Mouse IL-1β is a 17.3–17.5 kDa protein comprising 153 amino acid residues, derived from proteolytic cleavage of a 31 kDa inactive precursor (pro-IL-1β) by caspase-1 . Key features include:
Gene: Il1b (Entrez Gene ID: 16176), located on chromosome 2 in mice .
Post-translational processing: Requires inflammasome activation (e.g., NLRP3, AIM2) for caspase-1-mediated cleavage .
Secretion mechanism: Non-classical pathway involving gasdermin-D pores, independent of ER/Golgi .
IL-1β drives immune and inflammatory responses through:
Immune activation: Stimulates prostaglandin synthesis, neutrophil recruitment, and Th17 cell differentiation .
Pathogen defense: Essential for containment of Mycobacterium tuberculosis via prostaglandin E2 (PGE2)-mediated pathways .
Disease associations: Elevated in chronic inflammation (e.g., arthritis, colitis) and cancer models .
In Il1b⁻/⁻ mice:
Absent acute-phase response to localized inflammation (e.g., turpentine) .
Normal systemic response to lipopolysaccharide (LPS), highlighting IL-1α redundancy in endotoxic shock .
Turpentine-induced inflammation: Impaired acute-phase protein production .
Cancer studies: IL-1β neutralization reduces melanoma lung nodules in DJ-1 KO mice but enhances them in wild types, indicating context-dependent roles .
IDOL mice: Enable real-time imaging of IL-1β activity via luciferase reporters, showing organ-specific inflammation (liver, pancreas) during hepatitis or pancreatitis .
L2-IL-1β mice: Exhibit esophageal squamous inflammation progressing to metaplasia, modeling Barrett’s esophagus .
Model Type | Phenotype/Application | Reference |
---|---|---|
Il1b⁻/⁻ | Impaired local inflammation response | |
IDOL (luciferase) | In vivo IL-1β activity tracking | |
L2-IL-1β transgenic | Esophageal metaplasia and cancer |
Neutralizing antibodies: Anti-IL-1β (e.g., AF-401-NA) blocks D10.G4.1 T-cell proliferation (ND₅₀ ≤0.25 µg/mL) .
Clinical relevance:
When activated, macrophages produce Interleukin-1 beta (IL-1β), a cytokine that plays a crucial role in stimulating the immune system. IL-1β exerts its effects by promoting the proliferation of thymocytes, which are precursor cells of T lymphocytes, through the induction of interleukin-2 (IL-2) release. Furthermore, IL-1β contributes to the maturation and proliferation of B cells, enhancing the humoral immune response. In addition to its immune-modulating properties, IL-1β also stimulates fibroblast growth factor activity, contributing to tissue repair and wound healing. Notably, IL-1β is recognized as an endogenous pyrogen, indicating its involvement in the induction of fever, a hallmark of the inflammatory response. Moreover, studies have demonstrated that IL-1β can stimulate the production of prostaglandins from synovial cells, highlighting its potential role in inflammatory joint conditions.
IL-1 beta (interleukin-1 beta) is a potent proinflammatory cytokine that mediates a wide range of immune and inflammatory responses in mice. It is initially synthesized as a 31 kDa inactive pro-form that accumulates in the cytosol of producer cells such as monocytes, macrophages, and dendritic cells. This precursor requires proteolytic processing to generate the biologically active form. The activation of inflammasomes—multi-protein complexes that respond to pathogens, stress conditions, and other danger signals—triggers the processing of caspase-1, which in turn cleaves pro-IL-1 beta into its active 17 kDa form. The mature mouse IL-1 beta is a 17.5 kDa protein containing 153 amino acid residues. Unlike many secreted proteins, IL-1 beta lacks a signal sequence peptide for the classical ER/Golgi secretory pathway and is released through an alternative and not fully characterized mechanism, often alongside active caspase-1 .
