IL 1 alpha Rat

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

Description

Inflammatory Responses

  • Local Inflammation: IL-1α is produced at sites of peripheral inflammation, such as subcutaneous air pouches injected with lipopolysaccharide (LPS). Concentrations of IL-1α rise significantly within 1 hour post-LPS exposure, preceding IL-1 receptor antagonist (IL-1ra) induction, and correlate with fever development .

  • Systemic Effects: While IL-1β dominates systemic responses, IL-1α contributes to localized inflammation, with minimal detection in plasma .

Reproductive Physiology

  • Parturition: IL-1α and its receptors (IL-1R1, IL-1R2) are dynamically regulated in uterine tissues during late gestation. IL-1R1 mRNA and protein increase at term, while IL-1R2 decreases, enhancing uterine sensitivity to IL-1α signaling during labor .

Blood-Testis Barrier (BTB) Regulation

  • Sertoli Cell Function: Intratesticular IL-1α administration disrupts BTB integrity by altering the localization of tight junction proteins (OCLN, F11R, TJP1) and actin cytoskeleton organization in Sertoli cells, facilitating germ cell translocation .

Receptor Interactions

  • IL-1R1 Signaling: IL-1α binds IL-1R1, which complexes with IL-1RAcP to activate NF-κB and MAPK pathways, driving inflammation .

  • Decoy Receptor IL-1R2: Acts as a sink for IL-1α, modulating its activity. IL-1R2 abundance decreases during parturition, amplifying IL-1α effects .

Cytoskeletal Modulation

  • Actin Dynamics: IL-1α induces disorganization of filamentous actin at the BTB and apical ectoplasmic specializations, disrupting cell adhesion without altering protein expression levels .

Table 1: IL-1α in LPS-Induced Inflammation (Rat Subcutaneous Air Pouch Model)1

Time Post-LPS (h)IL-1α Concentration (pg/mL)Fever Correlation
1120 ± 15Rising phase
3135 ± 20Peak fever
8130 ± 18Resolution phase

Table 2: IL-1α Effects on Blood-Testis Barrier (Rat Model)3

ParameterObservation
Germ cell loss (15 days post-treatment)~50% of seminiferous tubules affected
BTB permeabilityIncreased inulin-FITC leakage
Actin filament organizationDisrupted at BTB and ectoplasmic sites

Table 3: Disease Associations of IL-1α in Rats8

ConditionRole of IL-1α
HyperalgesiaMediates pain sensitization
HypotensionInduces vasodilation and shock
Interstitial lung diseasePromotes fibrotic responses
UveitisDrives ocular inflammation

Therapeutic and Pathological Implications

  • Inflammatory Diseases: IL-1α’s early and localized production makes it a target for anti-inflammatory therapies. Blocking IL-1R1 or enhancing IL-1ra reduces fever and tissue damage .

  • Reproductive Health: Progesterone suppresses IL-1R1 expression, delaying labor, while RU486 (a progesterone antagonist) upregulates IL-1R1, accelerating delivery .

  • Neurological Damage: In ischemic brain injury, IL-1α contributes to neuronal death, but chronic deletion shows compensatory mechanisms that limit therapeutic targeting .

Comparative Insights

  • Species-Specific Regulation: Rats exhibit distinct IL-1α/IL-1ra dynamics compared to mice. For example, IL-1ra in mice is highly induced systemically, whereas rats rely more on local IL-1α/IL-1β balance .

  • Redundancy in IL-1 Isoforms: IL-1α and IL-1β show overlapping roles in inflammation, but IL-1α’s constitutive expression grants it unique homeostatic functions .

Product Specs

Introduction
Interleukin-1 alpha (IL-1 alpha) is a cytokine that plays a key role in inflammation. It is produced by various cells, including those involved in immune responses and tissue repair. When released, IL-1 alpha triggers a cascade of signaling events that contribute to inflammation, fever, and other physiological responses.
Description
This product consists of recombinant rat Interleukin-1A, a non-glycosylated polypeptide chain containing 155 amino acids. It has a molecular mass of 17.7 kDa and is produced in E. coli. Purification is achieved using proprietary chromatographic techniques.
Physical Appearance
Sterile, white, lyophilized (freeze-dried) powder.
Formulation
The lyophilized protein was prepared in a sterile solution of 50mM Tris-HCl, pH 8, at a concentration of 1 mg/ml.
Solubility
To reconstitute, add sterile 18 MΩ-cm H2O to a concentration of at least 100 µg/ml. This solution can be further diluted in other aqueous solutions.
Stability
Lyophilized Interleukin-1a remains stable at room temperature for up to 3 weeks. For extended storage, keep desiccated below -18°C. After reconstitution, store at 4°C for 2-7 days. For long-term storage, add a carrier protein like HSA or BSA (0.1%) and store below -18°C. Avoid repeated freeze-thaw cycles.
Purity
Purity exceeds 98.0%, as determined by RP-HPLC and SDS-PAGE analyses.
Biological Activity
The biological activity, measured as the effective dose for half-maximal stimulation (ED50) of murine D10S cells, is less than 0.005 ng/ml. This corresponds to a specific activity of 200,000,000 IU/mg.
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
The sequence of the first five N-terminal amino acids was determined and was found to be Ala-Pro-His-Ser-Phe.

