IL 3 Rat

Interleukin-3 Rat Recombinant
Shipped with Ice Packs
In Stock

Description

Biological Functions

Rat IL-3 exerts pleiotropic effects on hematopoietic and immune cells:

  • Hematopoiesis: Stimulates proliferation and differentiation of pluripotent stem cells into myeloid lineages (neutrophils, macrophages, megakaryocytes) .

  • Mast Cell Regulation: Synergizes with stem cell factor (SCF) to enhance histamine production and mast cell protease II (RMCP-II) expression in bone marrow-derived mast cells (BMMCs) .

  • Microglial Activation: Autocrine production by rat microglia promotes proliferation and immune recruitment in the central nervous system .

Key Functional Pathways

  • JAK2/STAT5: Primary signaling pathway for survival and anti-apoptotic effects .

  • Ras Pathway: Suppresses apoptosis in progenitor cells .

Table 1: Key Preclinical Studies on Rat IL-3

Study FocusMethodologyKey ResultsReference
Microglial ProliferationIL-3 mRNA detection via PCRMicroglia self-produce IL-3, enhancing proliferation via autocrine signaling .
Mast Cell DifferentiationBMMC culture with IL-3 + SCFCombined cytokines increased histamine content by 300% over 21 days .
Receptor Binding AnalysisChimeric IL-3 proteins + mAb mappingIdentified N- and C-terminal helices as critical receptor interaction sites .

Clinical and Therapeutic Insights

  • Post-Chemotherapy Recovery: Accelerates platelet and granulocyte regeneration in preclinical models .

  • Limitations: Monotherapy showed limited efficacy in myelodysplastic syndromes but synergizes with GM-CSF and SCF for stem cell mobilization .

  • Neuroimmunology: Microglial IL-3 may compensate for weak T-cell responses in brain lesions .

Future Directions

  • Targeted Therapies: Engineering IL-3 variants with enhanced receptor specificity .

  • Neuroinflammatory Disease: Exploring IL-3’s role in Alzheimer’s and multiple sclerosis .

Product Specs

Introduction
Interleukin-3 (IL-3) is a cytokine with multiple functions, mainly produced by activated T cells. It is believed to interact with specific receptors on cell surfaces, promoting the growth, specialization, and survival of hematopoietic cell lines. Additionally, IL-3 has demonstrated influence over the activity of other cell types, such as mast cells, eosinophils, megakaryocytes, and basophils.
Description
Recombinant Rat Interleukin-3, produced in E. coli, is a single-chain polypeptide. It is non-glycosylated and consists of 144 amino acids, with a molecular weight of 16.3 kDa. The purification of IL-3 is carried out using proprietary chromatographic methods.
Physical Appearance
The product appears as a sterile, white powder that has been lyophilized (freeze-dried).
Formulation
The lyophilization process for Rat Interleukin-3 was performed using a concentrated solution in PBS at a pH of 7.4, which was previously filtered through a 0.2µm filter.
Solubility
For reconstitution of the lyophilized Interleukin-3, it is advised to use sterile 18MΩ-cm H2O at a concentration not lower than 100µg/ml. This solution can be further diluted using other aqueous solutions.
Stability
Lyophilized Interleukin-3 remains stable at room temperature for a period of 3 weeks. However, for optimal storage, it should be kept desiccated at a temperature below -18°C. After reconstitution, it is recommended to store IL3 at 4°C for a duration of 2-7 days. For long-term storage, freezing at -18°C is advisable. To enhance stability during long-term storage, consider adding a carrier protein like 0.1% HSA or BSA. Repeated freezing and thawing should be avoided.
Purity
The purity level is determined to be greater than 97.0% using the following methods: (a) Analysis by Reverse-Phase High-Performance Liquid Chromatography (RP-HPLC). (b) Analysis by Sodium Dodecyl Sulfate-Polyacrylamide Gel Electrophoresis (SDS-PAGE).
Biological Activity
The ED50, determined by the dose-dependent stimulation of thymidine uptake in murine MC-9 cells, is less than 10 ng/ml. This corresponds to a specific activity exceeding 1.0 x 10^5 units/mg.
Synonyms
MCGF (Mast cell growth factor), Multi-CSF, HCGF, P-cell stimulation factor, IL-3, MGC79398, MGC79399.
Source
Escherichia Coli.
Amino Acid Sequence
MISDRGSDAH HLLRTLDCRT IALEILVKLP YPQVSGLNNS DDKANLRNST LRRVNLDEFL KSQEEFDSQD TTDIKSKLQK LKCCIPAAAS DSVLPGVYNK DLDDFKKKLR FYVIHLKDLQ PVSVSRPPQP TSSSDNFRPM TVEC.