Recombinant mouse IL-1 beta used in laboratory research is typically an E. coli-derived protein comprising amino acids Val118-Ser269 of the native sequence, with an additional N-terminal methionine residue. This corresponds to the mature, processed form of the cytokine rather than the pro-form. Commercial preparations have a molecular weight of approximately 17.3-17.5 kDa and show a single band at around 19 kDa when resolved by SDS-PAGE under reducing conditions. The recombinant protein maintains the functional characteristics of naturally occurring IL-1 beta, including its ability to bind to IL-1 receptors and elicit biological responses. For instance, recombinant mouse IL-1 beta stimulates cell proliferation of the D10.G4.1 mouse helper T cell line with an ED50 (effective dose for 50% maximal response) of 2-10 pg/mL, demonstrating its high potency .
Mouse IL-1 beta signals through two distinct receptors: IL-1RI (type I receptor) and IL-1RII (type II receptor), both of which are also shared with IL-1 alpha. The signaling process involves a sequential binding mechanism where IL-1 beta first binds directly to IL-1RI, followed by association with the IL-1R accessory protein (IL-1R3/IL-1R AcP) to form a high-affinity receptor complex that initiates signal transduction. In contrast, IL-1RII has high affinity for IL-1 beta but functions as a decoy receptor that binds the cytokine without initiating signaling, thereby serving as a negative regulator of IL-1 beta activity. Additionally, IL-1 receptor antagonist (IL-1Ra) acts as a competitive inhibitor by occupying IL-1 receptors without triggering signaling, providing another level of regulation. The IL-1 receptor is particularly highly expressed in endocrine beta cells compared to other mouse tissues, making these cells especially responsive to IL-1 beta effects. This receptor system allows for precise control of IL-1 beta activity in different physiological and pathological conditions .
For optimal maintenance of mouse IL-1 beta activity, proper storage and reconstitution protocols are critical. Commercial preparations typically come in two forms: lyophilized with a carrier protein (commonly BSA) or as carrier-free solutions. For lyophilized preparations (e.g., catalog #401-ML), reconstitution should be performed at a concentration of 100 μg/mL in sterile PBS containing at least 0.1% human or bovine serum albumin. After reconstitution, the protein should be stored at temperatures below -20°C, preferably in a manual defrost freezer to prevent degradation from temperature fluctuations. It is crucial to avoid repeated freeze-thaw cycles, as these can significantly reduce bioactivity. For carrier-free preparations (e.g., #401-ML/CF), which are supplied as filtered solutions in PBS, immediate storage at recommended temperatures upon receipt is essential. When working with recombinant IL-1 beta in experimental settings, researchers should prepare single-use aliquots to minimize exposure to freeze-thaw cycles and maintain consistent activity across experiments .
Accurate measurement of mouse IL-1 beta production requires consideration of both the pro-form and mature form of the cytokine. Several complementary approaches are recommended:
When interpreting results, researchers should consider that IL-1 beta can be secreted in both processed and unprocessed forms under certain conditions, necessitating methods that distinguish between them for comprehensive analysis.
The choice between carrier-free and BSA-containing formulations of recombinant mouse IL-1 beta depends on the specific experimental application:
Characteristic | BSA-Containing (e.g., 401-ML) | Carrier-Free (e.g., 401-ML/CF) |
---|---|---|
Formulation | Lyophilized from PBS with BSA | 0.2 μm filtered solution in PBS |
Stability | Enhanced protein stability | May have reduced stability |
Shelf life | Extended | Potentially shorter |
Storage concentration | Can be stored at more dilute concentrations | May require higher concentrations for stability |
Shipping conditions | Ambient temperature | Shipped with dry ice or equivalent |
Recommended applications | Cell/tissue culture, ELISA standards | Applications where BSA might interfere |
Potential interference | BSA may interfere with certain assays | Minimal interference with sensitive assays |
Reconstitution | Requires reconstitution in PBS with human/bovine serum albumin | Ready to use as supplied |
IL-1 beta plays a significant role in the age-associated decline of pancreatic beta cell function, which represents a key risk factor for type 2 diabetes development. Research using mouse models has revealed several mechanisms:
Altered IL-1 system balance: During aging, the expression of protective IL-1 receptor antagonist (IL-1Ra) decreases in pancreatic islets, while IL-1 beta gene expression increases specifically in the CD45+ islet immune cell fraction. This shift creates an imbalance favoring IL-1 beta-mediated inflammatory effects.
