IL 1 alpha Human, His

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

Description

Production and Purification

IL-1α Human, His is synthesized in E. coli and purified via immobilized metal affinity chromatography (IMAC) leveraging the His tag . Key steps:

  • Expression: Optimized in E. coli for high yield, avoiding eukaryotic post-translational modifications .

  • Cleavage: The 31 kDa precursor is processed by proteases like calpain to generate the bioactive 17–18 kDa mature form .

  • Quality Control: Validated by SDS-PAGE, reverse-phase HPLC, and SEC-MALS for monodispersity .

In Vitro Bioassays

  • Cell Proliferation: IL-1α Human, His stimulates dose-dependent proliferation in murine D10.G4.1 T cells (EC₅₀: 0.05–0.15 ng/mL), neutralized by anti-IL-1α antibodies .

  • Cytokine Induction: Upregulates KC (CXCL1) in macrophages, a chemokine critical for neutrophil recruitment .

  • Receptor Binding: Binds IL-1R1 with high affinity, activating NF-κB and MAPK pathways .

In Vivo Relevance

  • Inflammation Models: IL-1α drives acute-phase responses (e.g., fever, leukocytosis) and chronic pathologies (e.g., autoinflammation) .

  • Wound Healing: Enhances fibroblast and keratinocyte proliferation via FGF/EGF upregulation .

Table 2: Key Functional Assays

Assay TypeReadoutEC₅₀/ND₅₀Citation
D10.G4.1 Proliferation[³H]-thymidine uptake0.05–0.15 ng/mL
KC Induction (LPS/ATP)CXCL1 secretionIL-1α-dependent

Applications in Research

  • Drug Development: Used to screen IL-1α inhibitors (e.g., IL-1Ra, neutralizing antibodies) .

  • Structural Studies: Facilitates crystallography and receptor interaction analyses .

  • Disease Modeling: Applied in sepsis, arthritis, and cancer studies to dissect IL-1α-specific pathways .

Product Specs

Introduction
Interleukin-1 alpha (IL-1 alpha) is a pro-inflammatory cytokine with a wide range of cellular origins, including macrophages, osteoblasts, monocytes, and hepatocytes. Typically found in low concentrations in circulation, IL-1 alpha levels elevate in response to stimuli such as inflammation, infection, or microbial endotoxins. Its biological activities are diverse, and it exerts its effects by binding to specific cell surface receptors.
Description
Recombinant human IL-1A, expressed in E. coli, is a non-glycosylated polypeptide chain containing 159 amino acids (fragment 113-271). It includes an amino-terminal hexahistidine tag and has a molecular mass of 22.5 kDa. The purification of the His-tagged IL-1 alpha is achieved using proprietary chromatographic techniques.
Physical Appearance
A clear, sterile-filtered solution.
Formulation
The His-tagged IL-1 alpha protein is supplied in a buffer of 1x PBS with 50% glycerol.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For long-term storage, it should be stored frozen at -20°C. Repeated freezing and thawing should be avoided.
Purity
The purity is determined to be greater than 95.0% using the following methods: (a) Reverse-phase high-performance liquid chromatography (RP-HPLC) analysis and (b) Sodium dodecyl-sulfate polyacrylamide gel electrophoresis (SDS-PAGE) analysis.
Synonyms
Hematopoietin-1, Lymphocyte-activating factor (LAF), Endogenous Pyrogen (EP), Leukocyte Endogenous Mediator (LEM), Mononuclear Cell Factor (MCF), IL-1 alpha,IL1, IL-1A, IL1F1.
Source
Escherichia Coli.

Q&A

What is the molecular structure of human IL-1α and how does the His-tag modification impact its biological activity?

Human IL-1α is a 31 kDa pro-protein belonging to the interleukin-1 family. Unlike many cytokines, IL-1α is biologically active in its unprocessed pro-form and contains a functional nuclear localization signal (NLS) in its N-terminal domain. This allows pro-IL-1α to shuttle to the nucleus upon translation in multiple cell types including epithelial cells, myeloid cells, and keratinocytes .