Q&A

What is the optimal concentration of recombinant rat IL-3 for bone marrow-derived mast cell cultures?

For rat bone marrow-derived mast cell cultures, the optimal concentration of recombinant rat IL-3 typically ranges from 10-20 ng/mL. This concentration has been demonstrated to effectively support mast cell development and proliferation when culturing bone marrow cells for 4-6 weeks. In standard protocols, bone marrow cells are cultured in medium containing IL-3 (often derived from WEHI-3-conditioned medium or recombinant sources) until the cultures contain >98% mast cells, as verified by flow cytometry for c-Kit and FcεRI expression . It is advisable to titrate the concentration for your specific cell population, as requirements may vary slightly based on the rat strain and experimental objectives.

How do rat and mouse IL-3 differ in their biological activities and cross-reactivity?

Rat and mouse IL-3 share approximately 54% amino acid sequence homology but exhibit significant differences in their cross-reactivity. While mouse IL-3 shows limited activity on rat cells, rat IL-3 demonstrates substantial cross-reactivity with mouse cells. This asymmetrical cross-reactivity is particularly important when designing experiments using conditioned media or when comparing results across species. For studies specifically requiring rat IL-3 activity, it is recommended to use species-specific recombinant proteins rather than cross-species application, as the potency and specificity of biological responses can vary significantly . When using WEHI-3-conditioned medium as a source of IL-3, researchers should be aware that this mouse-derived cell line produces mouse IL-3, which may have reduced efficacy in rat cell systems.

What is the recommended protocol for generating rat bone marrow-derived mast cells using IL-3?

The recommended protocol for generating rat bone marrow-derived mast cells (BMCMCs) involves:

  • Harvest bone marrow cells from rat femurs and tibias under aseptic conditions

  • Remove red blood cells using hypotonic lysis buffer

  • Culture bone marrow cells at a density of 5×10⁵ cells/mL in complete medium supplemented with 10-20 ng/mL recombinant rat IL-3 or 10-20% WEHI-3-conditioned medium

  • Incubate at 37°C with 5% CO₂

  • Replace half the medium with fresh IL-3-containing medium twice weekly

  • After 4-6 weeks, verify mast cell purity (>98%) via flow cytometry for c-Kit and FcεRI expression

This protocol consistently yields homogeneous populations of functional mast cells suitable for further experimentation . For optimal results, maintaining consistent IL-3 levels throughout the culture period is essential, as fluctuations can affect cell differentiation and functional characteristics.

How can I verify the purity and functionality of rat mast cells cultured with IL-3?

Verification of rat mast cell purity and functionality after IL-3-dependent culture should include multiple complementary approaches:

  • Phenotypic characterization: Flow cytometry analysis for expression of c-Kit (CD117) and FcεRI, which should be >98% positive in mature cultures

  • Morphological assessment: Toluidine blue or Alcian blue/safranin staining to visualize metachromatic granules

  • Functional assays:

    • β-hexosaminidase release assay following IgE sensitization and allergen challenge

    • Cytokine production (IL-4, IL-6, IL-13) in response to various stimuli

    • Calcium flux measurements after activation

  • Molecular verification: RT-PCR for mast cell-specific proteases and receptors

A fully functional rat mast cell population should respond to IgE-mediated activation with degranulation and cytokine production. Researchers should establish baseline responses for their specific cell populations, as functional parameters can vary based on culture conditions and rat strain .

How does the signaling pathway of IL-3 in rat mast cells differ from other species, and what are the implications for experimental design?

