IL 1 alpha Human

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

Description

Introduction to IL-1 Alpha (Human)

Interleukin-1 alpha (IL-1α) is a pleiotropic cytokine encoded by the IL1A gene on human chromosome 2. It plays a central role in innate immunity, inflammation, and tissue repair by activating immune responses through the interleukin-1 receptor (IL-1R1) . Unlike IL-1β, IL-1α is bioactive in its precursor form and functions as an "alarmin," initiating inflammatory cascades during cellular stress or damage . It is produced by macrophages, neutrophils, endothelial cells, and epithelial cells, with roles spanning fever induction, hematopoiesis, and wound healing .

Gene and Protein Features

  • Gene: IL1A (chromosome 2q14.2) encodes a 271-amino acid precursor protein (31 kDa) .

  • Mature form: Processed into a 17 kDa active cytokine via proteolytic cleavage, though the precursor retains bioactivity .

  • Structure: Comprises 12 β-strands in a β-trefoil fold, conserved across mammals .

Unique Pro-Domain Properties

The IL-1α precursor contains a nuclear localization sequence (NLS) and histone acetyltransferase (HAT)-binding domains, enabling nuclear translocation to regulate gene expression independently of IL-1R signaling .

In Vitro Activities (Picomolar–Femtomolar Range)

  • Stimulates fibroblast proliferation and collagen synthesis .

  • Induces IL-6, prostaglandin E2 (PGE2), and cyclooxygenase-2 (COX-2) in epithelial cells .

  • Enhances neutrophil recruitment via keratinocyte-derived chemokines (e.g., CXCL1/KC) .

In Vivo Roles

  • Inflammation: Triggers acute-phase protein synthesis, fever, and muscle proteolysis .

  • Wound healing: Promotes fibroblast growth factor (FGF) and epidermal growth factor (EGF) expression in keratinocytes .

  • Radiation protection: Mitigates γ-irradiation lethality in mice via hematopoietic recovery .

Mechanism of Action

ComponentDetail
ReceptorBinds IL-1R1 with IL-1R3 as a co-receptor .
SignalingActivates NF-κB and MAPK pathways, amplifying TNF-α, IL-6, and chemokines .
RegulationAntagonized by IL-1 receptor antagonist (IL-1Ra), which competes for IL-1R1 binding .

Clinical Trials

  • Oncology: Phase 3 trials of MABp1 (anti-IL-1α antibody) show efficacy in advanced colorectal cancer .

  • Transplant medicine: IL-1α (50 ng/kg) accelerates platelet recovery post-autologous bone marrow transplantation .

  • Dermatology: Targeted inhibition reduces acne severity by suppressing follicular inflammation .

Preclinical Insights

  • Leukemia stem cells (LSCs): IL-1α/IL-1R1 axis drives proliferation in chronic myeloid leukemia (CML); combinatory therapy with tyrosine kinase inhibitors enhances efficacy .

  • Graft-versus-host disease (GVHD): IL-1α blockade ameliorates intestinal mucositis and GVHD in murine models .

Nuclear Functions

Pro-IL-1α interacts with histones via its conserved pro-domain, suggesting epigenetic regulatory roles in inflammation and senescence .

Species-Specific Roles

FeatureHumanMouse
Primary IL-1 in inflammationIL-1β dominates IL-1α drives systemic cytokines .
IL-1Ra regulationLimited basal expression High constitutive levels buffer IL-1α/β effects .

Functional Redundancy and Divergence

  • IL-1α is critical for CXCL1/KC production in macrophages, while IL-1β preferentially amplifies TNF-α .

  • In COVID-19, dual IL-1α/β blockade (anakinra) reduces mortality, whereas IL-1β-specific inhibitors (canakinumab) fail .

Challenges and Future Directions

Despite IL-1α's therapeutic potential, unanswered questions persist:

  • Mechanisms governing its nuclear-cytosolic shuttling .

  • Role of caspase-1-independent cleavage in chronic inflammation .

  • Biomarker development to stratify patients for IL-1α-targeted therapies .