The addition of a histidine tag, typically comprising 6-10 histidine residues, facilitates protein purification via metal affinity chromatography but requires validation to ensure preservation of biological activity. Researchers should implement the following methodological approach:

  • Compare the biological activity of His-tagged versus untagged IL-1α in standard bioassays measuring IL-6 or TNF-α production

  • Evaluate receptor binding efficiency using surface plasmon resonance

  • Assess nuclear translocation capability through immunofluorescence microscopy

  • Consider removable His-tags using protease cleavage sites if structural interference is observed

How is IL-1α expression regulated at the transcriptional level and what experimental approaches best capture these regulatory mechanisms?

The IL-1α gene (IL1A) has a distinct promoter structure lacking canonical TATA and CAAT box regulatory regions. Instead, it contains binding sites for the transcription factor Sp1, which mediates basal expression during homeostasis . During inflammatory conditions, IL1A expression is upregulated through binding sites for AP1 and NF-κB transcription factors .

For effective experimental investigation of IL-1α transcriptional regulation:

  • Employ chromatin immunoprecipitation (ChIP) assays to identify transcription factor binding to the IL1A promoter under different conditions

  • Utilize reporter assays with wild-type and mutated promoter constructs to evaluate the contribution of specific transcription factor binding sites

  • Consider the role of the long noncoding antisense IL1A transcript (AS-IL-1α) which is required for IL1A transcription in myeloid cells

  • Analyze epigenetic modifications at the IL1A locus using bisulfite sequencing or ATAC-seq

What distinguishes IL-1α from IL-1β despite their shared receptor, and how can researchers experimentally differentiate their biological effects?

While both IL-1α and IL-1β signal through the same IL-1 receptor (IL-1R), they differ substantially in expression patterns, processing requirements, and cellular localization:

CharacteristicIL-1αIL-1β
Cell expressionConstitutively expressed in epithelial, endothelial, and stromal cellsExpression largely restricted to immune cells
BioactivityActive in both pro-form and mature formRequires inflammasome-mediated processing for activity
Cellular localizationContains NLS; can localize to nucleusPrimarily cytosolic
Release mechanismPrimarily passive release during cell deathActive secretion following inflammasome activation
Role as alarminPrimary alarmin during sterile injurySecondary response

To experimentally differentiate their effects:

  • Use genetic approaches with IL-1α or IL-1β knockout models, as demonstrated in Ptpn6 spin mice where IL-1α but not IL-1β deletion protects against neutrophilic dermatosis

  • Employ neutralizing antibodies specific to each cytokine

  • Design cell-type specific deletion strategies to address the different cellular sources

  • Analyze the kinetics of expression and release, as IL-1α often precedes IL-1β expression in many inflammatory contexts

How can researchers effectively study the nuclear functions of IL-1α independent of its receptor-mediated signaling?

IL-1α's capacity to localize to the nucleus and potentially regulate gene expression independently of receptor binding represents a unique feature requiring specialized experimental approaches:

  • Generate IL-1α constructs with mutations in the nuclear localization signal (residues 79-86) to create cytoplasm-restricted variants

  • Develop cell lines expressing IL-1α fused to a regulatable nuclear import system (e.g., hormone-responsive domains)

  • Perform nuclear-cytoplasmic fractionation followed by co-immunoprecipitation to identify nuclear binding partners

  • Utilize ChIP-seq approaches to map genomic binding sites of nuclear IL-1α, considering its reported interactions with histone acetyltransferases p300, PCAF, and GCN5

  • Implement RNA-seq analysis comparing cells expressing wild-type IL-1α versus NLS-mutant forms to identify nuclear-function-dependent gene regulation

The interaction of pro-IL-1α with HAX-1 (HS-1-associated protein X), a protein associated with mitochondrial, endoplasmic reticulum, and nuclear membranes, promotes its nuclear localization and may serve as an additional target for investigation .

What post-translational modifications regulate IL-1α function and what techniques should be employed to investigate their effects?

IL-1α undergoes several post-translational modifications that potentially regulate its function:

  • Myristoylation or acetylation on Lys82

  • Phosphorylation at Ser90

  • Proteolytic processing by calpain and other proteases

To investigate these modifications systematically:

  • Employ site-directed mutagenesis to generate modification-resistant variants (e.g., K82R to prevent acetylation, S90A to prevent phosphorylation)

  • Use mass spectrometry-based approaches (MS/MS) to identify and quantify modification sites under different conditions

  • Develop modification-specific antibodies for immunoblotting and immunoprecipitation

  • Apply pharmacological inhibitors of modifying enzymes (e.g., calpain inhibitors, kinase inhibitors) in cellular models

  • Assess the impact of modifications on subcellular localization through immunofluorescence microscopy and fractionation approaches

The functional consequences of these modifications remain largely unknown and represent an important frontier in IL-1α research .

