Recombinant Human Interleukin-3 (IL3), partial (Active) (GMP)

Shipped with Ice Packs
In Stock

Description

Definition and Production

Recombinant Human IL-3 (partial, Active, GMP) is a truncated form of the native IL-3 protein (amino acids 20–152) produced using expression systems such as E. coli or HEK293 cells . The partial sequence excludes the N-terminal signal peptide, enhancing solubility while retaining bioactivity . GMP-grade production ensures compliance with stringent quality controls, including animal-free reagents and endotoxin limits (<0.1 EU/μg) .

Key Functional Metrics

  • ED₅₀/EC₅₀: 0.02–2.0 ng/mL in TF-1 cell proliferation assays .

  • Specific Activity: Ranges from >1.7 × 10⁶ IU/mg (R&D Systems) to >1.0 × 10⁷ IU/mg (Cusabio) .

  • Mechanisms:

    • Activates JAK2/STAT5 and PI3K/AKT pathways .

    • Inhibits osteoclast differentiation via NF-κB suppression .

Species Specificity

Exhibits no cross-reactivity with murine cells due to low sequence homology (29% vs. human IL-3) .

Therapeutic Outcomes

  • Myelodysplastic Syndromes: Increased neutrophil and platelet counts in 30–60% of patients .

  • Aplastic Anemia: Modest hematopoietic restoration, particularly thrombopoiesis .

  • Secondary Hematopoietic Failure: Improved recovery post-chemotherapy/radiotherapy .

Adverse Effects

  • Low-grade fever, headaches, and transient thrombocytopenia at doses ≥250 μg/m² .

GMP Compliance

  • Produced under ISO 13485 or cGMP guidelines .

  • Animal-component-free (ACF) and xeno-free (XF) processes .

Stability

  • Storage: Lyophilized form stable at -80°C for years; reconstituted aliquots stable at 4°C for 1 week .

Comparative Product Analysis

ManufacturerExpression SystemPuritySpecific ActivityEndotoxin
R&D Systems E. coli≥97%>1.7 × 10⁶ IU/mg<0.1 EU/μg
Abcam HEK293≥95%Not quantified<0.005 EU/μg
Cusabio E. coli>98%>1.0 × 10⁷ IU/mg<0.01 EU/μg
PTGLab HEK293>95%5.0–9.2 × 10⁵ IU/mg<0.1 EU/μg

Research Limitations and Future Directions

  • Variability in Bioactivity: ED₅₀ differences reflect assay conditions (e.g., cell line sensitivity) .

  • Combination Therapies: Ongoing trials explore synergies with GM-CSF or erythropoietin .

Product Specs

Buffer
Lyophilized from a 0.2 µm filtered concentrated solution in phosphate-buffered saline (PBS), pH 7.4, containing 0.02% Tween-20.
Form
Lyophilized powder
Lead Time
Product shipment typically occurs within 5-10 business days of order receipt. Delivery times may vary depending on the purchasing method and location. Please consult your local distributor for precise delivery estimates.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to pellet the contents. Reconstitute the protein in sterile, deionized water to a final concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and storing in aliquots at -20°C or -80°C. Our standard protocol uses 50% glycerol.
Shelf Life
Shelf life depends on several factors including storage conditions, buffer composition, temperature, and the inherent stability of the protein. Generally, the shelf life of the liquid formulation is 6 months at -20°C/-80°C, while the lyophilized form has a shelf life of 12 months at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C or -80°C. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag-Free
Synonyms
Colony stimulating factor multiple; Hematopoietic growth factor; IL 3; IL-3; IL3; IL3_HUMAN; Interleukin 3 (colony stimulating factor; multiple); Interleukin 3; Interleukin-3; Mast cell growth factor; MCGF; MGC79398; MGC79399; Multi CSF; Multilineage colony stimulating factor; Multipotential colony stimulating factor; Multipotential colony-stimulating factor; OTTHUMP00000065963; P cell stimulating factor ; P-cell-stimulating factor
Datasheet & Coa
Please contact us to get it.
Expression Region
20-152aa
Mol. Weight
15.1 kDa
Protein Length
Partial
Purity
> 98 % by SDS-PAGE and HPLC analyses.
Research Area
Immunology
Source
E.Coli
Species
Homo sapiens (Human)
Target Names
IL3
Uniprot No.