Q&A

What is IL-1 alpha and how does it differ from IL-1 beta in rats?

IL-1 alpha is a non-secreted pro-inflammatory cytokine produced mainly by activated macrophages, neutrophils, epithelial cells, and endothelial cells in rats. Unlike IL-1 beta, IL-1 alpha remains biologically active in both its precursor and mature forms. Structurally, rat IL-1 alpha contains 156 amino acids with a molecular weight of approximately 17.8 kDa .

Both IL-1 alpha and IL-1 beta bind to the same receptor (IL-1RI) and share similar biological properties, but they are encoded by different genes and exhibit distinct expression patterns and cellular localization . IL-1 alpha typically remains associated with the cell membrane or is released upon cell death, whereas IL-1 beta requires processing by inflammasomes before secretion.

Cross-species homology analysis reveals important evolutionary relationships:

SpeciesHuman IL-1AMouse IL-1ARat IL-1A
Human IL-1A---55.5%58.7%
Mouse IL-1A55.5%---78.8%
Rat IL-1A58.7%78.8%---

This significant conservation across species (particularly the 78.8% similarity between rat and mouse) indicates shared evolutionary pressures, though researchers must consider the 41.3% difference between human and rat when translating findings .

What are the primary biological functions of IL-1 alpha in rat models?

IL-1 alpha serves as a critical mediator of host defense responses to inflammation and injury in rats. It possesses metabolic, physiological, and hematopoietic activities, playing a central role in regulating immune responses . Key functions include:

  • Mediating fever responses during inflammation

  • Regulating local and systemic inflammatory processes

  • Modulating neuronal activity and neurorepair processes through microglial expression

  • Contributing to tissue inflammation in metabolic disorders, particularly in pancreatic islets of diabetic rats

  • Influencing brain damage in ischemic, excitotoxic, and traumatic injury models

Unlike many cytokines, IL-1 alpha exhibits dual functionality - acting as both an intracellular regulator when retained within cells and as a traditional cytokine when released into the extracellular environment. This dual nature makes it particularly important in conditions where cell damage occurs, such as sterile inflammation or tissue injury .

How is IL-1 alpha signaling regulated in rat models?

IL-1 alpha signaling in rats involves a complex regulatory system with multiple control points:

  • Receptor Engagement: IL-1 alpha signals by binding to IL-1 receptor type I (IL-1RI), which then associates with an accessory protein (IL-1RAcP) to initiate signal transduction .

  • Competitive Inhibition: IL-1 receptor antagonist (IL-1ra) serves as a natural inhibitor by binding to IL-1RI without initiating signaling. IL-1ra acts as a competitive receptor antagonist that blocks all known actions of IL-1 .

  • Temporal Regulation: The appearance of IL-1ra is typically delayed (appearing around 2 hours after inflammatory stimulus) compared to the more rapid induction of IL-1 alpha, creating a time-dependent regulatory mechanism .

  • Concentration Differential: IL-1ra prevents IL-1 actions at molar ratios of 500:1 or greater, and circulating IL-1ra concentrations in disease states are much higher than those of IL-1 .

  • Decoy Receptor: A second IL-1 receptor (IL-1RII) binds IL-1 but fails to initiate signal transduction, potentially limiting IL-1 bioavailability .

The balance between IL-1 agonists (IL-1 alpha and IL-1 beta) and antagonists (IL-1ra) determines the ultimate inflammatory response in various physiological and pathological conditions in rat models .

How does microglial-specific IL-1 alpha influence neuronal activity and neurorepair in rat models?