Product Specs

Introduction

Interleukin-1 alpha (IL-1α) is a cytokine that plays a crucial role in the body's immune response. Primarily produced by activated macrophages, IL-1α stimulates the proliferation of thymocytes (immune cells in the thymus) by triggering the release of interleukin-2 (IL-2). Additionally, it contributes to the maturation and proliferation of B cells, which are responsible for producing antibodies. IL-1α also influences fibroblast growth factor activity, which is essential for tissue repair and wound healing. As a key player in the inflammatory response, IL-1α is recognized as an endogenous pyrogen, meaning it can induce fever. Furthermore, studies have shown that it can stimulate the production of prostaglandin, a hormone-like substance involved in inflammation and pain, from synovial cells found in joints.

Description
Recombinant human Interleukin-1 alpha, expressed in E. coli, is a non-glycosylated polypeptide chain consisting of 159 amino acids. This single-chain protein has a molecular weight of 18,022 Daltons. The purification of IL-1A is achieved using proprietary chromatographic methods.
Physical Appearance
The product appears as a sterile, white powder that has been lyophilized (freeze-dried).
Formulation
The protein was lyophilized from a sterile solution at a concentration of 1 mg/ml, containing 25 mM Tris-HCl buffer at a pH of 8.
Solubility
To reconstitute the lyophilized Interleukin-1 alpha, it is advised to dissolve it in sterile 18 MΩ-cm H2O to a concentration of at least 100 µg/ml. This solution can be further diluted in other aqueous solutions as needed.
Stability
Lyophilized Interleukin-1 alpha exhibits stability at room temperature for a period of 3 weeks. However, for long-term storage, it is recommended to store the desiccated product at temperatures below -18°C. After reconstitution, IL-1a should be stored at 4°C for a duration of 2 to 7 days. For extended storage, it is advisable to store it below -18°C. Repeated freeze-thaw cycles should be avoided.
Purity
The purity is determined to be greater than 97.0% through the following methods:
(a) Analysis using Reverse-Phase High-Performance Liquid Chromatography (RP-HPLC).
(b) Analysis using Sodium Dodecyl Sulfate-Polyacrylamide Gel Electrophoresis (SDS-PAGE).
Biological Activity
The ED50, which represents the effective concentration required to achieve 50% of the maximum response, was determined to be less than 0.001 ng/ml in a murine D10S cell stimulation assay. This corresponds to a specific activity of 1 x 109 IU/mg.
Protein Content
Protein quantification was conducted using two independent methods:
1. UV spectroscopy at a wavelength of 280 nm, utilizing an absorbency value of 1.13 as the extinction coefficient for a 0.1% (1 mg/ml) solution. This value was calculated using the PC GENE computer analysis program for protein sequences (IntelliGenetics).
2. Analysis by RP-HPLC, employing a standard solution of IL-1 as a reference standard.
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.
Amino Acid Sequence
SAPFSFLSNVKYNFMRIIKYEFILNDALNQSIIRANDQYLTAAALHNLDEAV KFDMGAYKSSKDDAKITVILRISKTQLYVTAQDEDQPVLLKEMPEIPKTITG SETNLLFFWETHGTKNYFTSVAHPNLFIATKQDYWVCLAGGPPSITDFQILE NQA.

Q&A

What is IL-1α and how is it synthesized in human cells?

IL-1α is a cytokine of the interleukin 1 family encoded by the IL1A gene in humans . Unlike most secreted proteins, IL-1α is initially synthesized as a 31kDa precursor lacking a signal peptide fragment (similar to IL-1β and IL-18) . The synthesis occurs in association with cytoskeletal structures, particularly microtubules, rather than on ribosomes associated with rough endoplasmic reticulum .

Methodologically, when studying IL-1α synthesis, researchers should note that both the 31kDa precursor and the 18kDa mature form (processed by calpain, a calcium-activated cysteine protease) are biologically active . This dual activity has significant implications for experimental design, as isolation protocols must account for both intracellular and extracellular forms. Western blotting with antibodies recognizing both forms is recommended for comprehensive detection.