Target Background

Function

Granulocyte/macrophage colony-stimulating factors (GM-CSFs) are cytokines crucial in hematopoiesis. They regulate the production, differentiation, and function of granulocytes and monocytes/macrophages, two closely related white blood cell populations. This specific CSF stimulates the production of granulocytes, macrophages, mast cells, stem cells, erythroid cells, eosinophils, and megakaryocytes.

Gene References Into Functions
  1. IL-3's potential involvement in immediate memory deficits during the chronic phase of schizophrenia. PMID: 29554544
  2. IL-3's role in enhancing human mesenchymal stem cell (MSC) migration by regulating the CXCR4/SDF-1α axis, suggesting its potential in regenerative cell therapies. PMID: 28705238
  3. IL-3's regulation of endothelial cell-extracellular vesicle release, cargo, and angiogenic paracrine action via STAT5. PMID: 27157262
  4. The reciprocal blunting effect of T-GM-CSF and -IL-3 on the receptor binding and myeloid progenitor cell proliferation activity of FL-GM-CSF and -IL-3, both in vitro and in vivo. PMID: 28344320
  5. IL-3's induction of signaling pathways associated with increased cell survival under oxidative stress, correlating with glucose uptake stimulation. PMID: 27862234
  6. The effects of IL-3/GM-CSF on the myofibroblastic differentiation of human adipose-derived stromal cells (hASCs) and human dermal fibroblasts (HDFs). PMID: 28377320
  7. The use of genetically engineered mesenchymal stromal cells producing IL-3 and TPO to improve human scaffold-based xenograft models. PMID: 28456746
  8. IL-3's minimal downregulation of its receptor's alpha-chain while preserving the common beta-chain, leading to sustained STAT5 activation. PMID: 27443880
  9. The potential influence of IL-3 rs181781 and CTLA4 rs4553808 gene polymorphisms on tacrolimus dose-adjusted concentrations. PMID: 28112181
  10. The lack of association between IL-3 and IL-13 polymorphisms and rheumatoid arthritis (RA), except for a potential increased risk associated with IL-13 rs1800925 in patients with ESR <25.00. PMID: 27323078
  11. IL-3 and IL-12p40 as potential molecular predictors for recurrent wheezing due to respiratory syncytial virus (RSV) infection. PMID: 26299549
  12. IL3 as an important genetic regulator of human brain volume variation, potentially subject to positive selection during human evolution. PMID: 26875095
  13. The IL-3 dependence of thymic stromal lymphopoietin activation of basophils in allergic asthma patients. PMID: 25962901
  14. Lack of confirmation of an association between IL-3 SNPs and schizophrenia. PMID: 26277822
  15. Association of high plasma IL-3 levels with high mortality in sepsis. PMID: 25766237
  16. Association of genetic variation in the interleukin-3 gene with breast cancer risk. PMID: 24670917
  17. The role of HuR in IL-3 3'-UTR-mediated post-transcriptional control in T-cells. PMID: 24658545
  18. IL-13 rs20541 SNP as a risk factor for asthma. PMID: 23978640
  19. The link between beta-catenin activation and IL-3, suggesting that targeting IL-3 signaling may inhibit beta-catenin activity in some acute myeloid leukemia (AML) patients. PMID: 24598054
  20. Significant association of IL3 SNP rs40401 with asthma risk in young adult Japanese women. PMID: 24684517
  21. Significant positive association between IL3 SNP rs40401 variant and the risk of rhinoconjunctivitis. PMID: 23953855
  22. Association of IL3 rs2073506 G>A polymorphism with increased risk of nodal and metastatic esophageal cancer. PMID: 23726808
  23. IL3 promoter genetic variation's effect on human brain volume by regulating neural progenitor proliferation and survival. PMID: 23226269
  24. The use of transgenic mice to study the developmental regulation of the IL-3 and GM-CSF locus and identify DNA enhancer elements. PMID: 23024272
