IL 1 alpha Mouse, His

Interleukin-1 alpha Mouse Recombinant, His Tag
Shipped with Ice Packs
In Stock

Description

Production and Formulation

  • Expression Systems:

    • E. coli: Yields non-glycosylated, bioactive IL-1α (17.9 kDa) .

    • HEK293: Produces glycosylated IL-1α with near-native folding, critical for receptor-binding studies .

  • Reconstitution: Lyophilized protein is soluble in sterile water (≥100 µg/mL) and stable in PBS (pH 7.4) .

Biological Mechanism

IL-1 alpha Mouse, His binds the IL-1 receptor type 1 (IL-1R1), activating downstream signaling via MyD88 and NF-κB pathways . Key activities include:

  • Pro-Inflammatory Effects: Synergizes with TNF-α to induce fever, neutrophil recruitment, and acute-phase proteins .

  • Immune Cell Activation: Stimulates macrophages, fibroblasts, and endothelial cells to secrete cytokines (e.g., IL-6, CXCL1/KC) .

  • Wound Healing: Promotes keratinocyte proliferation and growth factor release (FGF, EGF) .

4.1. Inflammatory Disease Models

  • Skin Inflammation: IL-1α drives neutrophilic dermatoses in Ptpn6 mutant mice. Genetic ablation of IL-1α (but not IL-1β) prevents disease progression .

  • Autoinflammatory Disorders: CRISPR-generated Il1a-KO line2 mice exhibit normal IL-1β expression, clarifying IL-1α-specific roles in chronic inflammation .

4.2. Therapeutic Insights

  • COVID-19: Anakinra (IL-1α/β inhibitor) reduced clinical progression in trials, whereas IL-1β-specific blockers (e.g., canakinumab) showed no efficacy, underscoring IL-1α’s dominance in hyperinflammation .

4.3. Functional Assays

  • KC/CXCL1 Production: IL-1α-deficient macrophages show impaired neutrophil chemoattractant release, validating its role in innate immunity .

  • Radiation Protection: IL-1α mitigates γ-irradiation lethality in mice via hematopoietic recovery .

Comparative Insights: IL-1α vs. IL-1β

FeatureIL-1αIL-1β
Precursor ProcessingCalpain-mediated cleavage Caspase-1 (ICE)-dependent
Cellular SourceKeratinocytes, epithelial cellsActivated macrophages
BioactivityActive as precursor and mature formRequires processing for activity
Knockout PhenotypeProtects against skin inflammationLess impact on neutrophilic diseases

Challenges and Future Directions

  • Model Limitations: Earlier Il1a-KO line1 mice showed reduced IL-1β expression, confounding phenotype interpretation. The CRISPR-generated line2 model resolves this .

  • Therapeutic Targeting: IL-1α’s dual nuclear/cytosolic localization complicates drug design, necessitating cell-specific delivery strategies .

Product Specs

Introduction
Interleukin-1 alpha (IL-1 alpha) is a proinflammatory cytokine produced by various cell types, including macrophages, osteoblasts, monocytes, and hepatocytes. Typically, circulating levels of IL-1 alpha are low and only elevate upon stimulation by agents associated with inflammation, infection, or microbial endotoxins. IL-1 alpha exhibits a wide range of biological activities and exerts its effects by binding to specific cell surface receptors.
Description
Recombinant Mouse IL-1 alpha, produced in E. coli, is a single, non-glycosylated polypeptide chain comprising 179 amino acids (115-270 a.a). It has a molecular mass of 20.4 kDa. The IL-1 alpha is fused to a 23 amino acid His-tag at the N-terminus and purified using proprietary chromatographic techniques.
Physical Appearance
Sterile Filtered colorless solution.
Formulation
The IL-1 alpha protein solution (1 mg/ml) is supplied in Phosphate buffered saline (pH 7.4) with 10% glycerol.
Stability
For short-term storage (2-4 weeks), store at 4°C. For extended periods, store frozen at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Purity is determined to be greater than 85% by SDS-PAGE analysis.
Synonyms
Interleukin-1 alpha, IL-1 alpha, Il1a.
Source
E.coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSSAPYTYQ SDLRYKLMKL VRQKFVMNDS LNQTIYQDVD KHYLSTTWLN DLQQEVKFDM YAYSSGGDDS KYPVTLKISD SQLFVSAQGE DQPVLLKELP ETPKLITGSE TDLIFFWKSI NSKNYFTSAA YPELFIATKE QSRVHLARGL PSMTDFQIS.