What are the alternative names for IL-1α that appear in scientific literature?

When conducting literature searches or designing experiments involving IL-1α, researchers should be aware of its numerous alternative designations. IL-1α is also known as fibroblast-activating factor (FAF), lymphocyte-activating factor (LAF), B-cell-activating factor (BAF), leukocyte endogenous mediator (LEM), epidermal cell-derived thymocyte-activating factor (ETAF), serum amyloid A inducer or hepatocyte-stimulating factor (HSP), catabolin, hemopoetin-1 (H-1), endogenous pyrogen (EP), osteoclast-activating factor (OAF), and proteolysis-inducing factor (PIF) .

When searching databases, use comprehensive search terms including these alternatives to ensure complete literature coverage. Additionally, keyword variations should be incorporated into manuscripts to improve discoverability.

Which cell types are the primary sources of IL-1α in humans?

IL-1α is produced primarily by activated macrophages, as well as neutrophils, epithelial cells, and endothelial cells . In skin tissue, IL-1α is constitutively produced at high levels and contributes to maintaining skin barrier function against pathogens .

For experimental isolation of IL-1α-producing cells, researchers should consider tissue-specific sources. Flow cytometry with intracellular cytokine staining can identify specific cellular sources, while single-cell RNA sequencing provides comprehensive transcriptional profiling of IL1A expression across cell populations. When designing cell culture experiments, consider that keratinocytes represent a major reservoir of preformed IL-1α precursor .

How does IL-1α exert its biological effects at the cellular and molecular levels?

IL-1α exerts biological effects in the picomolar to femtomolar range through binding to the interleukin-1 receptor (IL-1R) . Experimentally, researchers should note several key activities when designing functional assays:

In vitro, IL-1α:

  • Stimulates autocrine secretion from keratinocytes and macrophages

  • Induces pro-collagen type I and III synthesis

  • Causes fibroblast proliferation and collagenase secretion

  • Induces cytoskeletal rearrangements

  • Triggers IL-6 and G-CSF secretion

  • Induces cyclooxygenase synthesis and prostaglandin PGE2 release

  • Causes phosphorylation of heat shock proteins

  • Stimulates smooth muscle cell and keratinocyte proliferation

  • Induces TNFα release from endothelial cells

  • Stimulates acute-phase protein secretion from hepatocytes

  • Promotes proliferation of CD4+ cells and IL-2 production

In functional studies, dose-response curves should be carefully established due to the high potency of IL-1α. Additionally, researchers should account for the synergistic effects between IL-1α and TNF, which has been demonstrated in numerous biological contexts including radioprotection, the Shwartzman reaction, PGE2 synthesis, and others .

What are the methodological approaches for investigating IL-1α in inflammatory responses?

When investigating IL-1α in inflammatory responses, researchers should employ multiple complementary approaches:

  • In vitro systems: Use primary human cells (macrophages, neutrophils, epithelial cells) stimulated with appropriate triggers (PAMPs, DAMPs, or pathogens) to study IL-1α production. Measure both cell-associated (precursor) and released (mature) forms.

  • Ex vivo analysis: Isolate cells from inflamed tissues to assess the IL-1α production profile across different cell types using flow cytometry or immunohistochemistry.

  • In vivo models: Consider that IL-1α interacts with other cytokines, particularly TNF, forming inflammatory feedback loops that can exacerbate neutrophil infiltration following bacterial infections like S. aureus or C. trachomatis . Design experiments that can distinguish between the direct effects of IL-1α and secondary effects mediated through induced cytokines.

  • Neutralization experiments: Use specific anti-IL-1α antibodies to block its activity while leaving IL-1β signaling intact, allowing discrimination between the two cytokines' effects.

Studies show that IL-1α functions as an alarmin that instigates host defense against multiple infectious agents, including M. tuberculosis, where IL-1α signaling cooperates with TNF signaling to promote protective granuloma formation .

How should researchers differentiate between the roles of IL-1α and IL-1β in experimental systems?