  25. The role of eosinophils as a source of proMMP-9 in microenvironments rich in betac-family cytokines and TNF, highlighting synergistic interaction between TNF and betac-family cytokines for proMMP-9 synthesis. PMID: 22321809
  26. Association of the minor allele for IL13 rs1295686 SNP with increased likelihood of reporting breast pain before surgery. PMID: 22515947
  27. The potential of targeting IL-3 to hamper tumor angiogenesis through an IL-3 autocrine loop driving a tumor endothelial switch. PMID: 21643009
  28. IL-3's potential neuroprotective role in Alzheimer's disease by providing cellular protection against amyloid-beta neurotoxicity. PMID: 20964623
  29. Protective effect of IL3 rs40401 and rs40401 genotypes against malaria attacks. PMID: 21224257
  30. The role of PLCγ2 and Ca(2+) signaling in IL3/GM-CSF stimulated human hematopoietic stem/progenitor cells via MEK/ERK modulation. PMID: 21506110
  31. Increased risk for Graves' disease and Graves' ophthalmopathy conferred by the interaction of 6 loci from 5 interleukin genes compared to single risk alleles. PMID: 20332709
  32. Significant relationship between IL-3 genotypes and SNPs in acute rejection subjects compared to patients without acute rejection. PMID: 21168724
  33. The support of human alveolar macrophage development and human immune responses in the lung by human IL-3/GM-CSF knock-in mice. PMID: 21262803
  34. RhoH's function as a negative regulator of IL3-induced signals through JAK-STAT pathway modulation. PMID: 20738848
  35. The importance of the domain 1 D-E loop disulfide of hβc and β(IL-3) in maintaining ligand-binding residue positions for high-affinity binding and signaling. PMID: 20516062
  36. Two different modes of βc binding utilized in the presence of hIL-3Rα isoforms. PMID: 20472554
  37. Valproic acid's (VPA) novel role in enhancing IL-3 stimulation of megakaryopoiesis and erythropoiesis. PMID: 20381581
  38. Progressive activation of the IL-3/GM-CSF locus during T cell differentiation, with stepwise increases in active modifications and cytokine-expressing cells. PMID: 20147630
  39. IL-3 and oncogenic Abl's regulation of the myeloblast transcriptome by altering mRNA stability. PMID: 19829692
  40. Binding kinetics of native IL-3 and several variants to the IL-3 receptor. PMID: 11700046
  41. IL-3's induction of MHC class II and B7.2 expression on eosinophils, enabling T cell proliferation support. PMID: 11714768
  42. Signaling inhibition by antisense oligodeoxynucleotides targeting the common beta chain of receptors. PMID: 11763346
  43. Ectopic IL3 expression in myeloid leukemic cells with t(5;12)(q31;p13), suggesting deregulation by the translocation and a variant leukemogenic mechanism. PMID: 11861295
  44. The influence of the antiapoptotic cytokine IL-3, along with SCF and FLT3L, on the proliferation of γ-irradiated AC133+/CD34+ progenitor cells. PMID: 12002675
  45. Differentiation of monocytes cultured with IL-3 (plus IL-4) into dendritic cells producing less IL-12 and shifting T helper (Th) cell responses toward a Th2 cytokine pattern. PMID: 12055233
  46. Increased activity of mutated interleukin 3 due to a change from a rare to a common ligand, enhancing IL-3-dependent signaling. PMID: 12093816
  47. IL-3's role in potentiating hematopoietic cell migration. PMID: 12135758
  48. Regulation of the IL-3 gene by two enhancers with distinct but overlapping tissue specificities. PMID: 12165512
  49. IL-3 stimulation of mcl-1 gene transcription through the SIE motif, involving p38(MAPK)-dependent phosphorylation of PU.1 at serine 142. PMID: 12612065
  50. IL-3-induced reduced expression of IL-5Rα in eosinophils, while increasing IL-3Rα expression, leaving GM-CSF receptor α expression unaffected. PMID: 12759409
Database Links