Q&A

Overview of Key Research Findings

Recent advancements in IL-1α mouse models have clarified distinct roles of IL-1α independent of IL-1β, resolving long-standing contradictions in the field. The development of the CRISPR-Cas9 generated IL-1α knockout mouse line (IL-1α-KO line2) has addressed previous limitations in studying IL-1α function. This model demonstrates that IL-1α specifically regulates KC/CXCL1 expression, while confirming that IL-1β production is not dependent on IL-1α as previously thought .

What is IL-1α and how does it differ from other IL-1 family members?

IL-1α is a founding member of the IL-1 cytokine family and functions as a critical inflammatory mediator in both host defense and disease pathogenesis. Unlike IL-1β, which requires processing by inflammasomes for activation, IL-1α can act as both a nuclear transcription regulator and a cytokine when released. The distinct functions of IL-1α include specific regulation of neutrophil chemoattractant KC/CXCL1, which is not dependent on IL-1β . Methodologically, researchers can distinguish IL-1α activity by measuring KC/CXCL1 levels following stimulation with PAMPs or pathogens, as IL-1α knockout significantly reduces KC production while IL-1β knockout does not affect or even increases KC levels .

How are IL-1α knockout mouse models generated?

Two primary approaches have been used to generate IL-1α knockout mice:

  • Traditional gene targeting (IL-1α-KO line1): This earlier model showed interdependent expression of IL-1α and IL-1β, with substantial reduction in IL-1β production when IL-1α was deleted .

  • CRISPR-Cas9 technology (IL-1α-KO line2): This newer approach targeted exons 2-5 of the IL-1α gene by simultaneous injection of two guide RNAs with Cas9 mRNA into pronuclear-staged C57BL/6J zygotes . Verification of successful deletion is performed through:

    • Targeted deep sequencing

    • PCR amplification of genomic DNA

    • Western blot analysis confirming loss of IL-1α protein production

This improved model shows normal IL-1β induction in response to microbial PAMPs and pathogens, enabling more precise study of IL-1α-specific functions .

What is the role of His-tagged IL-1α in mouse research models?

While the search results don't specifically mention His-tagged IL-1α, His-tagging is a common approach for protein purification and detection in research. For IL-1α research, His-tagged recombinant proteins can be used to study binding interactions with the IL-1 receptor, evaluate antibody specificity, and investigate protein-protein interactions. When designing experiments with His-tagged IL-1α, researchers should verify that the tag doesn't interfere with the protein's biological activity through functional assays comparing tagged and untagged versions .

How can researchers distinguish IL-1α-specific effects from IL-1β effects in inflammatory responses?

Distinguishing IL-1α-specific effects requires careful experimental design using appropriate knockout models. The newer CRISPR-generated IL-1α-KO line2 offers significant advantages because:

  • It shows normal induction and activation of IL-1β in response to stimuli

  • It demonstrates normal inflammasome activation

  • It confirms IL-1α-specific regulation of KC/CXCL1

Methodologically, researchers should:

  • Use both IL-1α and IL-1β knockout models in parallel experiments

  • Measure multiple inflammatory markers (not just IL-1β levels)

  • Evaluate temporal dynamics of cytokine production (early vs. late timepoints)

  • Consider using neutralizing antibodies as complementary approaches

  • Include appropriate controls for inflammasome activation

A specific protocol involves stimulating bone marrow-derived macrophages with LPS or pathogens and measuring both IL-1β and KC/CXCL1 production by ELISA or western blot, where KC production will be significantly reduced only in IL-1α knockout cells .

How should researchers address contradictory findings when comparing different IL-1α knockout mouse lines?

The conflicting results observed between different IL-1α knockout lines highlight important considerations for experimental design and interpretation:

  • Verify the genetic background of the knockout line (extensive backcrossing to C57BL/6 is recommended)

  • Characterize the molecular defect in each knockout line (deletion size, potential off-target effects)

  • Assess temporal dynamics of cytokine production (the IL-1α-KO line1 showed pronounced reduction in IL-1β only at early timepoints)

  • Consider context-dependent effects (acute vs. chronic inflammation)

  • Validate findings using complementary approaches (neutralizing antibodies, conditional knockouts)

When publishing results, researchers should clearly specify which knockout line was used and discuss potential limitations of the model. For chronic disease studies, both IL-1α-KO lines may yield similar results due to the temporal dynamics of IL-1β production .

What experimental protocols are recommended for studying IL-1α-dependent neutrophil recruitment?