Differentiating between IL-1α and IL-1β functions remains challenging since both cytokines signal through the same IL-1R receptor. Methodologically, researchers should:

  • Use specific knockout models: The newly generated CRISPR IL-1α knockout mice (Il1a-KO line2) provide a more accurate model for differentiating IL-1α-specific functions compared to earlier knockout models (Il1a-KO line1) that showed reduced IL-1β expression . When selecting knockout models, investigate whether the model has confounding effects on IL-1β expression.

  • Apply selective neutralization: Use cytokine-specific neutralizing antibodies that target either IL-1α or IL-1β with minimal cross-reactivity.

  • Consider temporal dynamics: Research indicates that IL-1α and IL-1β may have different kinetics. The reduction of IL-1β expression in Il1a-KO line1 cells was pronounced only at early time points following stimulation, while prolonged stimulation resulted in similar levels of IL-1β in both wild-type and knockout cells .

  • Examine cell-specific effects: IL-1α functions as a nuclear transcription factor in addition to its extracellular cytokine role, whereas IL-1β functions exclusively as a secreted cytokine. Design experiments that can distinguish between these dual functions.

  • Control for inflammasome activation: IL-1β, but not IL-1α, requires inflammasome-mediated processing for activation, providing another experimental distinction point .

What are the optimal methods for detecting and quantifying IL-1α in human samples?

For reliable detection and quantification of IL-1α in human samples, researchers should consider:

  • ELISA/Immunoassays: Sandwich immunoassays like the Q-Plex assay provide sensitive detection with a range of 4,000 – 5.49 pg/mL and a lower limit of detection of 5.43 ng/mL . These assays require minimal sample volume (25μL) and provide results in approximately 2.25 hours.

  • Western Blotting: For distinguishing between precursor (31kDa) and mature (18kDa) forms of IL-1α, western blotting remains the gold standard. Include positive controls from stimulated macrophages or keratinocytes.

  • Immunohistochemistry/Immunofluorescence: For tissue localization studies, use antibodies validated for human tissues with appropriate blocking steps to minimize non-specific binding.

  • Gene Expression Analysis: qRT-PCR for IL1A mRNA provides information about transcriptional regulation, but should be complemented with protein analysis due to post-transcriptional regulation.

  • Bioactivity Assays: Functional bioassays using IL-1-responsive cell lines can assess the biological activity of IL-1α in samples.

For all detection methods, consider:

  • Sample preparation (serum vs. plasma vs. tissue lysates)

  • Timing of collection (IL-1α levels may fluctuate during inflammatory responses)

  • Storage conditions (-80°C recommended with minimal freeze-thaw cycles)

  • Appropriate normalization strategies for comparative analyses

What experimental controls should be included when studying IL-1α signaling pathways?

When investigating IL-1α signaling pathways, include these essential controls:

  • Positive Controls:

    • Recombinant human IL-1α at established concentrations

    • Samples from LPS-stimulated macrophages (known IL-1α producers)

    • Positive control cell lines with well-characterized IL-1R expression

  • Negative Controls:

    • IL-1R antagonist (IL-1Ra) to confirm receptor specificity

    • Anti-IL-1α neutralizing antibodies

    • Samples from IL-1α knockout models (preferably Il1a-KO line2)

    • IL-1R knockout cells or receptor blocking

  • Specificity Controls:

    • IL-1β stimulation to compare shared downstream pathways

    • TNF stimulation to identify synergistic or distinctive pathway components

  • Pathway Validation Controls:

    • Pharmacological inhibitors targeting specific nodes in the IL-1 signaling pathway

    • siRNA/shRNA knockdown of pathway components

    • Phosphorylation-specific antibodies to track activation states

Remember that mTOR activity significantly influences IL1A mRNA translation, and IL-1α and NF-κB mutually induce each other in a positive feedback loop . Experimental designs should account for these regulatory relationships.

How can researchers effectively study the complex interplay between IL-1α and TNF?

The synergistic relationship between IL-1α and TNF is one of the most clinically relevant cytokine interactions. To study this interplay effectively:

  • Dose-Response Matrices: Test combinations of IL-1α and TNF at various concentrations to identify synergistic vs. additive effects using isobologram analysis.