IL-3 signaling in rat mast cells follows the canonical Jak/STAT pathway but exhibits species-specific differences in downstream effector activation:

  • The rat IL-3 receptor complex shows higher constitutive association with Jak2 compared to mouse counterparts

  • Rat mast cells demonstrate stronger and more sustained STAT5 phosphorylation following IL-3 stimulation

  • PI3K/Akt pathway activation by IL-3 in rat cells exhibits differential kinetics compared to mouse cells

These differences necessitate careful experimental design considerations:

  • Timing considerations: Peak signaling events may occur at different time points in rat versus mouse or human systems

  • Inhibitor concentrations: May require adjustment when translating protocols from other species

  • Readout selection: Optimal downstream targets for monitoring IL-3 activity may differ

Researchers should validate signaling pathway kinetics in their specific rat model rather than directly applying parameters established in mouse systems. When measuring phosphorylation events, it is advisable to perform detailed time-course experiments specific to rat cells rather than assuming conserved kinetics across species .

What cross-talk exists between IL-3 and TIM family receptors in rat immune cells, and how can this be experimentally manipulated?

IL-3 and T-cell immunoglobulin and mucin domain (TIM) family proteins engage in significant cross-talk in rat immune cells, particularly in mast cells and basophils:

  • TIM-1 and TIM-3 (but not TIM-2 or TIM-4) are constitutively expressed on rat peritoneal mast cells and bone marrow-derived cultured mast cells (BMCMCs)

  • IL-3 signaling influences the expression patterns of TIM proteins: IgE+antigen stimulation downregulates TIM-1 expression while upregulating TIM-3 on BMCMCs

  • Reciprocally, TIM-1 and TIM-3 activation enhances IL-3-dependent cytokine production, particularly IL-4, IL-6, and IL-13

This cross-talk can be experimentally manipulated through:

  • Antibody-mediated approaches: Using anti-TIM-3 polyclonal antibodies (20 μg/mL) to enhance cytokine production without affecting degranulation in IL-3-cultured mast cells

  • Recombinant protein modulation: Applying recombinant mouse TIM-4 (20 μg/mL), a ligand for TIM-1, to promote Th2 cytokine production in mast cells

  • Genetic approaches: Selective knockdown of TIM family members to isolate their contributions to IL-3-dependent responses

The timing of such manipulations is critical, as TIM receptor expression is dynamically regulated following activation. For optimal results, experimental designs should account for both the baseline expression patterns and activation-induced changes in receptor expression .

How can contradictory data between in vitro IL-3 effects on rat cells and in vivo observations be reconciled methodologically?

Reconciling contradictions between in vitro IL-3 effects and in vivo observations in rat models requires systematic methodological approaches:

  • Concentration discrepancies: In vitro studies typically use consistent, high IL-3 concentrations (10-20 ng/mL), while in vivo concentrations fluctuate and are often much lower. Solution: Conduct dose-response experiments using physiologically relevant IL-3 concentrations determined from in vivo measurements.

  • Temporal differences: In vitro systems often examine acute responses, while in vivo effects may develop over longer timeframes. Solution: Design time-course experiments spanning hours to days to better capture the full spectrum of responses.

  • Microenvironmental factors: In vivo, IL-3 acts within complex tissue microenvironments containing multiple cell types and signaling molecules. Solution: Develop more sophisticated co-culture systems incorporating stromal cells and other relevant cell types.

  • Compensatory mechanisms: In vivo systems may engage compensatory pathways absent in reductionist in vitro models. Solution: Use selective inhibitors or genetic approaches to block compensatory pathways in vivo.

  • Strain-dependent variations: Different rat strains can exhibit distinct IL-3 response patterns. Solution: Validate key findings across multiple rat strains (e.g., Sprague-Dawley, Wistar, Lewis) to identify strain-specific versus conserved responses.

When contradictions persist despite these approaches, researchers should consider that such differences may reflect genuine biological complexities rather than methodological artifacts, and these differences themselves may provide valuable insights into IL-3 biology .

What methodological approaches can detect IL-3-dependent epigenetic modifications in rat immune cells?