Impact on insulin secretion: One-year-old mice with whole-body knockout of IL-1 beta demonstrate higher insulin secretion both in vivo and in isolated islets compared to wild-type controls. This suggests that IL-1 beta negatively regulates insulin secretion during aging.
Effects on beta cell proliferation and mass: IL-1 beta knockout mice show enhanced expression of the proliferation marker Ki67 and increased size and number of islets, indicating that IL-1 beta suppresses beta cell proliferation and mass expansion in aging animals.
Cell-specific effects: Myeloid cell-specific IL-1 beta knockout preserves glucose-stimulated insulin secretion during aging, while it progressively declines in control mice. This demonstrates that immune cell-derived IL-1 beta is particularly important in age-related beta cell dysfunction.
Molecular targets: IL-1 beta treatment of isolated islets reduces expression of key genes including E2f1 (involved in cell cycle regulation), Ins2 (insulin gene), and Kir6.2 (potassium channel subunit critical for insulin secretion). These molecular changes likely contribute to the functional decline of beta cells.
These findings collectively demonstrate that IL-1 beta from myeloid cells contributes to age-associated decline in beta cell function by affecting both functional properties and proliferative capacity of beta cells .
IL-1 beta expression and its effects vary considerably across different tissues in mouse models, reflecting tissue-specific roles in homeostasis and disease:
Pancreatic islets: Islet-resident macrophages express remarkably high levels of IL-1 beta and NLRP3 inflammasome components compared to macrophages in other tissues, even in the absence of metabolic challenge. These macrophages are constitutively M1-polarized, suggesting chronic IL-1 activity within islets that may impact beta cell function and mass during normal aging and in metabolic disorders.
Endocrine beta cells: The IL-1 receptor (IL-1R1) is highly expressed in pancreatic beta cells relative to other mouse tissues, making these cells particularly sensitive to IL-1 beta effects. This receptor expression pattern explains why beta cells are prime targets for both physiological and pathological IL-1 beta signaling.
Colonic tissue: Similar to islet macrophages, colonic macrophages express elevated levels of IL-1 beta and inflammasome components, suggesting parallel inflammatory mechanisms in these tissues.
Myeloid cells: Myeloid-specific deletion of IL-1 beta demonstrates that these cells are a critical source of the cytokine affecting metabolic function. Mice with myeloid cell-specific IL-1 beta knockout maintain better glucose-stimulated insulin secretion during aging compared to control mice.
Central nervous system: Microglial activation leads to IL-1 beta production in the brain, which contributes to neurotoxicity through interactions between the NLRP3 inflammasome and mitophagy pathways. This mechanism is relevant for neuroinflammatory and neurodegenerative conditions.
This tissue-specific pattern of IL-1 beta expression and signaling has important implications for understanding its diverse roles in different physiological processes and disease states, as well as for designing targeted therapeutic approaches .
Several knockout mouse models have been developed to investigate IL-1 beta function in different physiological and pathological contexts:
Whole-body IL-1 beta knockout (IL-1β KO): These mice completely lack IL-1 beta protein in the circulation and in all tissues. They show no detectable IL-1 beta in cultured peritoneal macrophages. This model is useful for studying the global effects of IL-1 beta deficiency across all systems. Research with these mice has revealed improved glucose tolerance and enhanced insulin secretion in aging animals, suggesting a role for IL-1 beta in age-related metabolic decline.
Myeloid cell-specific IL-1 beta knockout (MyeloIL-1β KO): Generated using Cre-loxP technology with myeloid-specific promoters (typically LysM-Cre), these mice lack IL-1 beta specifically in myeloid cells while maintaining normal expression in other cell types. Studies with these mice have demonstrated that myeloid-derived IL-1 beta is particularly important for age-associated decline in beta cell function. Aged myeloIL-1β KO mice secrete more insulin and maintain better expression of genes essential for beta cell function (Ins2, Kir6.2) and cell cycle regulation (E2f1).