HGNC: 6011

OMIM: 147740

KEGG: hsa:3562

STRING: 9606.ENSP00000296870

UniGene: Hs.694

Protein Families
IL-3 family
Subcellular Location
Secreted.
Tissue Specificity
Activated T-cells, mast cells, natural killer cells.

Q&A

What is Recombinant Human IL-3 and what are its structural characteristics?

Recombinant Human Interleukin-3 (IL-3) is a pleiotropic cytokine belonging to the interleukin family with a molecular weight of approximately 15.1-15.2 kDa in its monomeric form . It is characterized by a four-helix bundle structure and shares structural similarities with GM-CSF and IL-5 . The protein sequence of the partial active form includes residues 20-152aa of the full-length protein, with the amino acid sequence beginning with APMTQTTSLKTS and ending with QQTTLSLAIF .

When analyzing IL-3 via SDS-PAGE, it migrates as a major band at approximately 15.2 kDa under both reducing and non-reducing conditions . The protein is typically tag-free in its recombinant form to ensure native-like activity and minimize interference with biological function . The recombinant protein possesses >98% purity as determined by SDS-PAGE and HPLC analyses, making it suitable for precise experimental applications .

How is the biological activity of Recombinant Human IL-3 measured?

The biological activity of Recombinant Human IL-3 is primarily assessed through cell proliferation assays using IL-3-dependent cell lines, most commonly TF-1 human myeloid leukemia cells . The activity is quantified by determining the ED50 (effective dose required for 50% maximal response), which typically ranges from less than 0.1 ng/ml to 0.5 ng/ml, corresponding to a specific activity of >1.0 × 10^7 IU/mg to >2 × 10^7 units/mg .

The standard protocol involves treating TF-1 cells in triplicate with serial dilutions of IL-3 for 72 hours, followed by measurement of cell viability using luminescence-based assays such as CellTiter-Glo . A typical dose-response curve might show an EC50 of approximately 102 pg/ml (6.7 pM) . For more rigorous analyses, researchers should validate activity across multiple biological readouts, including:

  • Cell proliferation kinetics

  • Phosphorylation of downstream signaling molecules (e.g., JAK/STAT, Ras-Raf-ERK)

  • Expression of anti-apoptotic bcl-2 family members

  • Colony formation of multiple hematopoietic lineages

What are the optimal reconstitution methods for lyophilized IL-3?

Optimal reconstitution of lyophilized Recombinant Human IL-3 requires careful attention to buffer conditions and handling procedures to maintain biological activity. The lyophilized protein is typically prepared from a filtered concentrated solution in PBS, pH 7.4, with 0.02% Tween-20, or from acetonitrile with TFA .

For reconstitution, the following methodology is recommended:

  • Briefly centrifuge the vial prior to opening to bring contents to the bottom.

  • Reconstitute the protein in deionized sterile water to achieve a concentration of 0.1-1.0 mg/mL .

  • For enhanced stability, add glycerol to a final concentration of 5-50%, with 50% being optimal for long-term storage .

  • Alternatively, reconstitute in 10 mM HCl at >50 μg/ml, particularly for preparations susceptible to aggregation .

  • If required for specific applications, add carrier protein after initial reconstitution.

  • Prepare single-use aliquots to avoid repeated freeze-thaw cycles.

It is essential to note that repeated freezing and thawing is not recommended as it may lead to protein degradation and loss of activity . Working aliquots can be stored at 4°C for up to one week, while long-term storage requires -20°C or preferably -80°C conditions .

How can researchers optimize IL-3 stability for long-term experimental protocols?

Stability optimization for IL-3 in extended experimental protocols requires strategic approaches to preserve structural integrity and biological activity. The shelf life of IL-3 preparations varies depending on storage conditions: generally, liquid forms maintain stability for approximately 6 months at -20°C/-80°C, while lyophilized forms can be stable for up to 12 months under the same conditions .

To maximize stability for long-term protocols, researchers should implement the following strategies:

  • Buffer Optimization: The choice of reconstitution buffer significantly impacts stability. For applications requiring extended stability, consider:

    • Using 10 mM HCl as the initial reconstitution solution

    • Adding protein stabilizers such as HSA or BSA (0.1-1%) for dilute solutions

    • Maintaining pH between 4.0-5.0 where IL-3 demonstrates greatest stability

  • Storage Temperature Hierarchy:

    • For daily use: 4°C (stable for up to 7 days)

    • For monthly use: -20°C (stable for up to 6 months in liquid form)

    • For long-term archiving: -80°C (preferred for maintaining activity beyond 6 months)

  • Aliquoting Strategy:

    • Calculate experimental needs and prepare appropriately sized single-use aliquots

    • Use low-binding microcentrifuge tubes to minimize protein loss through adsorption

    • Document freezing date and concentration on each tube

  • Quality Control Monitoring:

    • Periodically test activity using TF-1 proliferation assays to ensure maintained potency

    • Monitor for signs of aggregation or precipitation before each experimental use

How does IL-3 contribute to hematopoietic stem cell differentiation protocols?

IL-3 plays a pivotal role in hematopoietic stem cell (HSC) differentiation protocols due to its multilineage stimulatory capacity. As a multi-colony stimulating factor, IL-3 stimulates the development and colony formation of multiple lineages of hematopoietic cells by activating intracellular pathways such as Ras-Raf-ERK and JAK/STAT . It also inhibits apoptosis and promotes cell survival by targeting the anti-apoptotic bcl-2 gene family .

In research protocols, IL-3 is strategically employed in several differentiation pathways:

  • Myeloid Lineage Differentiation:

    • IL-3 is used as an early regulator for the differentiation of induced pluripotent stem cells into myeloid lineages .

