cold" tumors where lack of immune infiltration limits the efficacy of current immunotherapies.
Understanding IL-9's position in cytokine networks is crucial for predicting therapeutic outcomes:
Synergistic interactions:
Antagonistic relationships:
Network modeling approaches:
Systems biology tools to predict outcomes of multi-cytokine environments
In vitro validation using cytokine combinations
In vivo modeling using conditional knockout or reporter systems
A deeper understanding of these networks will inform more sophisticated combination therapy approaches that leverage IL-9's unique properties while mitigating potential antagonistic interactions.
Human Recombinant IL-9 is produced using Sf9 Baculovirus cells. This method involves using insect cells (Sf9) infected with a baculovirus that carries the gene for human IL-9. The resulting protein is a single, glycosylated polypeptide chain containing 132 amino acids, with a molecular mass of approximately 14.9 kDa . The recombinant IL-9 is fused to a 6 amino acid His tag at the C-terminus, which aids in its purification through chromatographic techniques .
The IL-9 solution is typically formulated with 10% glycerol and phosphate-buffered saline (pH 7.4) to maintain its stability . For short-term storage, it can be kept at 4°C if used within 2-4 weeks. For longer periods, it should be stored frozen at -20°C, with the addition of a carrier protein (0.1% HSA or BSA) to prevent degradation . It is important to avoid multiple freeze-thaw cycles to maintain the protein’s activity.