Recombinant IL-1β Human, His is synthesized using optimized bacterial expression systems:
Expression: Codon-optimized E. coli strains ensure high yield .
Purification: Nickel-affinity chromatography targets the His tag, followed by ion-exchange and size-exclusion chromatography .
Formulation: Lyophilized in phosphate buffer (pH 7.1–7.5) with stabilizers like trehalose .
IL-1β Human, His binds the IL-1 receptor (IL-1R1), activating downstream pathways such as NF-κB and MAPK, which drive inflammatory responses . Key functional roles include:
Immune Modulation: Induces thymocyte proliferation, B-cell maturation, and fibroblast growth factor activity .
Inflammasome Activation: Processed by caspase-1 into its mature form, enabling secretion .
Proinflammatory Effects: Stimulates prostaglandin release, adhesion molecule expression, and fever .
Autoinflammatory Syndromes: Used to study conditions like CAPS (Cryopyrin-Associated Periodic Syndromes) linked to NLRP3 inflammasome mutations .
Cancer Research: Facilitates investigation of IL-1β’s dual role in tumor progression and suppression (e.g., breast cancer metastasis, chemoresistance in mesothelioma) .
Drug Screening: Serves as a target for IL-1β inhibitors (e.g., anakinra, canakinumab) in rheumatoid arthritis and type 2 diabetes .
Biomarker Analysis: Quantified via ultrasensitive assays like Simoa® (LLOQ: 0.008 pg/mL) .
IL-1β blockade is effective in treating:
GMP Compliance: ELISA kits (e.g., resDetect™) validate IL-1β levels in CAR-T cell therapies .
Stability: Lyophilized protein remains stable for years at -80°C; reconstituted aliquots last 2–7 days at 4°C .
Gene Regulation: The rs16944 polymorphism in IL1B correlates with reduced mRNA stability in hyperglycemic patients .
Monocyte Subsets: Non-classical monocytes exhibit lower IL-1β secretion due to Hsp27-mediated mRNA decay .
Cancer Microenvironments: B cells enhance IL-1β-driven invasiveness in triple-negative breast cancer via NF-κB activation .
The IL-1b His tag protein is supplied in a solution containing 20mM Tris-HCl at pH 8.0 and 50% glycerol.
The purity is determined to be greater than 95.0% using SDS-PAGE analysis.
APVRSLNCTLRDSQQKSLVMSGPYELKALHLQGQDMEQQVVFSMSFV
QGEESNDKIPVALGLKEKNLYLSCVLKDDKPTLQLESVDPKNYPKKKME
KRFVFNKIEINNKLEFESAQFPNWYISTSQAENMPVFLGGTKGGQDITDFTMQFVSS
IL-1β (Interleukin-1 beta) is a proinflammatory cytokine encoded by the IL1B gene in humans. It serves as a critical mediator of inflammatory responses and is primarily produced by activated macrophages, monocytes, and a subset of dendritic cells known as slanDC . IL-1β exhibits multiple biological functions, including:
Mediation of inflammatory responses
Involvement in cellular proliferation, differentiation, and apoptosis
Induction of cyclooxygenase-2 in the central nervous system, contributing to inflammatory pain hypersensitivity
Amplification of B and T lymphocyte proliferation in response to antigens and mitogens
Stimulation of neutrophilia and acute phase protein production
Upregulation of chemokines and other inflammatory cytokines
Regulation of zinc and iron redistribution in tissues
Modulation of corticosteroid and glucose homeostasis
Functioning as a potent adjuvant in antigen-specific antibody responses
IL-1β is exceptionally potent, functioning at picomolar concentrations to elicit downstream effects as part of both innate and adaptive immunity .
