IL-6 Human Recombinant produced in E.Coli migrates to 25kDa and is fused to a 6 amino acid his tag at its C-terminus. IL-6 is purified by proprietary chromatographic technique.
Human IL-6 is a multifunctional cytokine that folds as a four α-helix bundle structure with up-up-down-down topology. The human IL-6 polypeptide spans from Pro29 to Met212 and has a molecular weight of approximately 25 kDa when produced recombinantly with a 6-amino acid histidine tag at the C-terminus . IL-6 functions as a broad-acting cytokine that contributes to host defense through stimulation of acute phase responses, hematopoiesis, and immune reactions .
The structure-function relationship of IL-6 has been extensively studied, with specific regions of the protein identified as critical for receptor binding. Research has shown that at least two regions of human IL-6 are crucial for efficient binding to the human IL-6 receptor: residues 178-184 in helix D and residues 63-113 in the region incorporating part of the putative connecting loop AB through to the beginning of helix C . These structural elements contribute to the diverse portfolio of functions that IL-6 performs in normal physiology and disease.
The IL-6 receptor system has a unique configuration that enables its diverse biological activities. IL-6 exerts its effects through two main molecules: IL-6R (IL-6 receptor) and gp130 . The signaling process follows these steps:
IL-6 binds to mIL-6R (membrane-bound form of IL-6R)
This binding induces homodimerization of gp130
A high-affinity functional receptor complex forms, consisting of IL-6, IL-6R, and gp130
The homodimerization of this receptor complex activates Janus kinases (JAKs)
JAKs phosphorylate tyrosine residues in the cytoplasmic domain of gp130
This activation triggers two main signaling pathways :
The gp130 Tyr759-derived SHP-2/ERK MAPK pathway
The gp130 YXXQ-mediated JAK/STAT pathway
Interestingly, IL-6 can also signal through a mechanism known as trans-signaling, where the soluble form of IL-6R (sIL-6R) binds with IL-6, and this IL-6-sIL-6R complex can then form a complex with gp130 . This alternative pathway allows IL-6 to affect cells that do not express membrane-bound IL-6R.
The 6-amino acid histidine tag (His-tag) fused to the C-terminus of recombinant human IL-6 serves several important research purposes :
Purification efficiency: The His-tag allows for single-step purification using metal affinity chromatography, resulting in high purity (>95% as determined by 10% PAGE with Coomassie staining)
Detection capability: The tag facilitates easier detection of the protein in experimental settings using anti-His antibodies
Minimal interference: When positioned at the C-terminus, the His-tag typically has minimal impact on the biological activity of IL-6
For research applications, His-tagged IL-6 is typically supplied in phosphate-buffered saline with 25mM K₂CO₃ . For optimal stability during long-term storage, it is recommended to add a carrier protein (0.1% HSA or BSA) and avoid multiple freeze-thaw cycles.
Human and mouse IL-6 display distinct species-specific activities despite having a relatively high degree of sequence similarity (42%) . These differences are important considerations for experimental design and interpretation:
Species-Specific Activity | Human IL-6 | Mouse IL-6 |
---|---|---|
Active on human cells | Yes | No |
Active on mouse cells | Yes | Yes |
The structural basis for these species-specific activities has been investigated through the construction of human/mouse IL-6 hybrid molecules. Research has identified that specific residues in helix D (positions 178-184) and in the region from connecting loop AB through the beginning of helix C (residues 63-113) are critical for efficient binding to the human IL-6 receptor .
For human IL-6, interactions between residues Ala-180, Leu-181, and Met-184 and residues in the N-terminal region appear to be critical for maintaining the structure of the molecule. Replacement of these residues with the corresponding residues in mouse IL-6 resulted in a significant loss of α-helical content and a 200-fold reduction in activity in mouse bioassays .
Measuring IL-6 signaling requires careful selection of methods that capture both immediate signal transduction events and downstream biological effects. A comprehensive approach should include:
Phosphorylation assays: Monitor JAK-STAT pathway activation by measuring phosphorylation of STAT3 (Tyr705) using:
Western blotting
Phospho-specific flow cytometry
ELISA-based phosphoprotein quantification
Reporter systems:
Construct STAT3-responsive reporter cell lines using luciferase or fluorescent protein readouts
Monitor activation of SOCS3 (suppressor of cytokine signaling 3) promoter, which is rapidly induced by IL-6
Transcriptional profiling:
qPCR analysis of canonical IL-6 responsive genes: SOCS3, CRP, SAA
RNA-seq for comprehensive transcriptional response profiling
Functional assays tailored to cell type:
Hepatocytes: Measure acute phase protein production (CRP, SAA, fibrinogen)
B cells: Analyze antibody production and plasma cell differentiation
T cells: Assess Th17 differentiation and IL-17 production
Myeloid cells: Evaluate changes in differentiation markers and cytokine production
When working with His-tagged IL-6, it is important to include appropriate controls to ensure that observed effects are due to IL-6 bioactivity rather than artifacts from the His-tag or preparation method. Biological activity validation can be performed using established cell lines such as HepG2 (human) or 7TD1 (mouse) .