IL-1 receptor antagonist knockout (IL-1Ra KO): While not directly targeting IL-1 beta itself, these mice lack the endogenous antagonist that regulates IL-1 signaling. Beta cell-specific deletion of IL-1Ra decreases insulin secretion by affecting E2f1 and Kir6.2 expression and prevents beta cell mass expansion, providing insight into how the balance between IL-1 beta and its antagonist regulates metabolic function.
IL-1 receptor type I knockout (IL-1RI KO): These mice lack the primary signaling receptor for both IL-1 alpha and IL-1 beta, effectively blocking all IL-1 signaling. They provide a model for studying the collective importance of IL-1 family cytokines in various biological processes.
These models can be used individually or in combination to dissect cell-specific contributions of IL-1 beta to different physiological and pathological processes, enabling researchers to delineate the complex roles of this cytokine in inflammation, metabolism, and aging .
When investigating IL-1 beta effects on pancreatic islets, several methodological considerations are critical for obtaining reliable and physiologically relevant results:
Islet isolation and culture:
Islets should be isolated using collagenase digestion followed by density gradient separation.
Allow 24-48 hours of recovery after isolation before IL-1 beta treatment to minimize effects of isolation stress.
Culture in low glucose (5.5-8.3 mM) RPMI 1640 or CMRL medium supplemented with 10% FBS, L-glutamine, and antibiotics.
Maintain at 37°C with 5% CO2 in non-adherent culture dishes to preserve islet architecture.
IL-1 beta treatment parameters:
Concentration: Effects are dose-dependent; low concentrations (0.01-0.1 ng/mL) may have different effects than higher concentrations (1-10 ng/mL).
Duration: Acute (1-24 hours) versus chronic (2-7 days) exposure produces distinct outcomes.
For studying age-related effects, compare responses in islets isolated from young (8-16 weeks) and aged (>52 weeks) mice.
Functional assessments:
Glucose-stimulated insulin secretion (GSIS): Measure insulin release at both basal (2.8 mM) and stimulatory (16.7 mM) glucose concentrations.
Use static incubation or perifusion systems for different temporal resolution of secretory responses.
Quantify both first-phase (0-10 minutes) and second-phase (10-60 minutes) insulin secretion when using perifusion.
Molecular analyses:
Monitor expression of key beta cell function genes: Ins1, Ins2, Kir6.2, Pdx1, MafA, Glut2.
Assess proliferation markers: Ki67, PCNA, BrdU incorporation.
Examine cell cycle regulators: E2f1, Cyclin D1, Cyclin D2.
Evaluate IL-1 system components: Il1b, Il1r1, Il1rn (IL-1Ra).
Control conditions:
Include vehicle-treated controls, IL-1Ra treatment groups, and IL-1 beta plus IL-1Ra co-treatment groups.
Consider comparing effects with other cytokines (TNF-α, IFN-γ) individually and in combination.
Include positive controls for islet dysfunction and death (e.g., high glucose, palmitate).
These methodological approaches allow for comprehensive assessment of how IL-1 beta affects beta cell function, proliferation, and survival in both physiological and pathological contexts, particularly in aging-related studies .
Distinguishing between direct and indirect effects of IL-1 beta in complex tissues like pancreatic islets or brain tissue requires sophisticated experimental approaches:
Cell-specific receptor knockout models:
Generate conditional knockouts of IL-1R1 in specific cell types using Cre-loxP technology.
Compare phenotypes when the receptor is deleted in one cell type versus another (e.g., beta cells versus macrophages in islets).
This approach allows determination of which cell types must directly sense IL-1 beta to produce observed effects.
Cell sorting and ex vivo analysis:
Dissociate complex tissues into single-cell suspensions.
Use fluorescence-activated cell sorting (FACS) with cell-specific markers to isolate pure populations.
Analyze sorted populations separately for IL-1 beta responses.
Compare responses in co-culture systems versus isolated cultures.