Human IL-1β possesses a conserved β-trefoil conformation characteristic of the IL-1 family of cytokines. Its structure includes:
12 β-sheets forming a β-trefoil architecture
A central hydrophobic core
Six β-sheets (β1, β4, β5, β8, β9, and β12) arranged in an anti-parallel β-barrel configuration
Six β-hairpins with consecutive β-sheet naming starting from the N-terminus
Key functional loops, including β4/5 and β11/12, which are crucial for interactions with the IL-1 receptor accessory protein (IL-1RAcP)
The three-dimensional structure of IL-1β has been determined at high resolution using X-ray crystallography. Despite sharing structural similarity with other IL-1 family members, IL-1β has relatively low sequence identity with these proteins .
Unlike constitutively expressed IL-1α, IL-1β expression is highly regulated and limited to specific cell types:
Transcription initiation: IL-1β is transcribed following activation of monocytes, macrophages, and dendritic cells through:
Production as inactive precursor: IL-1β is initially synthesized as an inactive precursor protein (pro-IL-1β)
Proteolytic activation: The precursor requires proteolytic processing by caspase-1 (also known as interleukin-1β converting enzyme or ICE) to generate the bioactive form
Inflammasome involvement: Caspase-1 activation requires inflammasome assembly, which is triggered by danger-associated molecular patterns (DAMPs)
This two-step regulation (transcriptional control plus proteolytic activation) provides tight control over IL-1β activity, which is crucial given its potent inflammatory effects.
IL-1β binding to its receptors involves a complex multi-step process:
Primary receptor binding: IL-1β binds to IL-1 receptor type I (IL-1RI), which contains three immunoglobulin-like domains (D1, D2, and D3) forming two distinct binding sites (A and B)
Interface characteristics:
Total buried surface area: 1932 Ų over 47 residues
Site A (formed by D1/D2): ~1000 Ų over 25 amino acids
Site B (formed by D3): Nearly equivalent sized interface over 21 amino acids
Five of six β-sheets from the Ig fold of D3 participate in the interface
A hydrogen bond forms between IL-1β and the linker between D1/2 and D3
Accessory protein recruitment: Following IL-1β binding to IL-1RI, the IL-1 receptor accessory protein (IL-1RAcP) is recruited to form a functional signaling complex
Critical structural elements: The β4/5 and β11/12 loops of IL-1β are essential for IL-1RAcP recruitment and subsequent signaling
Understanding these binding mechanisms has been crucial for developing therapeutics targeting IL-1β signaling, including the design of receptor antagonists with enhanced potency.
Research has revealed that chronic IL-1β exposure can induce epithelial-to-mesenchymal transition (EMT) in non-small cell lung cancer cells, a phenomenon with significant implications for cancer progression:
Gradual EMT progression: A subset of non-small cell lung cancer cells undergoes a gradually progressing EMT phenotype following 21-day exposure to IL-1β
EMT memory: The EMT and associated phenotypes (enhanced cell invasion, PD-L1 upregulation, chemoresistance) are sustained even after IL-1β withdrawal, a phenomenon termed "EMT memory"
SLUG-dependence: The transcription factor SLUG is indispensable for establishing EMT memory, and high SLUG expression in lung cancer patients correlates with poor survival
Epigenetic regulation: Chromatin immunoprecipitation and methylation-specific PCR revealed SLUG-mediated temporal regulation of epigenetic modifications, including:
Therapeutic implications: Chemical inhibition of DNA methylation restored E-cadherin expression in EMT memory cells and increased their susceptibility to chemotherapy-induced apoptosis
These findings highlight the complex role of IL-1β in cancer progression and suggest potential therapeutic strategies targeting EMT memory in tumors.