  • Sequential Stimulation: Apply IL-1α and TNF in different sequences with varying time intervals to determine temporal dependencies.

  • Selective Blocking: Use cytokine-specific neutralizing antibodies or soluble receptors to block either IL-1α or TNF individually while monitoring the other's activity.

  • Genetic Models: Utilize single knockout (IL-1α-KO or TNF-KO) and double knockout models to dissect individual and combined contributions.

  • Reporter Systems: Develop dual reporter systems that can simultaneously track IL-1α and TNF pathway activation.

  • Proteomics Approach: Apply phosphoproteomics to identify convergent and divergent signaling nodes between IL-1α and TNF pathways.

Research shows that IL-1α and TNF synergy has been demonstrated in numerous biological contexts, including radioprotection, the Shwartzman reaction, PGE2 synthesis, sickness behavior, nitric oxide production, nerve growth factor synthesis, insulin resistance, and chemokine synthesis . In infectious disease models, cooperation between IL-1α and TNF signaling promotes protective granuloma formation during M. tuberculosis infection and provides protection from gut-derived sepsis induced by P. aeruginosa .

What approaches should be used to investigate IL-1α in cancer biology?

IL-1α has complex roles in cancer biology, functioning both in tumor promotion and anti-tumor immunity. When investigating IL-1α in cancer contexts:

  • Expression Analysis: Characterize IL-1α expression in tumor cells, stromal cells, and infiltrating immune cells using immunohistochemistry, flow cytometry, and single-cell RNA sequencing.

  • Tumor Microenvironment Studies: Analyze how IL-1α affects the composition and function of the tumor microenvironment, particularly focusing on myeloid cell recruitment and activation.

  • Tumor Cell Autonomous Effects: Establish IL-1α-overexpressing and IL-1α-knockout tumor cell lines to study direct effects on proliferation, invasion, and metastasis.

  • Therapeutic Targeting: Evaluate anti-IL-1α therapeutic antibodies (like MABp1) for anti-neoplastic activity in solid tumors . Design studies that distinguish between effects on tumor cells versus effects on the tumor microenvironment.

  • IL-1α/TNF Interactions: Include experiments that assess potential synergy between IL-1α and TNF in the cancer setting, as this synergy is observed in multiple biological contexts .

  • Nuclear vs. Cytokine Functions: Design experiments that can differentiate between IL-1α's dual role as a nuclear factor and as a secreted cytokine.

Research indicates that IL-1α can kill a limited number of tumor cell types, suggesting context-dependent roles in cancer biology . Additionally, IL-1α is being evaluated in clinical trials as a potential therapeutic in oncology indications .

What methodological approaches should be used to investigate IL-1α in skin biology and wound healing?

IL-1α plays significant roles in skin biology and wound healing. When designing research in this area:

  • Skin Models: Use 3D organotypic skin models that contain both keratinocytes and fibroblasts to study IL-1α-mediated epithelial-mesenchymal interactions.

  • Wound Healing Assays: Implement in vitro scratch assays and in vivo excisional or incisional wound models, with or without IL-1α neutralization/supplementation.

  • Barrier Function Assessment: Measure transepidermal water loss and antimicrobial peptide production in relation to IL-1α activity.

  • Cellular Source Identification: Use cell type-specific knockouts or conditional deletion models to identify the relative contributions of IL-1α from different skin cell populations.

  • Growth Factor Interactions: Investigate the relationships between IL-1α and other growth factors like FGF and EGF that are induced by IL-1α in the wound healing context.

Research shows that IL-1α is constitutively produced at high levels in the skin and contributes to maintaining skin barrier function . Topically administered IL-1α stimulates expression of FGF and EGF, promoting fibroblast and keratinocyte proliferation . The presence of a large depot of IL-1α precursor in keratinocytes suggests that locally released IL-1α plays an important role in accelerating wound healing .

How should researchers approach the study of IL-1α knockout models given the conflicting observations in the literature?