How should researchers design experiments to investigate IL-1α's role as an alarmin during sterile inflammation?

IL-1α functions as a critical alarmin released during cell death to initiate inflammatory responses. To effectively study this function:

  • Establish necrotic cell administration models, as demonstrated by studies showing that infiltration of neutrophils into the peritoneal cavity upon administration of necrotic cells depends on IL-1α release

  • Implement tissue injury models (e.g., microabrasion of footpads) which have been shown to accelerate disease progression in IL-1α-dependent inflammatory conditions

  • Compare responses in wild-type versus IL-1α-deficient settings to delineate IL-1α-specific contributions

  • Use chimeric models to distinguish the role of IL-1α from hematopoietic versus non-hematopoietic sources

  • Analyze downstream inflammatory mediators including G-CSF, CXCL1, CXCL2, and TNF-α to map the inflammatory cascade initiated by IL-1α

Such approaches have revealed that IL-1α from non-hematopoietic cells can trigger robust inflammatory responses acting through myeloid cells in a SYK, MyD88, RIPK1, and TAK1-dependent manner .

What are the optimal models for studying IL-1α's role in inflammatory skin diseases and how should experiments be designed?

IL-1α plays a crucial role in various inflammatory skin conditions. For effective research:

  • Utilize the Ptpn6 spin mouse model of neutrophilic dermatosis, where genetic ablation of IL-1α, but not IL-1β, protects mice from disease development

  • Consider transgenic models that overexpress IL-1α in epidermal cells, which develop inflammatory skin disease highlighting IL-1α's pathogenic potential

  • Implement bone marrow chimeras to determine the cellular source of pathogenic IL-1α (hematopoietic vs. non-hematopoietic)

  • Use microabrasion injury models to study how mechanical damage accelerates disease progression through passive release of IL-1α from epidermal cells

  • Analyze downstream cytokine production (TNF-α, G-CSF, KC) to map the inflammatory cascade

Key experimental readouts should include:

  • Histological assessment of skin pathology

  • Flow cytometric analysis of inflammatory infiltrates

  • Cytokine profiling in tissue homogenates and serum

  • Gene expression analysis of inflammatory mediators

How can researchers effectively investigate the seemingly contradictory roles of IL-1α in cancer?

IL-1α exhibits dual roles in cancer, promoting tumor growth and metastasis in some contexts while enhancing anti-tumor immunity in others. To systematically investigate this dichotomy:

  • Compare models of endogenous IL-1α expression versus overexpression approaches:

    • Endogenous IL-1α often promotes tumor growth and angiogenesis

    • Overexpression of cytosolic or membrane-bound forms of IL-1α can lead to tumor regression

  • Evaluate different cellular compartments of IL-1α expression:

    • Membrane-bound IL-1α tends to promote anti-tumor immunity

    • Secreted IL-1α often supports tumor progression

  • Analyze immune cell populations to determine mechanisms:

    • IL-1α-mediated anti-tumor effects depend on CD8+ T cells, NK cells, and M1-differentiated macrophages

    • Pro-tumorigenic effects may involve myeloid-derived suppressor cells and tumor-associated macrophages

  • Study IL-1α in combination with cancer therapies:

    • IL-1α (and IL-1β) maturation by the NLRP3 inflammasome activated by chemotherapeutic drugs can contribute to anti-tumor CD8+ T cell responses

    • Simultaneously, chemotherapy-enhanced production of pro-inflammatory mediators may potentiate tumor invasiveness and metastasis

Clinical trials with IL-1α-blocking monoclonal antibody MABp1 have shown promising outcomes in patients with end-stage cancers, improving survival rate, increasing lean body weight, and decreasing systemic inflammation and cachexia .

What methodological approaches should be used to study IL-1α's role in host defense against microbial infections?