Single-cell transcriptomics and proteomics:
Apply single-cell RNA sequencing to tissues from IL-1 beta-treated versus control animals.
Identify cell type-specific transcriptional responses using bioinformatic approaches.
This can reveal which cell populations respond directly to IL-1 beta and which show secondary responses.
In situ approaches:
Use reporter mice that express fluorescent proteins under control of IL-1 responsive elements.
Perform immunohistochemistry for phosphorylated signaling molecules downstream of IL-1R1.
Apply RNA in situ hybridization techniques (e.g., RNAscope) to visualize IL-1 responsive genes in tissue context.
Temporal analysis:
Conduct time-course experiments to distinguish primary (rapid) versus secondary (delayed) responses.
Primary responses typically occur within minutes to hours after IL-1 beta exposure.
Secondary responses that require intermediate mediators develop over longer timeframes.
Pharmacological approaches:
Use cell type-specific drug delivery systems.
Apply inhibitors of secondary mediators to block indirect effects.
Combine IL-1 beta treatment with receptor antagonists that have different cell type accessibility.
These strategies, especially when used in combination, can effectively dissect the complex network of direct and indirect effects that IL-1 beta exerts in heterogeneous tissue environments, providing insight into the mechanisms by which this cytokine contributes to physiological and pathological processes .
When working with IL-1 beta in mouse models and experimental systems, researchers often encounter several technical challenges that can affect data reliability and interpretation:
Pro-form versus mature form discrimination:
Many antibodies and ELISA kits do not distinguish between the 31 kDa pro-IL-1 beta and the 17 kDa mature form.
Solution: Use Western blotting with appropriate antibodies to verify which form is being detected, or employ ELISAs specifically designed to detect only the mature form.
When reporting results, clearly specify which form(s) were measured.
Detection thresholds:
IL-1 beta often functions at picomolar concentrations that may fall below detection limits of standard assays.
Solution: Use high-sensitivity assays (detection limit <1 pg/mL) or bioassays that can detect functional effects at physiological concentrations (ED50 for mouse IL-1 beta in D10.G4.1 cell proliferation is 2-10 pg/mL).
Sample processing artifacts:
Mechanical disruption of cells during tissue processing can lead to artifactual activation of inflammasomes and processing of pro-IL-1 beta.
Solution: Use gentle isolation techniques, process samples rapidly at 4°C, and include appropriate controls to assess processing-induced activation.
Binding to soluble receptors and carrier proteins:
IL-1 beta can bind to soluble forms of IL-1 receptors or other plasma proteins, masking detection.
Solution: Include acid dissociation steps or use immunoprecipitation techniques to release bound cytokine before measurement.
Short half-life in vivo:
IL-1 beta has a short circulatory half-life, making timing of measurements critical.
Solution: Perform kinetic studies to determine optimal sampling times, or measure IL-1 beta-induced secondary mediators with longer half-lives.
Freeze-thaw instability:
Recombinant IL-1 beta and native IL-1 beta in biological samples can lose activity with repeated freeze-thaw cycles.
Solution: Prepare single-use aliquots of standards and samples, and avoid repeated freezing and thawing.
Carrier protein interference:
BSA in carrier-containing formulations can interfere with certain assays.
Solution: Use carrier-free formulations for applications where BSA might be problematic, such as mass spectrometry or certain binding studies.
Understanding these technical challenges and implementing appropriate controls and methodological adjustments is essential for obtaining reliable and reproducible data when studying IL-1 beta in research settings .
Optimizing IL-1 beta bioactivity in experimental systems requires careful attention to several factors that can influence cytokine potency and stability:
Proper storage and handling:
Store lyophilized recombinant IL-1 beta at -20°C to -80°C.
After reconstitution, prepare single-use aliquots to avoid repeated freeze-thaw cycles.
Use low-binding microcentrifuge tubes to minimize protein adsorption to vessel walls.
Include carrier protein (0.1-1% BSA) in storage buffer to enhance stability unless experimental conditions preclude this.
Reconstitution protocol:
Reconstitute lyophilized IL-1 beta at a concentration of 100 μg/mL in sterile PBS containing at least 0.1% human or bovine serum albumin.