The specific role of IL-1α in regulating KC/CXCL1 expression provides a foundation for studying IL-1α-dependent neutrophil recruitment. Recommended protocols include:

  • In vitro approach:

    • Isolate bone marrow-derived macrophages from WT, IL-1α-KO, and IL-1β-KO mice

    • Stimulate with PAMPs (LPS, Pam3) or pathogens

    • Measure KC/CXCL1 levels in supernatants by ELISA

    • Perform western blot analysis to confirm IL-1α and IL-1β protein expression

  • In vivo approach:

    • Induce inflammatory response in WT, IL-1α-KO, and IL-1β-KO mice

    • Collect peritoneal lavage fluid or other relevant tissues

    • Analyze neutrophil infiltration by flow cytometry

    • Measure KC/CXCL1 levels in lavage fluid or tissue homogenates

This methodology allows researchers to specifically attribute neutrophil recruitment effects to IL-1α or IL-1β in various inflammatory contexts.

What controls should be included when using IL-1α knockout mouse models?

Proper experimental design with appropriate controls is essential when working with IL-1α knockout mice:

  • Genetic controls:

    • Wild-type littermates as primary controls

    • IL-1β knockout mice to distinguish IL-1α-specific effects

    • IL-1R knockout mice to confirm receptor dependency

  • Validation controls:

    • Western blot analysis confirming absence of IL-1α protein expression

    • PCR genotyping of experimental animals

    • Analysis of basal immune cell populations to confirm normal development

  • Experimental controls:

    • Include both early (1-3h) and late (12-24h) timepoints when measuring cytokine responses

    • Use multiple stimuli (TLR ligands, pathogens, sterile insults)

    • Include positive controls for inflammasome activation (e.g., ATP treatment after LPS priming)

How do chronic versus acute inflammatory models differ when using IL-1α knockout mice?

The distinction between acute and chronic inflammation is critical when interpreting results from IL-1α knockout models:

  • Acute inflammation:

    • IL-1α-KO line1 shows pronounced defects in early IL-1β production

    • IL-1α-KO line2 maintains normal IL-1β production

    • Both lines show defects in KC/CXCL1 production and neutrophil recruitment

  • Chronic inflammation:

    • Both knockout lines eventually achieve similar levels of IL-1β production

    • Differential phenotypes between IL-1α and IL-1β knockout mice become more apparent

    • Studies using IL-1α-KO line1 have successfully identified unique functions in chronic autoinflammatory diseases despite early IL-1β defects

This temporal dynamic explains why previous studies using IL-1α-KO line1 in chronic disease models were able to distinguish IL-1α-specific functions despite the model's limitations in acute inflammatory studies .

What are the most reliable methods for validating IL-1α knockout efficiency?

Thorough validation of IL-1α knockout models should include multiple complementary approaches:

  • Genomic verification:

    • Targeted deep sequencing of the modified locus

    • PCR amplification using primers flanking the deletion site

    • Sequencing of the deletion junction

  • Protein expression analysis:

    • Western blot analysis of cell lysates following LPS stimulation

    • Intracellular flow cytometry

    • ELISA measurement of secreted IL-1α

  • Functional validation:

    • Assessment of KC/CXCL1 production in response to PAMPs or pathogens

    • Normal induction of non-IL-1α-dependent cytokines (e.g., TNF)

    • Normal inflammasome activation (caspase-1 cleavage, IL-1β maturation)

How should researchers interpret discrepant results between IL-1α and IL-1β knockout models?

When faced with discrepant results between IL-1α and IL-1β knockout models, researchers should consider:

  • Temporal dynamics: IL-1α and IL-1β may have different kinetics of expression and action. The IL-1α-KO line1 showed reduction in IL-1β primarily at early timepoints .

  • Context-specific effects: The increased KC production observed in IL-1β knockout cells treated with LPS+ATP suggests competition between IL-1α and IL-1β for IL-1R binding, where increased availability of IL-1R for IL-1α binding may promote hyperexpression of select inflammatory factors in the absence of IL-1β .

  • Compensatory mechanisms: Long-term absence of one cytokine may lead to compensatory production of other inflammatory mediators.

  • Cell type-specific responses: Different cell types may have distinct dependencies on IL-1α versus IL-1β signaling.

What statistical approaches are recommended for analyzing IL-1α-dependent inflammatory responses?

When analyzing data from IL-1α knockout experiments, appropriate statistical approaches include:

  • For comparing cytokine levels between genotypes:

    • Two-way ANOVA with Tukey's post-hoc test for multiple comparisons across genotypes and treatments

    • Repeated measures analysis for time-course experiments

  • For neutrophil recruitment studies:

    • Non-parametric tests (Mann-Whitney) if data doesn't follow normal distribution

    • Consider power analysis to determine appropriate sample sizes based on expected effect size

  • For comprehensive evaluation:

    • Principal component analysis for multiparameter cytokine profiling

    • Hierarchical clustering to identify patterns in inflammatory responses

    • Correlation analysis between IL-1α expression and downstream mediators

Sample sizes should be determined based on anticipated biological variability, with a minimum of 3-5 biological replicates per condition for in vitro studies and 8-12 animals per group for in vivo experiments.