Detection of IL-3-dependent epigenetic modifications in rat immune cells requires specialized methodological approaches:

  • Chromatin Immunoprecipitation (ChIP) for histone modifications:

    • Optimize fixation time (typically 10-15 minutes with 1% formaldehyde) specifically for rat cells

    • Use antibodies validated for rat histones to assess H3K4me3 (activation mark) and H3K27me3 (repressive mark)

    • Include IL-3-responsive genes (e.g., GATA-2, PU.1) as positive controls

  • DNA methylation analysis:

    • Bisulfite sequencing of promoter regions for IL-3-responsive genes

    • Genome-wide approaches such as reduced representation bisulfite sequencing (RRBS)

    • Compare methylation patterns before and after IL-3 exposure at multiple time points (6h, 24h, 72h)

  • Accessibility assays:

    • ATAC-seq optimized for rat immune cells (100,000-50,000 cells)

    • DNase-seq to map open chromatin regions

    • Footprinting analysis to identify transcription factor binding sites

  • Functional validation:

    • Use epigenetic inhibitors (e.g., DNA methyltransferase inhibitors, histone deacetylase inhibitors) to confirm causative relationships

    • Perform time-course experiments to establish the sequence of epigenetic events following IL-3 stimulation

What are the optimal storage and handling protocols for maintaining rat IL-3 bioactivity?

Maintaining the bioactivity of rat IL-3 requires strict adherence to proper storage and handling protocols:

  • Storage conditions:

    • Store lyophilized recombinant rat IL-3 at -20°C to -80°C

    • After reconstitution, prepare single-use aliquots to avoid freeze-thaw cycles

    • Store reconstituted IL-3 at -80°C for long-term storage (up to 6 months)

    • For short-term use (1-2 weeks), 4°C storage is acceptable if preserved with 0.1% BSA

  • Reconstitution recommendations:

    • Reconstitute in sterile PBS or balanced salt solution

    • For low concentrations, include carrier protein (0.1-0.5% BSA) to prevent adsorption to tubes

    • Avoid introducing bubbles through gentle pipetting rather than vortexing

    • Filter sterilize using low protein-binding 0.22 μm filters

  • Working solution preparation:

    • Prepare fresh working solutions for each experiment when possible

    • Maintain sterile technique throughout handling

    • Use polypropylene tubes rather than glass or polystyrene to minimize protein adsorption

    • Keep solutions on ice during experiment preparation

  • Bioactivity monitoring:

    • Periodically verify activity using proliferation assays with IL-3-dependent cell lines

    • Include positive controls (freshly reconstituted IL-3) alongside stored aliquots

Following these protocols ensures consistent bioactivity across experiments. Researchers should document lot numbers, reconstitution dates, and storage conditions for all IL-3 preparations to facilitate troubleshooting if unexpected results occur .

How can researchers effectively troubleshoot failed IL-3-dependent rat cell cultures?

When troubleshooting failed IL-3-dependent rat cell cultures, researchers should systematically evaluate multiple parameters:

  • IL-3 source and quality:

    • Test IL-3 bioactivity using a known responsive cell line

    • Verify protein concentration using quantitative methods

    • Check for proper storage conditions and freeze-thaw history

    • Consider using an alternative lot or source of IL-3

  • Cell isolation and viability issues:

    • Assess initial bone marrow cell viability (should be >90%)

    • Verify proper red blood cell lysis procedure

    • Check for bacterial or fungal contamination

    • Evaluate cell density (optimal: 5×10⁵ cells/mL initially)

  • Culture conditions:

    • Confirm incubator parameters (37°C, 5% CO₂, humidity)

    • Assess media quality (pH, color, particulates)

    • Verify that all supplements (glutamine, antibiotics, serum) are fresh and correctly added

    • Ensure regular feeding schedule (every 2-3 days)

  • Rat strain considerations:

    • Some rat strains may require strain-specific protocol modifications

    • Consider genetic background effects on IL-3 responsiveness

    • Age of rats can affect bone marrow cell quality (optimal: 6-12 weeks old)

  • Technical execution:

    • Maintain strict aseptic technique throughout the process

    • Use appropriate culture vessels (untreated plastic for suspension cultures)

    • Ensure gentle handling during feeding and passaging

If cultures still fail after addressing these issues, establishing a side-by-side comparison with a colleague's successful protocol can help identify subtle but critical differences in technique or reagents .

What specialized flow cytometry protocols best identify IL-3-responsive rat cell populations?