    • When combined with lineage-instructive cytokines, IL-3 facilitates development of specific cell types:

      • With GM-CSF and IL-4 for dendritic cell differentiation

      • With M-CSF for macrophage generation

  • Erythrocyte and Megakaryocyte Development:

    • IL-3 in combination with IL-12 promotes erythrocyte differentiation

    • When used with IL-6, it enhances megakaryocyte development

  • Hematopoietic Progenitor Expansion:

    • IL-3 acts synergistically with IL-6 to enhance proliferation of hematopoietic progenitors

    • This combination is particularly effective for ex vivo expansion protocols prior to transplantation

For optimal differentiation protocols, researchers should implement sequential cytokine exposure regimens, where IL-3 is typically introduced early in the differentiation process (days 0-5) at concentrations of 10-50 ng/mL, followed by lineage-specific cytokines at later stages.

What are the clinical implications of IL-3's effects on thrombopoiesis and granulopoiesis?

IL-3's ability to stimulate multiple hematopoietic lineages, particularly thrombopoiesis and granulopoiesis, has significant clinical research implications. Phase I/II trials with recombinant human IL-3 expressed in yeast have demonstrated promising effects in patients with various hematological conditions .

The clinical research findings regarding IL-3's effects on hematopoiesis include:

  • Dose-Dependent Platelet Response:

    • Subcutaneous administration of rhIL-3 at dosages between 30 and 500 μg/m² for 15 consecutive days resulted in a dose-dependent increase in platelet counts in patients with normal hematopoiesis .

    • This effect is particularly relevant for research involving thrombocytopenia models and potential therapies.

  • Multi-lineage Stimulation:

    • Administration of rhIL-3 led to substantial increases in circulating neutrophils, eosinophils, monocytes, and lymphocytes .

    • Erythropoiesis was less stimulated, with increased hemoglobin concentration observed only in a minority of patients .

  • Secondary Hematopoietic Failure Recovery:

    • In research models of secondary hematopoietic failure due to prolonged chemo/radiotherapy or bone marrow infiltration by tumor cells, IL-3 treatment leads to clinically significant restoration of hematopoiesis .

    • This finding suggests research applications in developing supportive therapies for cancer treatment-induced myelosuppression.

  • Myelodysplastic Syndrome Response:

    • IL-3 has been shown to improve neutrophil and platelet counts in patients with myelodysplastic syndromes .

    • This indicates potential research directions for investigating dysregulated hematopoiesis mechanisms.

The safety profile observed in clinical research shows predominantly mild adverse effects at clinically used dosages, including fever, bone pain, headache, and stiffness of the neck . Researchers should note that transient thrombocytopenia has been observed in some patients with myelodysplastic syndrome or aplastic anemia treated at higher dosages (250-500 μg/m²) .

How does IL-3 influence microglial function in neurodegenerative disease models?

Recent research has uncovered a surprising and significant role for IL-3 in neuroinflammatory processes, particularly in Alzheimer's disease (AD) models . While IL-3 was previously associated primarily with hematopoietic regulation, findings now demonstrate that it plays a protective role in neurodegenerative conditions through microglial modulation.

The mechanistic pathway by which IL-3 affects microglial function in AD models involves several key processes:

  • Receptor Upregulation in Response to Amyloid:

    • Microglia dramatically increase expression of the IL-3Rα receptor specifically when they encounter amyloid plaques .

    • This upregulation appears to be part of a protective response mechanism to neuronal damage.

  • Enhancement of TREM2 Signaling:

    • IL-3 strengthens signaling downstream of TREM2, a critical cell-surface receptor that evokes microglial changes in AD .

    • This enhancement potentially improves microglial phagocytic capacity and amyloid clearance.

  • Neuroprotective Effects in Amyloidosis Models:

    • Experimental evidence from mouse models demonstrates that knocking out IL-3 leads to increased plaque size and number .

    • IL-3-deficient mice show impaired performance in memory tasks such as water maze tests, suggesting cognitive deficits related to amyloid accumulation .

To effectively study IL-3's role in neuroinflammation, researchers should consider the following methodological approaches:

  • Primary Microglial Culture Systems: Evaluate IL-3 receptor expression changes upon exposure to aggregated Aβ peptides

  • Brain Slice Culture Models: Assess IL-3-dependent microglial migration toward plaques

  • Transgenic AD Mouse Models: Compare amyloid clearance capacity in IL-3 wild-type versus knockout backgrounds

  • TREM2 Signaling Analysis: Measure downstream phosphorylation events in microglial cells with and without IL-3 stimulation

What is the source of IL-3 in the brain and how does this influence experimental design?