Recent research has revealed that microglial-derived IL-1 alpha plays a distinct role in regulating neuronal function compared to IL-1 alpha from other cellular sources. Conditional knockout studies using microglial-specific deletion of IL-1 alpha (created by crossing IL-1αfl/fl mice with those expressing CX3CR1 promoter-driven Cre recombinase) have demonstrated significant effects on neuronal activity and repair mechanisms .

Microglial IL-1 alpha appears to function as a key mediator in neuron-microglia communication pathways. During injury or inflammation, activated microglia release IL-1 alpha, which then modulates neuronal excitability, synaptic transmission, and potentially influences recovery processes. This cell-specific role suggests targeted intervention strategies might provide therapeutic benefits while avoiding systemic side effects of global IL-1 inhibition .

In stroke models, the temporal expression pattern of microglial IL-1 alpha shows biphasic regulation - an initial increase during acute injury followed by a second wave during repair phases. This temporal pattern highlights the context-dependent role of IL-1 alpha, potentially serving different functions during different phases of neural injury and recovery .

For researchers investigating neuroinflammation and neurodegenerative disorders in rat models, manipulating microglial-specific IL-1 alpha expression offers a promising approach to dissect the cell-specific contributions to pathology and recovery.

What is the relationship between IL-1 alpha and IL-1 receptor antagonist (IL-1ra) in peripheral inflammation in rats?

The relationship between IL-1 alpha and IL-1ra in peripheral inflammation demonstrates sophisticated temporal and spatial regulation. In rat models of peripheral inflammation induced by lipopolysaccharide (LPS) injection into subcutaneous air pouches, researchers have observed distinct expression patterns:

  • Temporal Divergence: IL-1 alpha and IL-1 beta levels increase within 1 hour after LPS injection, while IL-1ra production is delayed until approximately 2 hours post-stimulation .

  • Differential Kinetics: Following the initial increase, IL-1 alpha concentrations remain relatively constant, while IL-1 beta and IL-1ra levels continue to increase in parallel with the development of fever .

  • Compartmentalization: IL-1ra, but not IL-1 alpha or IL-1 beta, is detected in significant quantities in the plasma of LPS-injected animals, suggesting differential distribution between local tissues and circulation .

  • Quantitative Imbalance: In disease states, the concentration of circulating IL-1ra is much higher than that of IL-1, requiring molar ratios of 500:1 or greater for effective antagonism .

These findings suggest that IL-1ra serves as a natural regulatory mechanism to limit IL-1-mediated inflammation once it has been initiated. The delayed appearance and prolonged production of IL-1ra relative to IL-1 alpha create a temporal window during which inflammation can develop before being actively contained .

How does IL-1 alpha contribute to tissue inflammation in diabetic rat models?

In type 2 diabetic GK rat models, research has revealed a critical inflammatory process within pancreatic islets involving IL-1 family cytokines. While much of the characterization has focused on IL-1 beta, these findings provide important insights into the IL-1 family's role in metabolic inflammation:

  • Increased Expression: GK rat islets display elevated expression of inflammatory markers including IL-1 beta, suggesting a local inflammatory process .

  • Inflammatory Cascade: IL-1 activity promotes the expression of other cytokines and chemokines (IL-6, TNFα, KC, MCP-1, and MIP-1α), creating an inflammatory amplification loop .

  • Immune Cell Recruitment: This inflammatory cascade leads to the recruitment of CD68+, MHC II+, and CD53+ immune cells into pancreatic islets .

  • Metabolic Consequences: The resulting inflammation impacts both β-cell functional mass and insulin sensitivity, contributing to hyperglycemia and altered proinsulin/insulin ratios .

Intervention studies using IL-1ra treatment in GK rats demonstrate significant therapeutic benefits:

  • Decreased hyperglycemia

  • Reduced proinsulin/insulin ratio

  • Improved insulin sensitivity

  • Reduced islet inflammation and immune cell infiltration

  • Diminished peripheral inflammation (particularly in the liver)

These findings suggest that targeting IL-1 signaling may provide therapeutic benefits in metabolic disorders through multiple mechanisms, including both direct effects on pancreatic islets and indirect effects on systemic inflammation and insulin sensitivity .

What are the optimal detection methods for measuring IL-1 alpha in rat samples?