Specialized flow cytometry protocols for identifying IL-3-responsive rat cell populations should incorporate multiple parameters:

  • Surface marker panel:

    • Primary markers: c-Kit (CD117), FcεRI, CD34, IL-3Rα (CD123)

    • Lineage markers: CD11b, Gr-1, B220, CD3, Ter119 (for negative selection)

    • Maturation markers: CD45, CD49b, ST2, CD16/32

    • Use fluorochrome combinations optimized for rat cells to minimize spectral overlap

  • Functional readouts:

    • Phospho-flow analysis of STAT5 phosphorylation after IL-3 stimulation (15-30 min)

    • Intracellular cytokine staining following 4-6 hour stimulation with PMA/ionomycin

    • Mitochondrial activity assessment using MitoTracker probes

    • Cell cycle analysis using EdU incorporation or DNA content staining

  • Sample preparation optimization:

    • Use enzymatic dissociation methods optimized for rat tissues

    • Include viability dyes (e.g., 7-AAD or fixable viability dyes)

    • Optimize fixation and permeabilization protocols for intracellular markers

    • Include unstained, single-stained, and FMO controls

  • Gating strategy:

    • Initial gating on viable, single cells

    • Exclude lineage-positive cells

    • Gate on c-Kit+ and/or CD123+ populations

    • Further refine using functional response to IL-3 stimulation

  • Data analysis approaches:

    • Consider high-dimensional analysis methods (tSNE, UMAP) for complex populations

    • Use proliferation tracking dyes to identify highly responsive subpopulations

    • Apply kinetic analysis for time-dependent responses

This comprehensive approach allows identification of not only canonical IL-3-responsive cells but also novel or rare responsive populations that might be missed with more limited panels .

How does the molecular structure of rat IL-3 impact its interaction with the IL-3 receptor compared to other species?

The molecular structure of rat IL-3 contains several species-specific features that impact its receptor interactions:

  • Structural differences:

    • Rat IL-3 contains 147 amino acids compared to 140 in mouse IL-3

    • Four-helix bundle structure with rat-specific residues in the AB and CD loops

    • Contains 3 conserved disulfide bonds that are critical for biological activity

    • N-terminal region shows higher variability compared to human and mouse orthologs

  • Receptor binding characteristics:

    • Rat IL-3 binds to a heterodimeric receptor composed of an IL-3-specific α chain (CD123) and a common β chain shared with GM-CSF and IL-5 receptors

    • Binding affinity (KD) for rat IL-3Rα is approximately 10⁻⁸ M, which is stronger than mouse-to-rat cross-species binding

    • The higher affinity is attributed to specific residues in the D-helix of rat IL-3

  • Species cross-reactivity mechanism:

    • Rat IL-3 shows significant activity on mouse cells due to conserved residues in site 2 (βc binding interface)

    • Mouse IL-3 shows minimal activity on rat cells due to divergent residues in site 1 (α chain interface)

    • This asymmetrical cross-reactivity is notable when comparing with other species pairs

  • Structural implications for experimental approaches:

    • Antibodies against mouse IL-3 may not effectively neutralize rat IL-3 due to structural differences

    • When designing IL-3 antagonists, species-specific approaches are necessary

    • Fusion proteins incorporating rat IL-3 require careful linker design to preserve bioactivity

Understanding these structural details helps researchers select appropriate reagents and interpret cross-species data correctly. For studies requiring precise manipulation of IL-3 signaling, species-matched reagents are strongly recommended .

What are the most effective gene editing approaches for studying IL-3 function in rat models?

Several gene editing approaches have proven effective for studying IL-3 function in rat models, each with specific advantages:

  • CRISPR/Cas9 system:

    • Most efficient for complete IL-3 or IL-3Rα knockout models

    • Optimal design includes dual gRNAs targeting critical exons (exons 1-3)

    • Delivery methods:

      • Embryo microinjection for germline modification

      • Lentiviral vectors for cell-specific targeting

      • Ribonucleoprotein (RNP) complexes for primary cell editing

    • Verification requires both sequencing and functional assays

  • Base editing approaches:

    • Suitable for introducing point mutations in regulatory regions

    • Allows study of specific amino acid contributions to IL-3 function

    • Reduced off-target effects compared to standard CRISPR/Cas9

    • Particularly effective for modifying glycosylation sites without disrupting protein expression

  • Conditional systems:

    • Cre-loxP approaches for tissue-specific or inducible IL-3 deletion

    • Tetracycline-inducible systems for temporal control of IL-3 expression

    • Particularly valuable for distinguishing developmental versus acute roles

  • RNA interference:

    • shRNA delivered via lentiviral vectors for stable knockdown

    • siRNA for transient suppression in primary cells

    • Valuable for dose-dependent studies of IL-3 function

  • Methodological considerations:

    • For IL-3 receptor studies, target the specific α chain rather than the shared βc

    • Include appropriate controls for off-target effects

    • Validate editing efficiency at both genomic and protein levels

    • Consider compensatory upregulation of related cytokines

When selecting an approach, researchers should consider the specific research question, required precision of genetic manipulation, and downstream analytical methods. For complex phenotypes, creating multiple models using different gene editing strategies can provide complementary insights and increase confidence in observed effects .