When working with IL-1α knockout models, researchers should consider:

  • Model Verification: Thoroughly characterize your knockout model through genomic DNA PCR, targeted deep sequencing, and western blot analysis to confirm complete loss of IL-1α protein production .

  • Background Strain Consideration: Be aware that genetic background can influence phenotypes. The newer CRISPR-generated IL-1α knockout mice used pronuclear-staged C57BL/6J zygotes to minimize background-related genetic issues .

  • IL-1β Expression Assessment: Evaluate whether your knockout model affects IL-1β expression, as the original IL-1α knockout line (Il1a-KO line1) showed reduced IL-1β expression, while the newer line (Il1a-KO line2) showed normal induction and activation of IL-1β .

  • Temporal Analysis: Assess cytokine production at multiple time points, as research shows that IL-1β reduction in Il1a-KO line1 cells was pronounced only at early time points, with prolonged stimulation resulting in similar levels in both wild-type and knockout cells .

  • Stimulus Diversity: Test multiple stimuli, as responses may vary between pathogen-associated molecular patterns (PAMPs) and live pathogens .

  • Immune Cell Characterization: Evaluate immune cellularity in the blood and tissues, although the newer Il1a-KO line2 mice did not show gross abnormalities in immune cellularity .

Studies using different IL-1α knockout mice have produced conflicting observations, highlighting the importance of understanding the specific knockout model being used . The development of the Il1a-KO line2 mouse line, which does not display acute or chronic defects in IL-1β production, may help address many critical questions regarding the shared and unique functions of IL-1α and IL-1β cytokines .

What experimental approaches are recommended for evaluating IL-1α-targeting therapeutics?

When evaluating IL-1α-targeting therapeutics, researchers should implement:

  • Specificity Testing: Confirm that the therapeutic agent selectively targets IL-1α without affecting IL-1β or other IL-1 family members using binding assays and functional readouts.

  • Dose-Response Studies: Establish dose-response relationships in relevant in vitro and in vivo models, considering that IL-1α is active at picomolar to femtomolar concentrations .

  • Pharmacokinetic/Pharmacodynamic Analysis: Determine the half-life, tissue distribution, and duration of action of the therapeutic agent.

  • Biomarker Development: Identify and validate biomarkers that reflect IL-1α pathway inhibition to serve as pharmacodynamic markers in clinical studies.

  • Disease Models: Test in multiple disease models where IL-1α plays distinct roles, such as:

    • Skin conditions like acne (anti-IL-1α antibodies are being developed for this purpose)

    • Autoinflammatory disorders

    • Cancer (anti-IL-1α antibody MABp1 is being evaluated for anti-neoplastic activity)

    • Infectious disease models where IL-1α promotes protective responses

  • Combination Approaches: Evaluate combined blockade of IL-1α and TNF given their synergistic relationship in multiple biological contexts .

Clinical trials have explored IL-1α administration in patients receiving autologous bone marrow transplantation, with treatment at 50 ng/kg resulting in earlier recovery from thrombocytopenia compared to historical controls . This suggests potential applications beyond just antagonism of IL-1α activity.

How can researchers effectively study IL-1α in autoinflammatory diseases?

To study IL-1α in autoinflammatory diseases:

  • Patient Sample Analysis: Quantify IL-1α levels in serum, plasma, or affected tissues from patients with autoinflammatory diseases compared to healthy controls.

  • Genetic Association Studies: Investigate IL1A gene polymorphisms and their association with disease susceptibility or severity.

  • Ex Vivo Stimulation: Culture peripheral blood mononuclear cells from patients and controls with various stimuli to assess differences in IL-1α production and regulation.

  • Mouse Models: Utilize models of autoinflammatory diseases (e.g., Ptpn6 spin mice) to study the role of IL-1α, noting that cooperation between IL-1α and TNF mediates autoinflammation in these models .

  • Therapeutic Intervention Studies: Test IL-1α blockade in animal models and clinical studies, comparing outcomes with IL-1β-specific and IL-1R blockade.

  • Mechanistic Studies: Investigate the specific mechanisms by which IL-1α contributes to disease pathogenesis, such as neutrophil recruitment, inflammasome activation, or cytokine networks.