Selecting the appropriate detection method for IL-1 alpha in rat samples depends on research objectives, sample types, and required sensitivity. Several approaches offer complementary advantages:

Enzyme-Linked Immunosorbent Assay (ELISA)

Sandwich ELISA represents the gold standard for protein-level quantification:

  • Sensitivity: Typically <0.5 pg/mL for optimized commercial kits

  • Detection Range: 4.7-300 pg/mL for standard assays

  • Sample Compatibility: Serum, plasma, cell culture supernatant, and tissue lysates

  • Advantages: High specificity, quantitative results, established protocols

  • Limitations: Requires relatively large sample volumes, limited multiplexing capability

Multiplex Bead-Based Immunoassays

For simultaneous analysis of multiple cytokines:

  • Advantages: Measures IL-1 alpha alongside other inflammatory mediators from the same sample

  • Applications: Particularly valuable for comprehensive inflammatory profiling or when sample volume is limited

  • Considerations: May have slightly lower sensitivity than dedicated ELISA

Quantitative PCR (qPCR)

For transcript-level analysis:

  • Applications: Detecting changes in IL-1 alpha gene expression

  • Advantages: Highly sensitive, requires minimal sample input

  • Limitations: Does not measure protein levels or biological activity

Immunohistochemistry/Immunofluorescence

For spatial localization studies:

  • Applications: Identifying cellular sources of IL-1 alpha within tissues

  • Advantages: Preserves tissue architecture, allows co-localization with cell markers

  • Considerations: Generally qualitative or semi-quantitative

Bioactivity Assays

For functional assessment:

  • Approach: Measure cellular responses to IL-1 alpha in samples (e.g., IL-6 production)

  • Advantages: Assesses biologically active IL-1 alpha

  • Considerations: May not distinguish between IL-1 alpha and IL-1 beta without additional blocking antibodies

For most applications, combining protein quantification (ELISA) with gene expression analysis (qPCR) and localization studies (immunohistochemistry) provides the most comprehensive characterization of IL-1 alpha in rat experimental models .

How can researchers effectively study IL-1 alpha signaling pathways in rat models?

Investigating IL-1 alpha signaling pathways in rat models requires a multi-faceted approach combining pharmacological, genetic, and molecular techniques:

Pharmacological Modulation

  • Receptor Antagonism: IL-1ra administration blocks all IL-1 signaling, allowing assessment of pathway dependency

  • Selective Inhibition: Targeted inhibitors of downstream signaling components (e.g., p38 MAPK, NF-κB) help dissect specific pathway branches

  • Dosage Considerations: Given the competitive nature of IL-1ra, proper dosing (considering the 500:1 molar ratio requirement) is essential for complete blockade

Genetic Approaches

  • Conditional Knockouts: Cell-specific deletion using Cre-loxP systems (e.g., microglial-specific IL-1 alpha deletion) reveals cell-type specific functions

  • CRISPR/Cas9 Modification: Targeted editing of IL-1 alpha or receptor genes provides precise genetic manipulation

  • RNA Interference: siRNA or shRNA approaches for transient knockdown in specific tissues

Molecular Signaling Analysis

  • Phosphoprotein Analysis: Western blotting or phospho-flow cytometry to detect activation of key signaling molecules (IRAK1/4, TRAF6, p38, JNK, NF-κB)

  • Transcriptional Profiling: RNA-seq or targeted gene expression panels to characterize downstream transcriptional responses

  • Protein-Protein Interaction: Co-immunoprecipitation studies to examine receptor complex formation

Cellular Models

  • Primary Cell Isolation: Studying IL-1 alpha responses in freshly isolated rat cells maintains physiological relevance

  • Ex Vivo Tissue Cultures: Maintaining tissue architecture while allowing experimental manipulation (e.g., pancreatic islets)

  • Cell-Type Specific Analysis: Flow cytometry or single-cell approaches to examine responses in heterogeneous populations

In Vivo Experimental Models

  • LPS-Induced Inflammation: Subcutaneous air pouch or systemic LPS administration

  • Disease-Specific Models: Diabetic models (e.g., GK rats) , stroke models (MCAO) , or other condition-specific models

  • Tissue-Specific Analysis: Examining pathway activation in relevant target tissues rather than systemic measures alone

Combined approaches yield the most comprehensive understanding. For example, researchers might use conditional knockout rats challenged with inflammatory stimuli, followed by comprehensive analysis of signaling cascades and functional outcomes in relevant tissues .

What are the best experimental models to study IL-1 alpha in neuroinflammation in rats?