IL-1α plays non-redundant roles in defense against various pathogens. Effective research approaches include:

  • Bacterial infection models:

    • Mycobacterium tuberculosis infection, where IL-1α cooperates with TNF to promote protective granuloma formation

    • Pseudomonas aeruginosa colonization with cyclophosphamide-induced immunosuppression to study protection from gut-derived sepsis

    • Staphylococcus aureus or Chlamydia trachomatis infection to examine the inflammatory feedback loop involving IL-1α-induced chemokines

  • Viral infection models:

    • Adenovirus interaction with β3 integrin on marginal zone macrophages to study IL-1α upregulation and neutrophil recruitment

    • RSV infection models to examine IL-1α-driven neutrophil infiltration in lungs

  • Fungal and protozoan models:

    • Candida albicans infection to study IL-1α production from keratinocytes and subsequent G-CSF production from endothelial cells

    • Cryptococcus neoformans to examine IL-1α expression in the brain following systemic infection

    • Leishmania major infection in susceptible BALB/c mice where IL-1α drives pathology

Key experimental readouts should include:

  • Pathogen clearance kinetics

  • Inflammatory cell recruitment profiles

  • Cytokine and chemokine production measurements

  • Tissue damage assessment

  • Survival analysis

How should researchers design experiments to elucidate the complex signaling pathways downstream of IL-1α?

IL-1α signaling involves complex interactions with multiple pathways. To effectively study these networks:

  • Employ genetic approaches with knockout models for key signaling components:

    • Studies with Ptpn6 spin mice revealed that SHP-1 controls activation of spleen tyrosine kinase (SYK), which regulates MyD88 phosphorylation in myeloid cells

    • Genetic deletion of RIPK1, TAK1, or SYK in the appropriate cellular compartments can help dissect signaling requirements

  • Use phosphoproteomic approaches to map signaling cascades:

    • Temporal profiling of phosphorylation events following IL-1α stimulation

    • Comparison of signaling in different cell types (e.g., epithelial vs. immune cells)

  • Implement pharmacological inhibition approaches:

    • TAK1 inhibitors to assess transforming growth factor-β activated kinase 1 involvement

    • RIPK1 inhibitors to distinguish between its scaffolding versus kinase functions

  • Apply single-cell approaches to address cellular heterogeneity:

    • Single-cell RNA-seq to identify cell-specific responses

    • Single-cell phospho-flow cytometry to quantify signaling at the individual cell level

Studies have shown that IL-1α from non-hematopoietic cells acts on myeloid cells through a pathway involving SYK, MyD88, RIPK1, and TAK1, which is negatively regulated by SHP-1 to restrain excessive IL-1R signaling and associated auto-inflammation .

What are the key considerations for studying IL-1α protein-protein interactions and how can they be effectively investigated?

IL-1α engages in multiple protein-protein interactions that regulate its function and localization:

  • Receptor interactions: IL-1α binds to IL-1R1, which then recruits IL-1RAcP to form a signaling complex

  • Intracellular binding partners: Pro-IL-1α interacts with HAX-1, p300, PCAF, and GCN5

  • Regulatory interactions: Intracellular IL-1R2 can mask the NLS of pro-IL-1α, leading to cytosolic retention

To investigate these interactions systematically:

  • Apply proximity-based approaches:

    • BioID or TurboID to identify proteins in close proximity to IL-1α in different cellular compartments

    • FRET or split-luciferase assays to monitor interactions in living cells

  • Use co-immunoprecipitation approaches:

    • Standard co-IP followed by mass spectrometry to identify novel binding partners

    • Sequential co-IP to identify multiprotein complexes

  • Implement structural biology techniques:

    • X-ray crystallography of IL-1α with binding partners

    • Hydrogen-deuterium exchange mass spectrometry to map interaction interfaces

    • Cryo-EM for larger complexes

  • Develop domain mapping strategies:

    • Generate truncated variants to identify interaction domains

    • Site-directed mutagenesis of candidate interaction residues

Understanding these interactions is crucial for developing targeted therapeutics and elucidating mechanisms of IL-1α regulation.

What are the most effective strategies for distinguishing between membrane-associated and secreted forms of IL-1α in experimental systems?