Dysregulation of IL-1β signaling contributes to numerous pathological conditions, including sepsis, rheumatoid arthritis, inflammatory bowel disease, acute and chronic myelogenous leukemia, insulin-dependent diabetes mellitus, atherosclerosis, neuronal injury, and aging-related diseases . Several approaches for IL-1β inhibition have been developed:
Receptor antagonists: Anakinra, a recombinant version of the natural IL-1 receptor antagonist (IL-1Ra), was the first FDA-approved IL-1 inhibitor (2001). IL-1Ra binds IL-1RI with high affinity but cannot recruit IL-1RAcP, thereby preventing signal transduction
Engineered antagonists: Structural knowledge of IL-1β/IL-1RI interactions has enabled the development of more potent inhibitors:
Targeting processing: Inhibition of caspase-1 to prevent pro-IL-1β cleavage represents another approach to limiting IL-1β activity
Neutralizing antibodies: Antibodies targeting IL-1β directly can prevent receptor binding
When designing IL-1β inhibition strategies, researchers should consider:
The specific disease context and relative contribution of IL-1β
Potential compensatory mechanisms through other cytokines
Systemic effects of IL-1β blockade on immune responses
Binding site preferences and kinetics of inhibitory molecules
Production of high-quality recombinant human IL-1β with a histidine tag requires careful consideration of expression systems and purification strategies:
Expression system selection:
E. coli: Most commonly used for IL-1β expression due to simplicity and high yield
Mammalian cells: Preferred when post-translational modifications are required
Baculovirus-infected insect cells: Offers intermediate complexity with higher protein folding fidelity than bacteria
Construct design considerations:
Position of His-tag: N-terminal tags are generally preferred as they minimize interference with the IL-1β receptor binding domains
Linker sequence: Including a flexible linker (e.g., GGGGS) between the His-tag and IL-1β sequence can reduce steric hindrance
Protease cleavage site: Incorporating a TEV or enterokinase site allows tag removal if needed for functional studies
Codon optimization: Adjust codons based on the expression system for improved protein yield
Purification protocol:
Initial capture: Immobilized metal affinity chromatography (IMAC) using Ni-NTA or Co-NTA resins
Secondary purification: Size exclusion chromatography to remove aggregates and ensure monomeric protein
Endotoxin removal: Critical for experiments where endotoxin contamination could confound IL-1β effects
Quality control: SDS-PAGE, Western blot, and functional assays to confirm identity and activity
Optimizing solubility:
Expression temperature: Lower temperatures (16-25°C) often improve proper folding
Lysis buffer composition: Including mild detergents and appropriate salt concentrations
Avoiding aggregation: Addition of stabilizing agents like glycerol or arginine
Activity verification:
IL-1β receptor binding assays
Cell-based assays measuring downstream signaling activation
Comparison with commercial non-tagged IL-1β standards
The addition of a histidine tag to IL-1β can potentially impact its structural integrity and biological activity:
Several robust methods can be employed to assess IL-1β activity in research settings:
NF-κB activation assays:
Reporter cell lines: Cells transfected with an NF-κB responsive element driving luciferase or GFP expression
EMSA (Electrophoretic Mobility Shift Assay): Detects NF-κB translocation to nucleus
Immunofluorescence: Visualizes p65 nuclear translocation
Typical detection range: 0.1-10 ng/ml IL-1β
Cytokine induction:
ELISA/MSD assays: Measure downstream cytokines (IL-6, IL-8, TNF-α) induced by IL-1β
qPCR: Quantifies upregulation of inflammatory gene expression
Multiplex bead arrays: Simultaneously measures multiple cytokines
Most sensitive detection method, with responses at 10-100 pg/ml IL-1β
Cellular phenotypic assays:
Signaling pathway analysis:
Western blotting: Detects phosphorylation of pathway components (IKK, IκB, MAPKs)
Phospho-flow cytometry: Measures signaling at single-cell resolution
Inhibitor studies: Pharmacological dissection of IL-1β-activated pathways
Considerations for His-tagged IL-1β:
Include appropriate controls (commercial non-tagged IL-1β)
Account for potential endotoxin contamination (include polymyxin B controls)
Test for tag-specific artifacts using anti-His antibodies as blocking controls
Based on research showing IL-1β-induced epigenetic modifications in cancer cells , the following methodologies are recommended for studying these changes:
DNA methylation analysis:
Methylation-specific PCR: Detects methylation status of specific promoters (e.g., CDH1/E-cadherin)
Bisulfite sequencing: Provides single-nucleotide resolution of methylation patterns
Genome-wide methylation arrays: Identifies global methylation changes across the genome
RRBS (Reduced Representation Bisulfite Sequencing): Cost-effective approach for genome-scale methylation analysis
Histone modification profiling:
ChIP-qPCR: Measures specific histone marks (H3K27me3, H3K9me3) at regions of interest
ChIP-seq: Genome-wide mapping of histone modifications
Cut&Run or CUT&Tag: More sensitive alternatives to traditional ChIP
Western blotting: Global levels of specific histone modifications
Chromatin accessibility:
ATAC-seq: Maps open chromatin regions genome-wide
DNase-seq: Identifies DNase I hypersensitive sites
MNase-seq: Determines nucleosome positioning
Transcription factor binding:
ChIP-qPCR/ChIP-seq for SLUG and other EMT-associated transcription factors
DNA-protein interaction assays (e.g., electrophoretic mobility shift assay)
Experimental design considerations:
Time-course experiments: Capture dynamic changes during chronic exposure
IL-1β withdrawal studies: Assess persistence of epigenetic modifications
Inhibitor studies: Use of DNMT inhibitors (5-azacytidine), histone methyltransferase inhibitors, or histone deacetylase inhibitors to reverse changes
Functional validation: Correlate epigenetic changes with phenotypic outcomes
IL-1β plays multifaceted roles in cancer biology, with significant implications for therapeutic strategies:
Cancer-promoting mechanisms:
Chronic inflammation: IL-1β is a key mediator of inflammation, a recognized hallmark of cancer
EMT induction: Prolonged IL-1β exposure drives epithelial-to-mesenchymal transition in cancer cells, promoting invasiveness
PD-L1 upregulation: IL-1β-induced EMT is associated with increased PD-L1 expression, potentially contributing to immune evasion
Chemoresistance: Cancer cells exposed to chronic IL-1β develop resistance to chemotherapeutic agents
Epigenetic reprogramming: IL-1β induces stable epigenetic changes that persist even after cytokine withdrawal
Therapeutic strategies targeting IL-1β in cancer:
Direct IL-1β inhibition: Antagonists and neutralizing antibodies
Epigenetic modifiers: DNA methyltransferase inhibitors can reverse IL-1β-induced epigenetic changes and restore chemosensitivity
SLUG inhibition: Preventing SLUG upregulation blocks IL-1β-induced EMT
Combination approaches: Pairing IL-1β inhibition with chemotherapy or immunotherapy
Biomarker potential:
IL-1β represents an evolutionarily conserved cytokine with fundamental roles in immune defense:
Evolutionary conservation:
Fundamental defensive functions:
Species-specific adaptations:
Variations in receptor binding interfaces
Differences in regulatory mechanisms
Species-specific inhibitory strategies
Implications for research:
Animal models may not fully recapitulate human IL-1β biology
Species-specific antibodies and reagents are necessary for research
Understanding evolutionary context helps interpret experimental results
Several innovative approaches for modulating IL-1β activity in inflammatory conditions have emerged:
Enhanced receptor antagonists:
Targeting IL-1β processing:
Inflammasome inhibitors preventing caspase-1 activation
Inhibitors of non-canonical processing pathways
Receptor-targeted approaches:
Antibodies blocking IL-1RI
Soluble receptor decoys
Inhibitors of IL-1RAcP recruitment
Downstream signaling inhibition:
Selective inhibitors of IL-1β-activated kinases
Novel NF-κB pathway modulators
Cell type-specific delivery:
Nanoparticle-based targeting of IL-1β inhibitors to specific cell populations
Gene therapy approaches for localized IL-1Ra production
Combination therapies:
IL-1β inhibition plus TNF-α blockade for synergistic anti-inflammatory effects
IL-1β antagonism with metabolic modulators for conditions like diabetes
Researchers working with recombinant IL-1β often encounter batch-to-batch variability that can confound experimental results:
Standardization practices:
Activity normalization: Establish a bioactivity unit rather than relying solely on protein concentration
Reference standards: Maintain an internal reference standard to calibrate each new batch
Functional validation: Test each batch in a dose-response assay against a consistent cell line
Documentation: Maintain detailed records of source, lot numbers, and activity measurements