Several well-established experimental models offer distinct advantages for investigating IL-1 alpha in neuroinflammatory conditions in rats:

Middle Cerebral Artery Occlusion (MCAO)

The gold standard for ischemic stroke research:

  • Implementation: Temporary or permanent occlusion of the middle cerebral artery

  • Relevance: Models acute neuroinflammation following ischemia-reperfusion injury

  • IL-1 Alpha Role: Significant contributions to post-stroke inflammation and tissue damage

  • Intervention Studies: IL-1ra administration reduces infarct volume by approximately 50%, demonstrating therapeutic potential

Direct CNS Cytokine Administration

For mechanistic studies of IL-1 alpha effects:

  • Implementation: Intracerebroventricular (ICV) injection of recombinant IL-1 alpha or IL-1ra

  • Applications: Isolating direct central effects from peripheral inflammation

  • Parameters: Dose-dependent effects can be studied with precise control

Microglial-Specific Genetic Manipulation

For cell-specific investigation:

  • Implementation: Conditional deletion using CX3CR1-CreERT2 systems

  • Advantages: Isolates microglial IL-1 alpha contribution from other cellular sources

  • Applications: Studying cell-autonomous versus non-cell-autonomous effects

LPS-Induced Neuroinflammation

For studying acute inflammatory responses:

  • Implementation: Systemic or central LPS administration

  • Features: Produces reliable neuroinflammatory response with increased IL-1 expression

  • Applications: Model for inflammation-induced cognitive impairment or sickness behavior

Traumatic Brain Injury Models

For post-traumatic neuroinflammation:

  • Implementation: Controlled cortical impact or fluid percussion injury

  • Relevance: Models acute and chronic neuroinflammation following mechanical injury

  • IL-1 Contribution: Significant role in secondary injury processes and potential recovery mechanisms

Chronic Neurodegeneration Models

For prolonged inflammatory processes:

  • Implementation: Neurotoxin-induced models (e.g., 6-OHDA for Parkinson's)

  • Applications: Studying IL-1 alpha in chronic neurodegenerative contexts

  • Advantages: Models slow progression of neuroinflammation over extended periods

When designing experiments, researchers should consider temporal dynamics (acute vs. chronic), region-specific effects (focal vs. global), and cell-specific contributions (microglia vs. astrocytes vs. neurons) to IL-1 alpha-mediated neuroinflammation .

How do researchers interpret conflicting data about IL-1 alpha function in different rat models?

When faced with seemingly contradictory findings regarding IL-1 alpha function across different rat models, researchers should systematically evaluate several key factors that might explain these disparities:

Model-Specific Context

Different disease models may reveal distinct facets of IL-1 alpha biology:

  • Sterile Inflammation: IL-1 alpha often acts as an early alarmin released from damaged cells

  • Infectious Challenges: May show different kinetics compared to aseptic inflammation

  • Acute vs. Chronic Models: IL-1 alpha may have distinct roles in different phases of disease

  • Tissue-Specific Responses: Functions may differ dramatically between CNS and metabolic contexts

Temporal Considerations

The timing of IL-1 alpha expression and intervention is critical:

  • Early Phase: Often pro-inflammatory and potentially harmful

  • Resolution Phase: May contribute to tissue repair and homeostasis restoration

  • Dynamic Balance: The relationship with IL-1ra changes over time, influencing net effects

Cell-Specific Origin

IL-1 alpha from different cellular sources may have divergent functions:

  • Microglial IL-1 alpha: Specific effects on neuronal activity and repair

  • Macrophage-Derived IL-1 alpha: Primarily pro-inflammatory in peripheral tissues

  • Epithelial IL-1 alpha: May serve distinct functions in barrier tissues

Methodological Variations

Technical factors can contribute to discrepant results:

  • Detection Methods: Different sensitivities or specificities of assays

  • Genetic Background: Strain differences among rat models (e.g., GK vs. Sprague-Dawley)

  • Intervention Approaches: Pharmacological vs. genetic targeting may yield different outcomes

Integration Strategies

To reconcile conflicting data, researchers should:

  • Design experiments with internal controls that directly compare conditions

  • Perform comprehensive temporal analyses rather than single time points

  • Use multiple complementary approaches to validate key findings

  • Consider combination of in vivo, ex vivo, and in vitro approaches to triangulate results

The IL-1 system's complexity requires nuanced interpretation. For example, the seemingly contradictory finding that IL-1 receptor knockout mice still develop ischemic brain damage comparable to wild-type might be explained by compensatory mechanisms, alternative receptors, or the balance with other cytokines in the inflammatory network.

What statistical approaches are most appropriate for analyzing IL-1 alpha expression data in rat studies?