A particularly intriguing discovery about IL-3 in the central nervous system is its cellular source. Contrary to the expected immune cell origin, research has identified astrocytes as the primary producers of IL-3 in the brain . This finding has important implications for experimental design when studying neuroinflammatory processes.

The astrocytic origin of IL-3 presents several considerations for neuroinflammatory research:

  • Specialized Astrocyte Subpopulation:

    • Only a small subset of astrocytes constitutively produces IL-3 in the brain .

    • This specialized production suggests a dedicated neuroprotective function rather than a generalized inflammatory response.

  • Astrocyte-Microglia Communication Axis:

    • The production of IL-3 by astrocytes and reception by microglia establishes a novel intercellular communication pathway in the brain.

    • This axis appears particularly important for coordinating responses to pathological protein aggregation.

  • Implications for Experimental Models:

    • When designing experiments to study IL-3 in neurodegeneration, researchers must account for this unique cellular source.

    • Cell-specific deletion approaches (e.g., using Cre-lox systems with astrocyte-specific promoters) would be more informative than global knockouts.

    • Co-culture systems should incorporate both astrocytes and microglia to preserve this natural signaling mechanism.

  • Methodological Approaches to Study This Pathway:

    • Single-cell RNA sequencing to identify the specific astrocyte subpopulation producing IL-3

    • Conditional knockout models targeting IL-3 production specifically in astrocytes

    • 3D organoid models incorporating both cell types to better recapitulate the natural microenvironment

    • Spatial transcriptomics to map IL-3 producing astrocytes in relation to amyloid plaques

This astrocyte-microglia IL-3 signaling represents a promising therapeutic target pathway for neurodegenerative diseases, suggesting that enhancing IL-3 signaling might improve microglial amyloid clearance functions .

How does IL-3 receptor signaling influence T cell function in inflammatory bowel disease?

Recent research has revealed unexpected roles for IL-3 in regulating chronic intestinal inflammation, with particular relevance to inflammatory bowel disease (IBD) . While earlier studies had not addressed IL-3's role in IBD, current findings demonstrate that IL-3 receptor signaling exerts important regulatory functions at the interface of T cell biophysical properties and migration patterns.

The mechanisms by which IL-3 receptor signaling affects T cell function in the context of IBD include:

  • T Cell Cytoskeletal Modulation:

    • IL-3 receptor signaling induces changes in kinase phosphorylation that alter actin cytoskeleton structure .

    • These structural modifications directly impact T cell mechanical deformability and migratory capacity.

  • Regulatory T Cell (Treg) Trafficking:

    • IL-3 signaling enhances the egress of Tregs from inflamed colon mucosa .

    • In the absence of IL-3 or IL-3 receptor signaling, experimental colitis is exacerbated due to altered Treg trafficking.

  • IL-3 Expression in IBD Patients:

    • Clinical samples from IBD patients show increased levels of IL-3 in inflamed mucosa .

    • This elevation suggests an adaptive response attempting to limit inflammation.

  • Human Treg Mechanobiology:

    • IL-3 controls mechanobiological properties in human Tregs, similar to observations in mouse models .

    • Increased mucosal Treg abundance in IBD patients is associated with IL-3 signaling.

For researchers studying IL-3 in inflammatory conditions, several methodological approaches are valuable:

  • Mechanical Testing of T Cells: Employ real-time deformability cytometry and atomic force microscopy to quantify IL-3's effects on T cell mechanical properties .

  • Structural Analysis: Use scanning electron microscopy to visualize cytoskeletal changes induced by IL-3 signaling .

  • Dynamic Imaging: Apply fluorescence recovery after photobleaching (FRAP) to assess cytoskeletal dynamics .

  • Migration Assays: Implement both in vitro and in vivo cell trafficking assays to track Treg movement in response to IL-3 .

What are the methodological considerations for studying IL-3 signaling pathways?

Investigating IL-3 signaling pathways requires precise methodological approaches to capture the complexity of downstream effects across different cell types. IL-3's pleiotropic nature necessitates careful experimental design to distinguish direct from indirect effects and to capture the temporal dynamics of signaling events.

Key methodological considerations include:

  • Receptor Complex Analysis:

    • IL-3 receptor is heterodimeric, composed of a receptor-specific α chain and a common β chain shared with GM-CSF and IL-5 receptors .