Research indicates that IL-1α can form inflammatory feedback loops that exacerbate neutrophil infiltration, potentially contributing to autoinflammatory pathogenesis . The interactions between IL-1α and TNF appear particularly important in the development of chronic autoinflammatory diseases .

What are the most pressing unresolved questions in IL-1α biology for researchers to address?

Several critical questions remain in IL-1α biology that warrant further investigation:

  • Nuclear vs. Extracellular Functions: How do the nuclear transcription factor functions of IL-1α integrate with its extracellular cytokine activities, and how can these distinct functions be experimentally isolated?

  • Cell-Type Specific Roles: What are the cell-type specific contributions of IL-1α in different disease contexts, and how does the cellular source influence biological outcomes?

  • Regulatory Mechanisms: What are the precise mechanisms that regulate IL-1α release from cells, particularly in non-cell death contexts?

  • Context-Dependent Effects: Why does IL-1α promote protective responses in some contexts (e.g., infections, wound healing) but pathological inflammation in others?

  • Translational Barriers: What has limited the clinical translation of IL-1α-targeting therapies compared to IL-1β-targeting approaches?

  • Biomarkers: What are reliable biomarkers of IL-1α activity that could guide clinical development of IL-1α-targeting therapies?

The development of new genetic tools like the Il1a-KO line2 mouse model offers opportunities to address many of these questions, potentially improving our understanding of the molecular basis of disease and informing therapeutic strategies .

What methodological advances would facilitate better research on IL-1α biology?

Advancing IL-1α research would benefit from these methodological innovations:

  • Selective Inhibitors: Development of small molecule inhibitors that can selectively target IL-1α without affecting IL-1β or requiring complete IL-1R blockade.

  • Conditional Knockout Models: Creation of cell type-specific and inducible IL-1α knockout models to dissect tissue-specific functions.

  • Biosensors: Development of real-time biosensors for IL-1α activity in living cells and tissues.

  • Single-Cell Technologies: Application of single-cell proteomics and transcriptomics to better characterize IL-1α-producing and responding cells.

  • Humanized Models: Engineering of humanized mouse models expressing human IL-1α and IL-1 receptors to improve translational relevance.

  • Structural Biology Approaches: Detailed characterization of structural differences between precursor and mature IL-1α and how they interact with receptors.

Product Science Overview

Structure and Production

IL-1α is initially produced as a precursor protein, known as pro-IL-1α, which is then proteolytically processed to generate the mature form of the cytokine. The precursor form of IL-1α can also be biologically active and is often found on the cell surface of monocytes and B lymphocytes . The mature IL-1α protein consists of 159 amino acids and has a molecular weight of approximately 18 kDa .

Biological Functions

IL-1α is a pleiotropic cytokine, meaning it has multiple effects on different cell types. It is involved in various immune responses, including the activation of T cells and the promotion of the proliferation of fibroblasts. IL-1α also induces the production of other proinflammatory cytokines and mediators, such as tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6), which contribute to the inflammatory response .

One of the key roles of IL-1α is in the regulation of sterile inflammation, which occurs in the absence of pathogens. During tissue injury or necrosis, IL-1α is released and recruits immune cells to the site of injury, promoting the healing process . Additionally, IL-1α has been shown to contribute to angiogenesis, the formation of new blood vessels, during hypoxic conditions .

Recombinant IL-1α

Recombinant IL-1α is produced using recombinant DNA technology, where the IL-1α gene is inserted into an expression system, such as bacteria or yeast, to produce the protein in large quantities. The recombinant protein is then purified to achieve high levels of purity, typically greater than 95% . Recombinant IL-1α is used in various research applications, including studies on immune responses, inflammation, and cell proliferation .

Applications in Research

Recombinant IL-1α is widely used in immunological research to study its effects on different cell types and its role in various biological processes. It is used in cell culture experiments to investigate the proliferation and activation of immune cells, as well as in animal models to study the effects of IL-1α in vivo . Additionally, recombinant IL-1α is used in assays to measure the production of other cytokines and inflammatory mediators .

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.