Robust statistical analysis of IL-1 alpha data requires careful consideration of the unique characteristics of cytokine expression patterns. The following approaches are recommended:

Data Distribution Assessment

IL-1 alpha expression typically exhibits:

  • Non-normal distribution: Often positively skewed with potential outliers

  • Heteroscedasticity: Variance frequently increases with higher means

  • Zero-inflation: Many samples may have undetectable levels at baseline

Recommended Approaches:

  • Shapiro-Wilk test for normality assessment

  • Log or other transformation when appropriate

  • Non-parametric tests when normal distribution cannot be achieved

Experimental Design Considerations

Statistical power in IL-1 alpha studies requires:

  • Adequate sample sizes: Minimum n=8-10 per group for typical variability

  • Appropriate controls: Including vehicle controls, isotype antibody controls, genetic background controls

  • Repeated measures: When studying temporal dynamics

Analysis Strategies for Different Study Types

For Dose-Response Studies:

  • ANOVA with post-hoc tests (Tukey or Bonferroni)

  • Non-linear regression for EC50/IC50 determination

For Time-Course Studies:

  • Repeated measures ANOVA with sphericity correction

  • Area-under-curve (AUC) analysis for cumulative responses

  • Mixed-effects models for handling missing data points

For Correlation Studies:

  • Spearman's rank correlation for non-parametric data

  • Multiple regression for controlling confounding variables

  • Path analysis for exploring potential mechanistic relationships

Multiple Testing Corrections

Essential when examining:

  • Multiple cytokines simultaneously

  • Multiple time points

  • Multiple tissues or compartments

Recommended Approaches:

  • Bonferroni correction (conservative)

  • False Discovery Rate methods (Benjamini-Hochberg)

  • Family-wise error rate control

Visualization Strategies

Effective data presentation includes:

For complex designs involving IL-1 alpha measurements across multiple conditions, tissues, and time points, consultation with a biostatistician experienced in immunological research is highly recommended to ensure appropriate analysis strategies and interpretation.

How should researchers account for the balance between IL-1 alpha and IL-1 receptor antagonist in experimental design?

The dynamic balance between IL-1 alpha and IL-1 receptor antagonist (IL-1ra) critically influences experimental outcomes. Effective experimental design must account for this relationship through several key considerations:

Measurement Parameters

Essential Analytes:

  • IL-1 alpha (both precursor and mature forms)

  • IL-1 beta (for context and comparison)

  • IL-1ra (essential for interpreting net IL-1 activity)

  • IL-1RI and IL-1RII expression (receptor availability)

Ratio Calculations:

  • IL-1:IL-1ra molar ratios (functional antagonism requires ~500:1 IL-1ra:IL-1)

  • Active vs. total IL-1 alpha (precursor vs. processed forms)

Temporal Sampling Design

Given the delayed appearance of IL-1ra relative to IL-1 alpha , sampling strategies should include:

  • Early time points (1-2 hours) where IL-1 effects predominate

  • Intermediate points (3-5 hours) during the rising IL-1ra response

  • Later time points (8-24 hours) when IL-1ra levels typically peak

  • Resolution phase (24-72 hours) to capture return to homeostasis

Compartmental Analysis

Different biological compartments show distinct IL-1/IL-1ra dynamics:

  • Local tissue: Often contains both IL-1 alpha and IL-1ra

  • Circulation: May contain predominantly IL-1ra with minimal IL-1 alpha/beta

  • Cell-associated: IL-1 alpha can remain cell-membrane associated

Intervention Study Design

When manipulating the IL-1 system:

For IL-1ra Administration:

  • Dose determination based on expected physiological ratios

  • Timing relative to disease onset or experimental stimulus

  • Route of administration (systemic vs. local)

  • Pharmacokinetic considerations (half-life, distribution)

For IL-1 Alpha Manipulation:

  • Consider compensatory upregulation of IL-1ra

  • Monitor secondary inflammatory mediators

  • Account for potential redundancy with IL-1 beta

Experimental Controls

Robust experimental designs should include:

  • Dose-response curves for both IL-1 alpha and IL-1ra

  • Time course studies capturing dynamic relationships

  • Genetic approaches (knockouts/knockdowns) complementing pharmacological interventions

  • Comprehensive cytokine profiling beyond just IL-1 family members

As demonstrated in the GK rat diabetes model , targeting the IL-1 system through IL-1ra administration can have profound effects on both local tissue inflammation and systemic metabolic parameters, highlighting the importance of comprehensive assessment of the entire IL-1 network rather than isolated components.