    • Flow cytometry with fluorescently-labeled antibodies against both chains can quantify receptor expression levels.

    • Co-immunoprecipitation assays help verify receptor complex formation and identify associated adaptor proteins.

  • Signaling Pathway Delineation:

    • IL-3 activates multiple intracellular pathways, including Ras-Raf-ERK and JAK/STAT .

    • Western blotting for phosphorylated signaling proteins at various time points (typically 5, 15, 30, 60 minutes) after IL-3 stimulation provides temporal resolution.

    • Pathway-specific inhibitors (e.g., JAK inhibitors, MEK inhibitors) help establish causality between pathway activation and biological outcomes.

  • Transcriptional Response Profiling:

    • RNA-seq at multiple time points following IL-3 stimulation identifies primary and secondary transcriptional responses.

    • ChIP-seq for STAT factors can identify direct transcriptional targets.

    • IL-3 targets anti-apoptotic bcl-2 gene family members, which should be specifically assessed .

  • Cell Type Specificity Considerations:

    • Effects of IL-3 vary between cell types (hematopoietic progenitors, microglia, T cells).

    • Cell isolation techniques must be optimized to maintain receptor expression levels.

    • Consider cell-specific knockout models to distinguish autonomous from non-autonomous effects.

  • Experimental Controls for Recombinant IL-3 Studies:

    • Heat-inactivated IL-3 serves as a protein control to rule out non-specific effects.

    • Receptor-blocking antibodies confirm specificity of observed effects.

    • Dose-response relationships should be established (typically testing 0.1-100 ng/mL).

How can IL-3 be integrated into iPSC differentiation protocols for myeloid lineages?

Recombinant human IL-3 plays a pivotal role in directed differentiation of induced pluripotent stem cells (iPSCs) toward myeloid lineages. Optimizing IL-3 utilization in these protocols requires careful consideration of timing, concentration, and combinatorial cytokine approaches.

Methodological framework for IL-3 integration in iPSC differentiation:

  • Temporal Staging of IL-3 Application:

    • Early stage (days 0-5): IL-3 serves as an early regulator for iPSC differentiation into myeloid progenitors .

    • Mid-stage (days 5-12): Continued IL-3 exposure supports expansion of committed progenitors.

    • Late stage (beyond day 12): Combinatorial cytokine treatment with lineage-specific factors directs terminal differentiation.

  • Optimal Concentration Determination:

    • Concentration typically ranges from 10-50 ng/mL for most applications.

    • Titration experiments are recommended for each iPSC line to determine optimal dose.

    • Higher concentrations (50-100 ng/mL) may be needed for initial commitment, followed by lower maintenance doses.

  • Combinatorial Cytokine Strategies:

    • General myeloid progenitors: IL-3 + SCF + FLT3L

    • Dendritic cells: Initial IL-3, followed by GM-CSF and IL-4

    • Macrophages: Initial IL-3, followed by M-CSF

    • Erythrocytes: IL-3 + IL-12

    • Megakaryocytes: IL-3 + IL-6

  • Quality Control Metrics:

    • Flow cytometric analysis of lineage markers at each differentiation stage

    • Functional assays specific to target cell types

    • Comparison to primary cell controls for validation

The integration of IL-3 into differentiation protocols requires careful optimization, but when properly implemented, enables efficient generation of diverse myeloid lineages from iPSCs for disease modeling, drug screening, and regenerative medicine applications.

What troubleshooting approaches can address common challenges in IL-3-dependent experimental systems?

Researchers working with IL-3 in experimental systems frequently encounter challenges that can affect reproducibility and interpretation of results. The following troubleshooting framework addresses common issues and provides methodological solutions:

  • Inconsistent Biological Activity:

    • Problem: Variation in cellular responses to IL-3 between experiments.

    • Diagnostic Approach: Verify protein activity using TF-1 proliferation assay as a standardized readout .

    • Solution: Standardize reconstitution procedures, prepare single-use aliquots, and use carrier protein for dilute solutions to prevent adsorption to tubes and loss of activity.

  • Cell Type-Specific Response Variation:

    • Problem: Different cell populations showing variable sensitivity to IL-3.

    • Diagnostic Approach: Flow cytometric analysis of IL-3 receptor expression levels.

    • Solution: Adjust IL-3 concentration based on receptor expression levels; consider priming cells with low-dose IL-3 (1-5 ng/mL) for 24 hours before experimental treatment.

  • Receptor Desensitization:

    • Problem: Diminished response to IL-3 during prolonged exposure.