Allow complete dissolution by gentle swirling rather than vigorous vortexing, which can cause protein denaturation.
Filter-sterilize using low protein-binding 0.2 μm filters if sterility is required.
Dose optimization:
Perform dose-response experiments for each specific application and cell type.
The ED50 for mouse IL-1 beta in D10.G4.1 cell proliferation assays is 2-10 pg/mL, but optimal concentrations may differ for other cell types and readouts.
Consider both concentration and exposure duration as variables.
Synergistic effects:
IL-1 beta often acts synergistically with other cytokines like TNF-α or IFN-γ.
Testing combinations at sub-optimal concentrations may reveal biologically relevant interactions.
Receptor expression:
Verify IL-1 receptor expression on target cells, as receptor density can significantly impact sensitivity.
Consider pre-treating cells with cytokines known to upregulate IL-1 receptors if responsiveness is low.
Controlling antagonistic factors:
Be aware that serum contains soluble IL-1 receptors and IL-1Ra that can neutralize IL-1 beta.
Consider using serum-free conditions or heat-inactivated serum to minimize this effect.
Test the impact of adding IL-1Ra-neutralizing antibodies if endogenous antagonism is suspected.
Biological validation:
Confirm bioactivity using a well-established assay (e.g., D10.G4.1 cell proliferation).
Include positive controls (e.g., IL-1 beta at known effective concentration) and negative controls (e.g., heat-inactivated IL-1 beta) in all experiments.
Validate bioactivity through measurement of canonical IL-1-induced responses such as NF-κB activation or IL-6 production.
By systematically addressing these factors, researchers can maximize IL-1 beta bioactivity in their experimental systems and ensure more consistent and reproducible results across studies .
Several cutting-edge research directions are expanding our understanding of IL-1 beta's roles in mouse models of disease:
Tissue-specific inflammasome activation:
Investigation of why certain tissue-resident macrophages (like those in islets and colon) constitutively express higher levels of IL-1 beta and NLRP3 inflammasome components compared to other tissue macrophages.
Exploration of tissue-specific triggers and regulation of inflammasome activation and IL-1 beta processing.
Development of tissue-targeted inflammasome inhibitors as potential therapeutic approaches.
Aging and senescence:
Characterization of the senescence-associated secretory phenotype (SASP) in different tissues, with IL-1 beta as a key SASP component.
Investigation of how age-associated changes in the balance between IL-1 beta and IL-1Ra contribute to tissue dysfunction.
Exploration of senolytic approaches targeting IL-1 beta-producing senescent cells to ameliorate age-related pathologies.
Metabolic regulation:
Further dissection of how IL-1 beta affects insulin secretion and beta cell function through novel molecular targets beyond E2f1 and Kir6.2.
Investigation of IL-1 beta's roles in adipose tissue inflammation and thermogenesis.
Exploration of IL-1 beta's contributions to exercise-induced metabolic adaptations.
Neuroinflammation:
Examination of microglial IL-1 beta production in models of neurodegenerative diseases.
Investigation of IL-1 beta's effects on synaptic plasticity and neurogenesis.
Development of targeted approaches to modulate IL-1 signaling in the CNS without affecting peripheral immunity.
Multi-omics approaches:
Integration of single-cell transcriptomics, proteomics, and metabolomics to map IL-1 beta-responsive networks in complex tissues.
Application of spatial transcriptomics to map IL-1 beta production and response patterns within tissue architecture.
Development of computational models to predict IL-1 beta-mediated cellular crosstalk in heterogeneous tissues.
Therapeutic targeting:
Development of mouse models with humanized IL-1 signaling components to better translate findings to human disease.
Exploration of cell type-specific IL-1 beta neutralization strategies.
Investigation of temporal aspects of IL-1 blockade to identify critical windows for therapeutic intervention.
These emerging research areas promise to deepen our understanding of IL-1 beta's complex roles in health and disease and may lead to more refined therapeutic approaches for inflammatory and metabolic disorders .