IL-1α can exist in multiple forms including nuclear, cytosolic, membrane-associated, and secreted variants. To effectively distinguish between these forms:

  • Implement cell fractionation approaches:

    • Separate nuclear, cytosolic, membrane, and extracellular fractions

    • Use appropriate markers to validate fraction purity (histone H3 for nuclear, GAPDH for cytosolic, Na+/K+ ATPase for membrane)

  • Generate form-specific IL-1α variants:

    • Membrane-anchored IL-1α by fusion with transmembrane domains

    • Nuclear-restricted forms through strong NLS addition

    • Secretion-enhanced variants through signal peptide addition

  • Apply imaging approaches:

    • Live-cell imaging with fluorescently tagged IL-1α variants

    • Super-resolution microscopy to visualize subcellular localization

    • FRAP (Fluorescence Recovery After Photobleaching) to measure mobility

  • Develop detection strategies that distinguish the forms:

    • Form-specific ELISA approaches

    • Flow cytometry for cell-surface IL-1α detection

    • Proximity ligation assays to detect specific interactions

Understanding the distinct biological activities of these different forms is critical, as studies suggest membrane-bound IL-1α may promote anti-tumor immunity while secreted forms often contribute to pathological inflammation .

How can researchers effectively address the technical challenges in studying IL-1α's role in the tumor microenvironment?

The tumor microenvironment presents unique challenges for studying IL-1α functions:

  • Implement advanced in vivo imaging techniques:

    • Intravital microscopy with fluorescently labeled IL-1α or reporter systems

    • PET imaging with radiolabeled anti-IL-1α antibodies to track distribution

  • Develop complex 3D culture systems:

    • Tumor spheroids co-cultured with stromal and immune cells

    • Microfluidic tumor-on-a-chip platforms with controlled cytokine gradients

    • Organoid models derived from patient samples

  • Apply spatial transcriptomics and proteomics:

    • Geo-seq or Visium spatial transcriptomics to map IL-1α expression patterns

    • Imaging mass cytometry to visualize protein distribution in tissue sections

    • Digital spatial profiling for high-plex protein and RNA analysis

  • Consider heterogeneity through single-cell approaches:

    • Single-cell RNA-seq of tumor and stromal populations

    • Single-cell secretome analysis to identify cellular sources of IL-1α

  • Design appropriate therapeutic intervention studies:

    • Local versus systemic IL-1α blockade

    • Combination approaches with immune checkpoint inhibitors

    • Timing considerations for intervention (early versus late-stage disease)

Clinical trials with the IL-1α-blocking monoclonal antibody MABp1 demonstrate that targeting IL-1α can improve outcomes in patients with advanced cancers, suggesting translational potential for these research approaches .

Product Science Overview

Introduction

Interleukin-1 alpha (IL-1α) is a cytokine belonging to the interleukin 1 family, which plays a crucial role in the regulation of immune responses, inflammation, and hematopoiesis. It is produced by various cell types, including macrophages, monocytes, and epithelial cells . The recombinant form of IL-1α, tagged with a histidine (His) tag, is widely used in research and therapeutic applications due to its ease of purification and enhanced stability.

Structure and Production

IL-1α is a non-glycosylated polypeptide chain consisting of 159 amino acids, with a molecular mass of approximately 22.5 kDa . The His tag, typically composed of six histidine residues, is added to the N-terminus of the protein to facilitate purification using affinity chromatography techniques . The recombinant IL-1α is commonly produced in Escherichia coli (E. coli) expression systems, which allow for high-yield production and cost-effective manufacturing .

Biological Functions

IL-1α is a pleiotropic cytokine involved in various biological processes, including:

  • Immune Response: IL-1α plays a central role in the activation and regulation of immune cells, such as T cells and macrophages .
  • Inflammation: It is a key mediator of the inflammatory response, promoting the production of other pro-inflammatory cytokines and chemokines .
  • Apoptosis: IL-1α can induce apoptosis in certain cell types, contributing to the removal of damaged or infected cells .
  • Hematopoiesis: It is involved in the regulation of hematopoietic stem cells and the production of blood cells .
Applications

Recombinant IL-1α with a His tag is used in various research and therapeutic applications, including:

  • Immunology Research: Studying the role of IL-1α in immune responses and inflammatory diseases.
  • Drug Development: Screening potential therapeutic agents targeting IL-1α or its signaling pathways.
  • Cell Culture: Using IL-1α to stimulate immune cells in vitro for research purposes.
Storage and Stability

Recombinant IL-1α (His tag) should be stored at -20°C to maintain its stability and activity. It is recommended to avoid repeated freeze-thaw cycles to prevent protein degradation .

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.