Quality control measures:
Endotoxin testing: Use LAL assays to confirm acceptably low endotoxin levels
Purity assessment: SDS-PAGE and mass spectrometry to verify protein integrity
Aggregation analysis: Size exclusion chromatography or dynamic light scattering
Receptor binding assays: Surface plasmon resonance to confirm binding kinetics
Experimental design considerations:
Single-batch experiments: Complete comparative studies using a single batch when possible
Internal controls: Include a consistent positive control in each experiment
Parallel testing: Run old and new batches side-by-side before switching
Calibration curves: Generate batch-specific dose-response curves
Storage optimization:
Aliquoting: Prepare single-use aliquots to avoid freeze-thaw cycles
Stabilizing additives: Consider carrier proteins or stabilizers for dilute solutions
Temperature monitoring: Ensure consistent storage conditions
Stability testing: Periodically test stored aliquots for activity retention
Several methodological challenges can complicate the interpretation of IL-1β signaling studies:
Endotoxin contamination:
Issue: Bacterial endotoxins can mimic or synergize with IL-1β effects
Solution: Rigorous endotoxin testing, inclusion of polymyxin B controls, endotoxin-removal protocols
Receptor expression heterogeneity:
Issue: Variable IL-1RI and IL-1RAcP expression across cell types and passages
Solution: Characterize receptor expression, consider receptor transfection for consistent expression, include positive control cell lines
Context-dependent responses:
Issue: IL-1β effects vary with cell density, serum conditions, and culture duration
Solution: Standardize culture conditions, perform time-course and dose-response studies, consider 3D culture systems
Signal amplification and feedback:
Issue: IL-1β induces its own expression, complicating interpretation of primary vs. secondary effects
Solution: Include protein synthesis inhibitors, use short time points for primary effects, consider IL-1Ra to block secondary signaling
His-tag specific considerations:
Issue: The His-tag may interfere with certain aspects of IL-1β function
Solution: Compare with untagged protein, consider tag removal, verify activity in multiple assay systems
Cell type selection:
Issue: Not all cell types respond equivalently to IL-1β
Solution: Validate responsiveness, consider primary cells vs. cell lines, examine heterogeneity within populations
Timing considerations:
Issue: Different signaling pathways and outcomes have distinct kinetics
Solution: Detailed time-course experiments, pulse-chase designs, consider both acute and chronic exposure models
IL-1β is initially produced as an inactive precursor protein, known as pro-IL-1β, which is synthesized in response to inflammatory stimuli. This precursor is a 31 kDa protein that accumulates in the cytosol of cells such as monocytes, macrophages, and dendritic cells . The activation of inflammasomes, which are multi-protein complexes responding to pathogens and stress conditions, triggers the processing of the caspase-1 precursor into its active form. Caspase-1 then cleaves pro-IL-1β into its active 17 kDa form .
Recombinant IL-1β proteins are produced using various expression systems, such as E. coli. These recombinant proteins often include a polyhistidine tag (His Tag) at the N-terminus to facilitate purification and detection . The His Tag allows for easy purification using nickel affinity chromatography, which binds to the histidine residues.
For example, the recombinant human IL-1β protein with a His Tag is expressed from E. coli cells and contains amino acids Ala117-Ser269 . This protein has a calculated molecular weight of approximately 19.3 kDa and migrates as 19-20 kDa under reducing conditions in SDS-PAGE . The purity of this protein is typically greater than 95% as determined by SDS-PAGE .
IL-1β is a potent immunomodulator that mediates a wide range of immune and inflammatory responses. It signals through two receptors, IL-1RI and IL-1RII, both of which are shared with IL-1 alpha . The activity of IL-1β can be moderated by the IL-1 Receptor Antagonist (IL-1RA), which blocks receptor binding through competitive inhibition .
IL-1β plays a significant role in innate host defense by triggering the production of other proinflammatory cytokines in target cells and initiating acute-phase responses to infection and injury . Elevated levels of IL-1β have been associated with various chronic inflammatory conditions, making it a target for therapeutic interventions .