Distinguishing between classic IL-6 signaling (via membrane-bound IL-6R) and trans-signaling (via soluble IL-6R) is crucial for understanding IL-6 biology in different physiological and pathological contexts. Researchers can employ the following methodological approaches:
Selective pathway inhibition:
Use sgp130Fc (soluble gp130-Fc fusion protein) to specifically block trans-signaling without affecting classic signaling
Compare with pan-IL-6 inhibitors like anti-IL-6 antibodies or anti-IL-6R antibodies that block both pathways
Cell-specific receptor expression analysis:
Flow cytometry to quantify membrane IL-6R expression on target cells
ELISA to measure soluble IL-6R in experimental systems
Genetically modified systems:
Use IL-6R knockout cells reconstituted with either membrane-bound or soluble IL-6R
Employ cells expressing non-cleavable IL-6R (resistant to shedding) to isolate classic signaling
Pathway-specific biomarkers:
Monitor differential gene expression patterns characteristic of each pathway
Analyze pathway-specific phosphorylation profiles
Signaling Type | Key Components | Inhibition Strategy | Characteristic Cellular Responses |
---|---|---|---|
Classic Signaling | IL-6 + membrane IL-6R + gp130 | Anti-IL-6R antibodies (e.g., tocilizumab) | Acute phase response, fever, hematopoiesis |
Trans-Signaling | IL-6 + soluble IL-6R + gp130 | sgp130Fc | Chronic inflammation, tissue remodeling, T cell recruitment |
This methodological distinction is particularly important when evaluating the potential therapeutic applications of IL-6 pathway inhibitors, as blocking specific modes of IL-6 signaling may provide more targeted approaches with fewer side effects .
The structure-function relationship of IL-6 has significant implications for both basic research and therapeutic development. Several structural considerations deserve attention:
Critical binding domains:
Research utilizing human/mouse IL-6 hybrid molecules has identified two regions that are crucial for efficient binding to the human IL-6 receptor:
Residues 178-184 in helix D
Residues 63-113 in the region incorporating part of the connecting loop AB through to the beginning of helix C
Mutations in these regions can dramatically alter receptor binding and biological activity.
Conformational integrity:
The alpha-helical content of IL-6 is essential for its proper function. Specific interactions between residues Ala-180, Leu-181, and Met-184 and residues in the N-terminal region are critical for maintaining the structure of human IL-6. Disruption of these interactions can lead to decreased alpha-helical content and reduced biological activity .
Post-translational modifications:
While recombinant E. coli-produced IL-6 lacks glycosylation, mammalian-expressed IL-6 can have variable glycosylation patterns that may affect:
Protein stability and half-life
Receptor binding kinetics
Immunogenicity profiles
Engineered variants:
Researchers have developed modified IL-6 molecules with altered properties:
Antagonistic variants that bind but do not activate signaling
Super-agonists with enhanced receptor binding or stability
Targeted fusion proteins for tissue-specific delivery
For therapeutic applications, understanding these structural determinants has guided the development of IL-6-targeted biologics such as tocilizumab, a humanized anti-IL-6 receptor antibody that has shown exceptional efficacy in treating rheumatoid arthritis and juvenile idiopathic arthritis .
Characterizing the role of IL-6 in disease models requires comprehensive experimental approaches that address both molecular mechanisms and functional outcomes. The following methodological framework is recommended:
Loss and gain of function studies:
Genetic approaches: IL-6 knockout models, conditional IL-6 or IL-6R deletion
Pharmacological approaches: Anti-IL-6 antibodies, anti-IL-6R antibodies (e.g., tocilizumab), small molecule inhibitors of downstream signaling
Overexpression models: Tissue-specific IL-6 overexpression, hydrodynamic delivery of IL-6 expression constructs
Temporal analysis of IL-6 signaling:
Acute vs. chronic IL-6 elevation
Time-course studies of signaling pathway activation
Inducible expression systems for temporal control
Spatial characterization:
Tissue-specific IL-6 production and response profiling
Cell-type specific IL-6R expression mapping
In situ analysis of IL-6 responsive genes
Integration with other cytokine networks:
Combinatorial cytokine treatments
Analysis of compensatory mechanisms in IL-6-deficient systems
Comparison of IL-6 blockade with inhibition of other inflammatory cytokines
In disease-specific contexts, functional outcome measures should be tailored to the condition being studied. For example:
Disease Model | Key IL-6 Functions | Essential Outcome Measures |
---|---|---|
Rheumatoid Arthritis | Joint inflammation, synovial hyperplasia, bone erosion | Joint histology, inflammatory infiltrate, cartilage/bone integrity, synovial fluid analysis |
Systemic Juvenile Idiopathic Arthritis | Systemic inflammation, fever, acute phase response | CRP/SAA levels, fever patterns, growth parameters, organ function |
Cancer Models | Tumor growth, metastasis, immune evasion | Tumor size, metastatic burden, tumor-infiltrating lymphocyte analysis, cancer stem cell markers |
Infection Models | Host defense, tissue injury control | Pathogen clearance, tissue damage assessment, survival analysis, immune cell recruitment |
Research has demonstrated that IL-6 plays diverse roles across these disease contexts, with both protective and pathological effects depending on the timing, duration, and context of expression .