Mouse models studying IL-1 beta provide valuable insights for translational research, though with important considerations for human application:
Comparative biology insights:
Mouse IL-1 beta shares 65-78% amino acid sequence identity with human IL-1 beta, with higher conservation in functional domains.
Both species use similar processing mechanisms via inflammasomes and caspase-1.
The IL-1 receptor system and downstream signaling pathways are highly conserved between mice and humans.
These similarities allow reasonable prediction of human responses from mouse data, while acknowledging species differences.
Age-related metabolic dysfunction:
Mouse studies showing that IL-1 beta contributes to age-associated decline in beta cell function have direct relevance to human type 2 diabetes, where aging is the primary risk factor.
The finding that myeloid cell-derived IL-1 beta specifically impacts beta cell function suggests targeted therapeutic approaches for human metabolic diseases.
Data from IL-1 beta knockout mice showing preserved insulin secretion with age aligns with clinical trials of IL-1 blocking agents in humans with diabetes.
Inflammatory disease mechanisms:
Mouse models have revealed that tissue-specific sources of IL-1 beta (e.g., from myeloid cells versus parenchymal cells) have distinct impacts on disease progression.
This insight has implications for designing more precise human therapeutic strategies that target IL-1 beta from specific cellular sources rather than global inhibition.
Therapeutic development considerations:
Evidence from mouse islets showing that IL-1 beta reduces expression of key genes like E2f1, Ins2, and Kir6.2 provides molecular targets for monitoring therapeutic efficacy in humans.
The observation that IL-1 beta and IL-1Ra balance changes during mouse aging suggests that age-appropriate dosing of IL-1 blocking agents may be necessary in human clinical applications.
Mouse studies demonstrating that IL-1 beta impacts both acute function and long-term proliferative capacity suggest that human therapeutic strategies should consider both immediate and sustained effects.
Biomarker development:
Mouse research identifying downstream molecular targets of IL-1 beta can inform the development of biomarkers to monitor treatment efficacy in human patients.
Understanding the temporal dynamics of IL-1 beta effects in mice helps in designing optimal sampling protocols for human clinical trials.
By carefully considering both the similarities and differences between mouse and human IL-1 beta biology, researchers can maximize the translational value of mouse studies for developing and refining human therapeutic approaches for inflammatory, metabolic, and age-related diseases .
IL-1β is synthesized as an inactive 31 kDa pro-form that accumulates in the cytosol . The activation of IL-1β requires cleavage by inflammasomes, which are multi-protein complexes that respond to pathogens, stress conditions, and other danger signals . The active form of IL-1β is a 17 kDa protein that is then released to exert its biological effects.
IL-1β is a potent mediator of inflammation and immune responses. It can induce fever, hypotension, and the release of adrenocorticotrophic hormone. Additionally, IL-1β stimulates the production of other cytokines, which further amplify the inflammatory response . This cytokine is involved in various inflammatory diseases, including sepsis, arthritis, and skin hypersensitivity reactions .
Recombinant murine IL-1β is produced using Escherichia coli (E. coli) as the expression system . The recombinant protein is a non-glycosylated polypeptide chain containing 153 amino acids and has a molecular mass of approximately 17.5 kDa . The protein is purified using proprietary chromatographic techniques to ensure high purity and biological activity.
Recombinant IL-1β is widely used in laboratory research to study its role in inflammation and immune responses. It is also used to develop and test anti-inflammatory drugs. For instance, transgenic mice expressing a luciferase gene driven by the IL-1β promoter have been used to monitor IL-1β expression in vivo using bioluminescence imaging . This model helps researchers understand the transcriptional regulation of IL-1β and evaluate the efficacy of potential therapeutic agents.
Lyophilized recombinant murine IL-1β is stable at room temperature for up to three weeks but should be stored desiccated below -18°C for long-term storage . Upon reconstitution, it should be stored at 4°C for short-term use and below -18°C for long-term use. To prevent degradation, it is recommended to add a carrier protein such as human serum albumin (HSA) or bovine serum albumin (BSA) and avoid freeze-thaw cycles .