What advanced analytical methods can quantify IL-3-induced changes in rat cell signaling networks?

Advanced analytical methods for quantifying IL-3-induced signaling networks in rat cells include:

  • Mass spectrometry-based phosphoproteomics:

    • SILAC or TMT labeling to compare stimulated versus unstimulated cells

    • Enrichment of phosphopeptides using TiO₂ or IMAC

    • Data-independent acquisition (DIA) for increased coverage

    • Bioinformatic analysis using pathway enrichment tools specific for rat proteins

  • Advanced imaging techniques:

    • Live-cell FRET biosensors to monitor kinase activities in real-time

    • Lattice light-sheet microscopy for 3D visualization of signaling dynamics

    • Single-molecule tracking to analyze receptor clustering and diffusion

    • Optimized immunofluorescence protocols for rat cells with validated antibodies

  • Single-cell approaches:

    • scRNA-seq to identify transcriptional responses across heterogeneous populations

    • CITE-seq for simultaneous surface protein and transcript analysis

    • Single-cell proteomics using microfluidic platforms

    • Computational integration of multi-omic data sets

  • Network analysis methods:

    • Bayesian network modeling of time-course data

    • Partial least squares regression to identify key network nodes

    • Boolean network models to predict signaling outcomes

    • Differential equation-based models for quantitative descriptions

  • Validation approaches:

    • Pharmacological inhibition at multiple nodes in the network

    • Genetic perturbation using CRISPR interference or activation

    • Cross-validation across multiple rat strains

    • Integration of in vitro and in vivo observations

These methods are most powerful when applied in combination and integrated through computational approaches. Researchers should design experiments with appropriate time points (ranging from minutes to hours) to capture both immediate signaling events and downstream network adaptations following IL-3 stimulation .

How do findings from rat IL-3 studies translate to human systems, and what methodological approaches improve translational relevance?

Translating findings from rat IL-3 studies to human systems requires careful methodological considerations:

  • Comparative receptor biology:

    • Rat IL-3 shares approximately 29% amino acid identity with human IL-3

    • The IL-3 receptor complex structure is conserved, but binding kinetics differ

    • Species-specific differences in downstream signaling intensity and duration exist

    • Methodological approach: Parallel signaling studies in rat and human cells using phospho-flow cytometry with cross-validated antibodies

  • Functional conservation assessment:

    • Many IL-3 functions are conserved (hematopoiesis, mast cell development), but quantitative differences exist

    • Rat cells generally show stronger proliferative responses to IL-3

    • Cytokine production profiles differ between species

    • Methodological approach: Comparative dose-response studies with standardized readouts

  • Translational experimental design:

    • Use humanized rat models where appropriate

    • Include human cell controls in key experiments

    • Validate key findings in human primary cells and tissues

    • Confirm pharmacological responses across species

    • Prioritize investigations of pathways with known conservation

  • Methodological refinements:

    • Develop cross-species reagents where possible

    • Use bioinformatic approaches to identify highly conserved regulatory elements

    • Apply systems biology approaches to map conserved network components

    • Design chimeric receptor studies to isolate species-specific components

  • Translational limitations awareness:

    • Acknowledge species differences in publications

    • Conduct literature-based meta-analyses of cross-species concordance

    • Use translational algorithms that account for species-specific factors

By incorporating these approaches, researchers can maximize the translational value of rat IL-3 studies while maintaining appropriate scientific caution about direct extrapolations to human biology .

What methodological controls should be included when studying IL-3 interactions with TIM family proteins and other immune regulators in rat models?