Optimizing recombinant IL-6 production and purification is critical for generating high-quality protein for research applications. The following methodological considerations are important:
Expression system selection:
E. coli: Typically yields high amounts of protein but lacks post-translational modifications. Commonly used for His-tagged IL-6 production
Mammalian cells: Provide proper folding and post-translational modifications but at lower yields
Insect cells: Offer a balance between yield and post-translational processing
Construct design considerations:
Purification strategy:
For His-tagged IL-6:
Immobilized metal affinity chromatography (IMAC) using Ni-NTA or Co-based resins
Selection of appropriate imidazole concentrations for binding, washing, and elution
Consideration of pH and salt concentrations to minimize non-specific binding
Quality control measures:
Purity assessment: SDS-PAGE with Coomassie staining (>95% purity is standard for research-grade IL-6)
Endotoxin testing: Critical for eliminating LPS contamination that could confound immunological experiments
Bioactivity validation: Cell-based assays using IL-6 responsive cell lines (e.g., HepG2, 7TD1)
Mass spectrometry confirmation of protein identity and integrity
Storage considerations:
Researchers should be aware that different purification and storage conditions can affect IL-6 bioactivity. Validation of each batch's activity using standardized bioassays is recommended before use in critical experiments.
When designing experiments to study IL-6 across species, researchers must account for the species-specific activities and structural differences that impact receptor binding and downstream signaling. The following methodological considerations are essential:
Species specificity awareness:
Human IL-6 is active on both human and mouse cells, while mouse IL-6 is active only on mouse cells . This asymmetrical cross-reactivity has important implications for experimental design, particularly when:
Using human IL-6 in mouse models
Developing translational studies from mouse to human
Interpreting xenograft studies where both human and mouse cells are present
Structural basis of species specificity:
Research has identified critical regions responsible for species-specific activities:
Residues 178-184 in helix D
Residues 63-113 in the region from connecting loop AB through the beginning of helix C
These regions should receive particular attention when analyzing sequences or designing mutants.
Experimental validation approaches:
In vitro bioassays: Compare activity using species-specific cell lines:
Human: HepG2 hepatocellular cells for acute phase protein induction
Mouse: 7TD1 hybridoma cells for proliferation assays
Receptor binding studies: Direct measurement of binding affinities to receptors from different species using:
Surface plasmon resonance
Bio-layer interferometry
Cell-based binding assays with labeled IL-6
Controls and standardization:
Include both species-matched and cross-species positive and negative controls
Standardize IL-6 concentrations across experiments using international units where possible
Consider using hybrid molecules as reference standards for comparative studies
Data interpretation framework:
Experimental Setting | Key Considerations | Recommended Approach |
---|---|---|
Human cells + mouse IL-6 | Minimal/no activity expected | Include human IL-6 positive control; consider higher doses of mouse IL-6 |
Mouse cells + human IL-6 | Activity expected but potentially with different potency | Perform dose-response studies; compare EC50 values with mouse IL-6 |
Humanized mouse models | Complex interplay of human and mouse components | Carefully track source of measured IL-6; consider potential feedback loops |
In vivo translation of in vitro findings | Species differences in downstream effects | Validate key findings with species-appropriate reagents |
Understanding these cross-species interactions is particularly important when developing and testing IL-6-targeted therapeutics, as preclinical studies in animal models may not fully predict human responses due to these species-specific differences .
Interleukin-6 (IL-6) is a potent pro-inflammatory cytokine primarily produced by activated T cells, endothelial cells, and macrophages. It plays a crucial role in immune responses, acute phase reactions, and hematopoiesis. The recombinant form of IL-6, tagged with a His tag, is widely used in research to study its molecular characteristics and functions.
The human recombinant IL-6 with a His tag is produced in Escherichia coli (E. coli) and migrates to approximately 25 kDa. The His tag, consisting of six histidine residues, is fused to the C-terminus of the protein, facilitating its purification through affinity chromatography . The recombinant IL-6 is supplied as a sterile filtered clear solution in phosphate-buffered saline and 25 mM potassium carbonate .
IL-6 is involved in a wide range of biological activities, including:
The recombinant IL-6 with a His tag is extensively used in immunological studies to explore its functions and interactions. It enables researchers to delve deeper into cytokine-mediated immune responses and understand the signaling pathways involved . The His tag allows for easy purification and detection of the protein, making it a valuable tool in laboratory research.
For short-term use, the recombinant IL-6 should be stored at 4°C. For long-term storage, it is recommended to keep it frozen at -20°C, with the addition of a carrier protein such as 0.1% human serum albumin (HSA) or bovine serum albumin (BSA) to prevent degradation. Multiple freeze-thaw cycles should be avoided to maintain the protein’s integrity .