What emerging technologies are advancing IL-1 alpha research in rat models?

Several cutting-edge technologies are transforming IL-1 alpha research in rat models, enabling more precise analysis of its biological roles:

CRISPR/Cas9 Genome Editing

Next-generation genetic manipulation allows:

  • Precise modification of IL-1 alpha, receptors, or pathway components

  • Generation of reporter rat lines with fluorescently tagged IL-1 system proteins

  • Creation of conditional knockouts with unprecedented specificity

  • Knock-in modifications that mimic human polymorphisms

Single-Cell Technologies

Resolving cellular heterogeneity through:

  • Single-cell RNA sequencing to identify cell-specific IL-1 alpha expression patterns

  • Single-cell proteomics for protein-level analysis

  • Spatial transcriptomics preserving tissue architecture context

  • CyTOF for simultaneous analysis of dozens of parameters in individual cells

Advanced Imaging Approaches

Visualizing IL-1 alpha dynamics through:

  • Intravital microscopy to track IL-1 alpha-expressing cells in vivo

  • PET imaging with radiolabeled IL-1ra to assess receptor occupancy

  • Bioluminescence resonance energy transfer (BRET) for real-time receptor activation studies

  • Light-sheet microscopy for whole-organ IL-1 system visualization

Organ-on-Chip and 3D Culture Systems

More physiologically relevant in vitro models:

  • Microfluidic devices mimicking tissue-specific environments

  • 3D organoids for studying IL-1 alpha in complex tissue architectures

  • Co-culture systems modeling cellular interactions

  • Perfusion systems replicating physiological cytokine gradients

Systems Biology Approaches

Holistic understanding through:

  • Multi-omics integration (genomics, transcriptomics, proteomics, metabolomics)

  • Computational modeling of IL-1 network dynamics

  • Machine learning for identifying complex patterns in IL-1 responses

  • Network analysis revealing IL-1 alpha's position in broader inflammatory cascades

These technologies are enabling researchers to address previously intractable questions about IL-1 alpha biology, including cell-specific functions in complex tissues, temporal dynamics at unprecedented resolution, and integration with broader inflammatory networks in health and disease .

How might research on IL-1 alpha in rat models translate to human therapeutic applications?

Research on IL-1 alpha in rat models offers several promising pathways to human therapeutic applications, though important translational considerations must be addressed:

Therapeutic Targeting Strategies

Rat models have validated multiple approaches that could translate to humans:

  • IL-1 Receptor Antagonism: IL-1ra administration in rats reduces inflammation and improves outcomes in multiple disease models

  • Cell-Specific Targeting: Microglial-specific IL-1 alpha manipulation suggests potential for cell-targeted approaches

  • Temporal Intervention: Rat studies revealing critical windows for IL-1 intervention inform optimal treatment timing

Disease Applications with Strong Translational Potential

Neurological Conditions:

  • Ischemic stroke: IL-1ra reduces infarct volume by approximately 50% in rat models

  • Traumatic brain injury: IL-1 pathway modulation shows neuroprotective effects

  • Neurodegenerative diseases: Microglial IL-1 alpha contributes to neuroinflammatory processes

Metabolic Disorders:

  • Type 2 diabetes: IL-1ra decreases hyperglycemia and improves insulin sensitivity in GK rats

  • Islet inflammation: Reduced pro-inflammatory cytokine production and immune cell infiltration

  • Hepatic inflammation: Improved inflammatory markers in the liver

Translational Challenges

Several factors require consideration when moving from rat models to human applications:

  • Species Differences: Rat IL-1 alpha shares only 58.7% homology with human IL-1 alpha

  • Disease Complexity: Human conditions often have greater heterogeneity than rat models

  • Dosing Translation: The 500:1 molar ratio required for IL-1ra efficacy necessitates careful dose scaling

  • Delivery Methods: Targeted delivery to affected tissues remains challenging

Biomarker Development

Rat studies inform potential biomarkers for patient stratification:

  • IL-1:IL-1ra ratios as predictors of inflammatory status

  • Tissue-specific versus systemic IL-1 measurements

  • Temporal profiles of IL-1 family members during disease progression

Combination Approaches

Findings from rat models suggest potential for combination therapies:

  • IL-1 blockade plus traditional disease-specific treatments

  • Targeting multiple inflammatory pathways simultaneously

  • Combining IL-1 antagonism with cellular or genetic therapies

The successful translation of IL-1ra from experimental models to approved human therapeutics (anakinra) demonstrates the feasibility of this pathway. Current rat research on cell-specific and context-dependent IL-1 alpha functions may lead to next-generation targeted approaches with improved efficacy and reduced side effects .