When studying IL-3 interactions with TIM family proteins and other immune regulators in rat models, comprehensive methodological controls are essential:

  • Antibody validation controls:

    • Include isotype-matched control antibodies at equivalent concentrations

    • Validate antibody specificity using knockout or knockdown cells

    • Test for cross-reactivity with related family members (e.g., TIM-1 antibodies against TIM-3)

    • Use multiple antibody clones targeting different epitopes when possible

  • Protein interaction controls:

    • Include recombinant protein-Fc fusions alongside native proteins

    • Test heat-inactivated proteins to control for non-specific effects

    • Use competition assays with soluble receptors to confirm specificity

    • Perform reciprocal co-immunoprecipitation experiments

  • Cellular response controls:

    • Compare effects on cells from multiple rat strains

    • Include time-matched unstimulated controls

    • Use cells with genetic modifications of key signaling components

    • Compare IL-3 effects with related cytokines (IL-5, GM-CSF) to identify specific versus shared responses

  • Specific TIM family interaction controls:

    • When studying TIM-1 and TIM-3 enhancement of Th2 cytokine production, include:

      • Anti-TIM-1 (20 μg/mL) and anti-TIM-3 (20 μg/mL) alongside isotype controls

      • Recombinant TIM-4 (20 μg/mL) as a TIM-1 ligand control

      • Medium-only controls to establish baseline cytokine production

      • Combined stimulation controls to assess additive or synergistic effects

  • Functional validation controls:

    • Include positive controls for maximal cell activation

    • Test multiple readouts (cytokine production, degranulation, apoptosis)

    • Validate in multiple cell types where receptors are co-expressed

    • Perform genetic knockdown validation of observed effects

These control strategies ensure that observed interactions are specific, reproducible, and biologically relevant, reducing the likelihood of misinterpreting artifacts or non-specific effects .

How can researchers design experiments to distinguish direct versus indirect effects of IL-3 on rat immune cell functions?

Designing experiments to distinguish direct versus indirect effects of IL-3 requires sophisticated experimental approaches:

  • Temporal resolution studies:

    • Implement high-resolution time courses (minutes to hours) to identify primary versus secondary responses

    • Use transcriptional inhibitors (e.g., actinomycin D) to block secondary gene expression

    • Apply protein synthesis inhibitors (e.g., cycloheximide) at various time points

    • Analyze signal propagation using phosphorylation kinetics

  • Cell purification and mixed culture approaches:

    • Conduct parallel experiments with highly purified populations and mixed cultures

    • Use transwell systems to separate cell populations while allowing soluble factor exchange

    • Implement cell-specific genetic modifications using targeted delivery systems

    • Apply single-cell analysis to heterogeneous populations

  • Receptor-specific approaches:

    • Use cells with genetic deletion or knockdown of IL-3Rα

    • Apply IL-3 receptor-blocking antibodies with careful titration

    • Implement receptor chimeras to isolate signaling components

    • Use mutant IL-3 variants with altered receptor binding properties

  • Secretome analysis:

    • Analyze conditioned media from IL-3-stimulated cells

    • Perform antibody-based neutralization of candidate mediators

    • Use mass spectrometry to identify secreted factors

    • Apply bioinformatic approaches to distinguish direct IL-3 targets from secondary mediators

  • Genetic approaches:

    • Implement rapid genetic modification systems (e.g., CRISPRi) to transiently suppress mediators

    • Use inducible expression systems to control timing of factor expression

    • Apply lineage-specific genetic modifications using Cre-loxP systems

    • Perform comparative studies between wild-type and receptor-deficient animals

These approaches are most powerful when implemented in combination, allowing researchers to triangulate between multiple lines of evidence. When designing such experiments, careful consideration should be given to timing, dosage, and potential compensatory mechanisms .

What statistical approaches are most appropriate for analyzing complex IL-3-dependent phenotypes in rat models?

Complex IL-3-dependent phenotypes in rat models require sophisticated statistical approaches for robust analysis:

  • Multivariate analysis methods:

    • Principal Component Analysis (PCA) to identify major sources of variation

    • Partial Least Squares Discriminant Analysis (PLS-DA) for group separation

    • MANOVA for assessing multiple dependent variables simultaneously

    • Hierarchical clustering to identify patterns across multiple parameters

  • Time series analysis approaches:

    • Mixed-effects models for longitudinal data with repeated measures

    • Functional data analysis for continuous temporal processes

    • Change-point detection methods to identify critical transition points

    • Area-under-curve calculations for cumulative response quantification

  • Dose-response modeling:

    • Four-parameter logistic regression for non-linear responses

    • Estimation of EC50 values with confidence intervals

    • Relative potency calculations across experimental conditions

    • Interaction analyses for combination treatments

  • Experimental design considerations:

    • Power analysis specifically calibrated for rat models (accounting for strain-specific variability)

    • Blocked designs to control for batch effects and animal variability

    • Latin square approaches for complex multi-factorial experiments

    • Sample size determination based on expected effect sizes from pilot studies

  • Advanced computational approaches:

    • Machine learning algorithms for pattern recognition

    • Network analysis for pathway interactions

    • Bayesian hierarchical modeling for integrating prior knowledge

    • Bootstrap and permutation tests for robust inference

When applying these methods, researchers should:

  • Pre-register analysis plans to avoid post-hoc bias

  • Consider correction for multiple testing (e.g., Benjamini-Hochberg procedure)

  • Report effect sizes alongside p-values

  • Provide complete methodological details to ensure reproducibility

  • Consider consulting with a biostatistician for complex experimental designs

How can researchers effectively integrate in vitro and in vivo rat IL-3 data to build comprehensive models of cytokine function?

Effective integration of in vitro and in vivo rat IL-3 data requires systematic methodological approaches:

  • Parallel experimental design:

    • Design matched in vitro and in vivo experiments with comparable endpoints

    • Use consistent IL-3 sources and concentrations when possible

    • Implement identical analysis methods for both systems

    • Include timing studies that reflect physiological kinetics

  • Translational parameters:

    • Identify and validate biomarkers that function in both systems

    • Develop pharmacokinetic/pharmacodynamic (PK/PD) models to relate in vitro concentrations to in vivo exposures

    • Measure IL-3 concentrations in relevant tissues to inform in vitro dosing

    • Characterize receptor expression profiles across systems

  • Integrative modeling approaches:

    • Develop multi-scale mathematical models incorporating cellular and organismal parameters

    • Use agent-based modeling to simulate tissue-level effects from cellular responses

    • Apply systems biology approaches to map network relationships

    • Implement machine learning to identify patterns across datasets

  • Validation strategies:

    • Test model predictions with targeted experiments

    • Perform interventional studies at equivalent doses across systems

    • Use genetic approaches consistently across in vitro and in vivo models

    • Apply emerging ex vivo systems (e.g., precision-cut tissue slices) as intermediate validation platforms

  • Data integration frameworks:

    • Develop standardized data structures for cross-system comparison

    • Implement dimension reduction techniques to identify core response patterns

    • Use Bayesian approaches to update in vitro-based predictions with in vivo data

    • Create visualization tools that present integrated datasets coherently

This integrated approach allows researchers to leverage the high control and mechanistic detail of in vitro systems alongside the physiological relevance of in vivo models, resulting in more comprehensive and predictive understanding of IL-3 biology .

Product Science Overview

Structure and Production

Recombinant rat IL-3 is typically produced in Escherichia coli (E. coli) and purified through sequential chromatography . The recombinant form is a single, non-glycosylated polypeptide chain containing 144 amino acids and has a molecular mass of approximately 16.3 kDa . The amino acid sequence of rat IL-3 includes several key regions that are essential for its biological activity .

Biological Activity

IL-3 is known for its ability to stimulate the proliferation and differentiation of various hematopoietic cell types. It exerts its effects by binding to specific receptors on the surface of target cells, leading to the activation of intracellular signaling pathways that promote cell growth and survival . The biological activity of recombinant rat IL-3 is often measured using a cell proliferation assay with NFS-60 mouse myelogenous leukemia lymphoblast cells .

Applications

Recombinant rat IL-3 is widely used in research to study its effects on hematopoietic cells and to explore its potential therapeutic applications. It is commonly used in cell culture experiments to promote the growth and differentiation of hematopoietic stem cells and progenitor cells . Additionally, IL-3 is used as a standard in enzyme-linked immunosorbent assays (ELISAs) and other immunoassays to quantify the presence of IL-3 in biological samples .

Storage and Stability

Recombinant rat IL-3 is typically supplied as a lyophilized powder and should be reconstituted in sterile, distilled water or phosphate-buffered saline (PBS) to regain full activity . It is recommended to store the reconstituted protein at -20°C or lower to maintain its stability and avoid repeated freeze-thaw cycles . The lyophilized form can be stored at 2°C to 8°C, preferably desiccated .

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.