    • Diagnostic Approach: Time-course analysis of receptor expression and signaling pathway activation.

    • Solution: Implement pulsed treatment regimens rather than continuous exposure; replenish IL-3 at 48-72 hour intervals.

  • Poor Reproducibility in Primary Cells:

    • Problem: Variable results when using primary cells from different donors.

    • Diagnostic Approach: Characterize baseline receptor expression and intrinsic responsiveness.

    • Solution: Pool cells from multiple donors for method development; include responder classification in experimental design; adjust IL-3 concentration based on donor-specific titration.

  • Unexpected Cross-Talk with Other Cytokines:

    • Problem: Altered IL-3 effects when used in combination with other factors.

    • Diagnostic Approach: Systematic analysis of pathway activation with single vs. combined cytokines.

    • Solution: Stagger cytokine addition with temporal separation (6-24 hours); adjust relative concentrations to achieve optimal synergy.

By implementing these methodological approaches, researchers can enhance the reliability and reproducibility of IL-3-dependent experimental systems, leading to more robust and translatable research outcomes.

What emerging applications of IL-3 show promise for translational research?

Recent discoveries about IL-3's functions beyond hematopoiesis are opening new avenues for translational research. Several promising directions warrant further investigation:

  • Neurodegenerative Disease Therapeutics:

    • The discovery that IL-3 protects against Alzheimer's disease through microglial modulation suggests therapeutic potential .

    • Research could focus on developing targeted IL-3 delivery systems to cross the blood-brain barrier and enhance microglial amyloid clearance.

    • Investigation of the specific astrocyte subpopulation that produces IL-3 could yield insights for cell-based therapies .

  • Inflammatory Bowel Disease Interventions:

    • IL-3's role in regulating T cell trafficking and function in intestinal inflammation suggests potential for targeted immunotherapy .

    • The mechanical and migratory properties of T cells affected by IL-3 represent an entirely new therapeutic paradigm beyond traditional immunosuppression.

    • Development of locally-delivered IL-3 formulations could enhance Treg retention in inflamed intestinal tissues.

  • Advanced Cell Therapy Applications:

    • IL-3's capacity to expand and differentiate hematopoietic stem cells has implications for optimizing cell therapy products .

    • Research into IL-3 preconditioning of immune cells before adoptive transfer may enhance therapeutic efficacy.

    • IL-3's synergistic effects with other cytokines warrant investigation for ex vivo cell manufacturing protocols.

  • Biomarker Development:

    • Correlations between IL-3 levels and disease states suggest potential diagnostic applications.

    • Further research could establish whether circulating IL-3 levels or receptor expression patterns predict treatment responses in inflammatory or hematologic conditions.

These emerging applications highlight the importance of continuing fundamental research on IL-3 biology while simultaneously exploring translational opportunities in multiple disease contexts.

How might genetic modification of IL-3 or its receptor enhance research applications?

Genetic engineering approaches targeting IL-3 or its receptor components offer powerful tools to enhance research applications and potentially develop novel therapeutics. Several strategic modifications show particular promise:

  • Receptor-Specific Variants:

    • Engineering IL-3 variants with altered binding characteristics for the α or β chains could create signaling-biased ligands.

    • Such modifications might selectively activate certain downstream pathways (e.g., preferentially activating JAK/STAT over Ras-Raf-ERK).

    • Methodological approach: Structure-guided mutagenesis targeting receptor interface residues, followed by signaling pathway analysis.

  • Fusion Proteins for Enhanced Functionality:

    • IL-3-antibody fusion constructs could direct the cytokine's activity to specific cell populations.

    • For neurodegenerative disease applications, fusion with brain-targeting peptides could enhance blood-brain barrier penetration.

    • IL-3-fluorescent protein fusions would enable real-time visualization of binding dynamics and trafficking.

  • Conditional Expression Systems:

    • Development of inducible IL-3 or IL-3R expression vectors for temporal control in experimental systems.

    • Cell type-specific promoters driving IL-3 expression could mimic physiological production patterns.

    • CRISPR/Cas9-mediated knock-in of reporter genes at IL-3 or IL-3R loci would allow monitoring of endogenous expression patterns.

  • Receptor Engineering:

    • Creating chimeric receptors incorporating the IL-3R signaling domain with alternative extracellular domains.

    • Engineering synthetic receptors responsive to small molecules but signaling through IL-3R pathways.

    • Developing dominant-negative IL-3R variants for pathway inhibition studies.

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.