What are the key unresolved questions about IL-1 alpha function in rat models?

Despite significant advances, several fundamental questions about IL-1 alpha function in rat models remain unresolved and represent critical areas for future research:

Receptor Complexity and Signaling

Unresolved Questions:

  • Do alternative IL-1 receptors exist beyond the canonical IL-1RI? Evidence suggests IL-1β can exacerbate ischemic brain damage independently of IL-1RI

  • What explains cell-type specific responses to seemingly identical IL-1 alpha stimulation?

  • How do different forms of IL-1 alpha (precursor vs. mature) signal differently?

  • What determines the balance between beneficial and detrimental IL-1 alpha effects?

Cellular Sources and Targets

Unresolved Questions:

  • Beyond microglia, what other CNS cell types produce functionally significant IL-1 alpha?

  • How do different cellular sources of IL-1 alpha (immune vs. non-immune) contribute to specific pathologies?

  • Which target cells are most responsive to IL-1 alpha during different disease phases?

  • How does cell-specific deletion of IL-1 alpha in non-microglial cells affect disease outcomes?

Temporal Dynamics and Resolution

Unresolved Questions:

  • What regulates the transition from IL-1 alpha-dominated early responses to IL-1ra-dominated later phases?

  • Does IL-1 alpha play distinct roles in the resolution of inflammation versus the acute phase?

  • How do aging and chronic disease alter the temporal dynamics of IL-1 alpha responses?

  • What determines whether IL-1 alpha responses resolve appropriately or become pathologically persistent?

Tissue-Specific Functions

Unresolved Questions:

  • Why does IL-1 alpha appear to have different, sometimes opposing effects in different tissue contexts?

  • How do tissue-specific microenvironments modify IL-1 alpha signaling outcomes?

  • What explains the predominance of IL-1 effects in certain tissues despite ubiquitous receptor expression?

  • How do tissue-resident versus infiltrating cells differ in their IL-1 alpha production and response?

Therapeutic Implications

Unresolved Questions:

  • Can IL-1 alpha be selectively targeted without affecting IL-1 beta in therapeutic contexts?

  • What biomarkers would identify patients most likely to benefit from IL-1 alpha-specific interventions?

  • How can temporal and spatial specificity of IL-1 targeting be achieved therapeutically?

  • What combination strategies might overcome compensation or redundancy in the IL-1 system?

Addressing these questions will require innovative approaches combining conditional genetic manipulations, advanced imaging techniques, single-cell analyses, and systems biology perspectives to fully unravel the complex biology of IL-1 alpha in health and disease .

Product Science Overview

Structure and Production

Recombinant rat IL-1α is typically produced in Escherichia coli (E. coli) and is a non-glycosylated polypeptide chain containing 155 amino acids with a molecular mass of approximately 17,703 Daltons . The recombinant form is often used in research to study its biological activities and interactions.

Biological Function

IL-1α is primarily expressed by monocytes, macrophages, and dendritic cells. It is coded by the IL1A gene located on the q arm of chromosome 2 at position 13 . This cytokine plays a significant role in innate host defense by triggering the production of other proinflammatory cytokines in target cells and initiating acute-phase responses .

Mechanism of Action

IL-1α signals through two receptors, IL-1RI and IL-1RII, both of which are shared with IL-1β . Upon binding to its receptors, IL-1α activates various intracellular signaling pathways that lead to the expression of genes involved in inflammation, immune response, and cell proliferation.

Applications in Research

Recombinant rat IL-1α is widely used in research to understand its role in various physiological and pathological processes. It is utilized in cell proliferation assays, where its activity is measured using specific cell lines such as D10.G4.1 mouse helper T cells . The recombinant protein is also employed in enzyme-linked immunosorbent assays (ELISAs) to quantify its concentration in biological samples .

Storage and Stability

Recombinant rat IL-1α is typically lyophilized from a filtered solution in phosphate-buffered saline (PBS) and can be reconstituted in sterile PBS containing at least 0.1% human or bovine serum albumin . It is shipped at ambient temperature and should be stored at -20 to -70°C to maintain its stability. Once reconstituted, it can be stored under sterile conditions at 2 to 8°C for up to one month or at -20 to -70°C for up to three months .

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.