What are common challenges in detecting IL-1α protein expression in mouse models?

Researchers frequently encounter difficulties in detecting IL-1α protein expression:

  • Low basal expression: IL-1α is often expressed at low levels in resting cells and requires stimulation for robust detection. Use strong TLR stimuli like LPS (100 ng/ml) for at least 3-6 hours to induce detectable expression .

  • Detection methods: Western blotting requires optimization of lysis conditions as IL-1α can be cell-associated. Include positive controls (LPS-stimulated wild-type macrophages) and verify antibody specificity using IL-1α knockout cells .

  • Subcellular localization: IL-1α can be nuclear, cytoplasmic, or membrane-associated depending on cell type and activation state. Consider fractionation approaches when analyzing localization.

  • Processing variations: IL-1α can exist as both full-length and processed forms. Use antibodies that recognize both forms or specific domains depending on experimental questions .

How can researchers address potential confounding factors when using IL-1α knockout mice?

Several strategies help minimize confounding factors in IL-1α knockout studies:

  • Genetic background standardization:

    • Use mice extensively backcrossed to C57BL/6J background

    • Include wild-type littermates as controls

    • Consider using CRISPR on inbred strains to minimize genetic variation

  • Microbiome considerations:

    • Co-house experimental groups or use littermates

    • Consider microbiome analysis in intestinal inflammation studies

    • Use germ-free facilities for highly sensitive phenotypes

  • Age and sex matching:

    • Use age-matched controls (particularly important in aging studies)

    • Include both sexes or justify single-sex usage

    • Analyze sex as a biological variable when appropriate

  • Experimental blinding:

    • Blind genotype information during data collection and analysis

    • Use objective, quantitative readouts when possible

What specialized techniques can improve the study of IL-1α in complex inflammatory models?

Advanced techniques for studying IL-1α in complex models include:

  • Conditional knockout approaches:

    • Cell-type specific deletion using Cre-loxP systems

    • Inducible deletion using tamoxifen-responsive Cre

    • Tissue-specific promoters driving Cre expression

  • Reporter systems:

    • IL-1α-GFP fusion proteins to track expression and localization

    • IL-1α promoter-driven reporter genes to monitor transcriptional regulation

    • Dual reporters for simultaneous tracking of IL-1α and IL-1β

  • Single-cell analysis:

    • scRNA-seq to identify IL-1α-producing cell populations

    • CyTOF for high-dimensional protein profiling

    • Imaging mass cytometry for spatial context

  • In vivo imaging:

    • Intravital microscopy to visualize neutrophil recruitment

    • Bioluminescence imaging using luciferase reporters

    • PET imaging with radiolabeled antibodies

Product Science Overview

Structure and Production

The recombinant mouse IL-1α protein is typically produced in Escherichia coli (E. coli) as a single, non-glycosylated polypeptide chain. This chain contains 179 amino acids and has a molecular mass of approximately 20.4 kDa . The protein is fused to a 23 amino acid His-tag at the N-terminus, which facilitates its purification through chromatographic techniques .

Biological Activity

IL-1α exerts its effects by binding to specific cell surface receptors. It is known to stimulate thymocyte proliferation by inducing the release of IL-2, promote B-cell maturation and proliferation, and enhance fibroblast growth factor activity . Additionally, IL-1α is identified as an endogenous pyrogen, meaning it can induce fever by stimulating the release of prostaglandins .

Applications

Recombinant IL-1α is widely used in laboratory research to study its role in inflammation and immune responses. It is also utilized in various assays to investigate its effects on different cell types and to understand the signaling pathways it activates .

Storage and Stability

For optimal stability, the IL-1α protein solution should be stored at 4°C if it will be used within 2-4 weeks. For longer storage periods, it is recommended to freeze the solution at -20°C and add a carrier protein such as 0.1% human serum albumin (HSA) or bovine serum albumin (BSA) to prevent degradation . It is important to avoid multiple freeze-thaw cycles to maintain the protein’s integrity.

Purity and Formulation

The recombinant IL-1α protein is typically formulated in a phosphate-buffered saline (PBS) solution with a pH of 7.4 and 10% glycerol . The purity of the protein is greater than 85% as determined by SDS-PAGE .

Safety and Usage

IL-1α is intended for laboratory research use only and is not suitable for use as a drug, agricultural or pesticidal product, food additive, or household chemical . Researchers handling this protein should follow appropriate safety protocols to ensure safe and effective use.

In summary, recombinant mouse IL-1α with a His-tag is a valuable tool for studying the mechanisms of inflammation and immune responses. Its production in E. coli and subsequent purification allow for high-quality protein suitable for various research 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.