IL 6 Human

Interleukin-6 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to Interleukin 6 (Human)

Interleukin 6 (Human), commonly abbreviated as IL-6, is a 20.8–28 kDa glycoprotein encoded by the IL6 gene on chromosome 7p21 . It functions as a pleiotropic cytokine with dual roles in pro-inflammatory responses and anti-inflammatory myokine activity . Produced by immune cells (e.g., macrophages, T cells), endothelial cells, and fibroblasts, IL-6 regulates immune homeostasis, hematopoiesis, metabolism, and tissue repair .

Molecular Structure and Receptor Signaling

IL-6 signals through a receptor complex composed of:

  • IL-6Rα (CD126): Ligand-binding subunit

  • gp130 (CD130): Signal-transducing subunit

Key Signaling Pathways

PathwayMechanismBiological Impact
ClassicalIL-6 binds membrane-bound IL-6Rα, activating gp130-JAK/STAT3 cascadeAcute-phase proteins, immune cell differentiation
Trans-signalingIL-6 binds soluble IL-6R (sIL-6R), enabling gp130 activation in non-immune cellsAngiogenesis, neuronal survival
Trans-presentationDendritic cells present IL-6/IL-6R complexes to T cellsTh17 cell differentiation

Structural studies reveal critical binding residues (e.g., R167, E171, R178, R181) in IL-6’s D-helix that modulate receptor affinity and gp130 activation .

Immune Regulation

  • Acute-phase response: Induces hepatic production of C-reactive protein (CRP) and serum amyloid A .

  • B cell maturation: Stimulates plasma cell differentiation and IgG production .

  • T cell polarization: Promotes Th17 differentiation (with TGF-β) and inhibits Treg development .

Metabolic and Homeostatic Functions

  • Hepcidin induction: Reduces iron availability during inflammation .

  • Energy allocation: Mobilizes lipids during exercise and transiently suppresses immunity .

Disease Associations

ConditionIL-6 RoleTherapeutic Target Status
Rheumatoid arthritisDrives synovial inflammation via Th17 cells and osteoclast activation Approved (tocilizumab, sarilumab)
COVID-19Correlates with severe hyperinflammation and poor prognosis Emergency use (anti-IL-6R agents)
Neuromyelitis opticaDisrupts blood-brain barrier via astrocyte activation Phase III trials (satralizumab)

Normal Physiological Levels

  • Healthy adults: Pooled mean = 5.186 pg/mL (95% CI: 4.631–5.740 pg/mL) .

  • Age correlation: Increases by 0.05 pg/mL per year .

Recent Advances in IL-6 Biology (2023–2024)

  • Structural insights: Mutagenesis studies identified R181 as non-essential for IL-6R binding but critical for gp130 complex stability .

  • Therapeutic innovations: Bispecific antibodies targeting IL-6 and TNF-α show efficacy in refractory autoimmune diseases .

  • Metabolic reprogramming: IL-6 trans-signaling enhances mitochondrial respiration in cachexia models .

Product Specs

Introduction
Interleukin 6 (IL-6) is a multifunctional cytokine that plays a role in various biological processes. It is crucial for the differentiation of B cells into antibody-secreting cells and influences the growth of myeloma and plasmacytoma cells. Additionally, IL-6 is involved in nerve cell differentiation and the induction of acute-phase reactants in hepatocytes.
Description
Recombinant Human Interleukin-6, produced in E. coli, is a single, non-glycosylated polypeptide chain consisting of 184 amino acids. With a molecular weight of 21 kDa, this purified protein is obtained through proprietary chromatographic techniques.
Physical Appearance
Sterile Filtered White lyophilized powder.
Formulation
The protein is lyophilized from a 0.2 μm filtered concentrated solution (1 mg/mL) in phosphate-buffered saline (PBS) at pH 7.4.
Solubility
To reconstitute the lyophilized Interleukin-6, it is recommended to dissolve it in sterile 18 MΩ-cm H2O to a concentration of at least 100 μg/mL. This solution can be further diluted in other aqueous solutions as needed.
Stability
Lyophilized Interleukin-6 remains stable at room temperature for up to 3 weeks. However, for long-term storage, it is recommended to store the desiccated protein below -18°C. After reconstitution, store IL-6 at 4°C for 2-7 days. For future use, store below -18°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for extended storage. Avoid repeated freeze-thaw cycles.
Purity
The purity of the protein is greater than 97.0% as determined by two methods: Reverse-Phase High-Performance Liquid Chromatography (RP-HPLC) analysis and Sodium Dodecyl Sulfate-Polyacrylamide Gel Electrophoresis (SDS-PAGE) analysis.
Biological Activity
The biological activity, represented by ED50 (half-maximal effective concentration), is determined by the dose-dependent stimulation of murine 7TD1 cells. The ED50 is less than 0.1 ng/mL, corresponding to a specific activity of 1.0 x 10,000,000 units per mg.
Protein Content
Protein quantification was performed using two independent methods. The first method involved UV spectroscopy at 280 nm, utilizing an absorbance value of 0.47 as the extinction coefficient for a 0.1% (1 mg/mL) solution. This value is calculated using the PC GENE computer analysis program (IntelliGenetics). The second method utilized RP-HPLC analysis with a standard solution of IL-6 as a reference standard.
Synonyms

B cell differentiation factor, BCDF, BSF-2, HPGF, HSF, MGI-2, B-cell stimulatory factor 2,Hybridoma growth factor, CTL differentiation factor, CDF, IL-6, HGF.

Source
Escherichia Coli.
Amino Acid Sequence
The sequence of the first five N-terminal amino acids was determined and was found to be Met-Pro-Val-Pro-Pro.

Q&A

What are the primary biological functions of human IL-6?

IL-6 is a multifunctional cytokine involved in diverse physiological processes. Its primary functions include:

  • Regulation of immune responses, particularly B-cell differentiation and antibody production

  • Mediation of acute and chronic inflammatory responses

  • Involvement in hematopoiesis (blood cell formation)

  • Participation in bone metabolism

  • Contribution to embryonic development

  • Regulation of metabolic processes similar to hormones

IL-6 was originally cloned as a factor acting on B cells to induce immunoglobulin production, but its role extends far beyond this initial discovery. It promotes antibody production both directly by acting on plasma cells and indirectly by promoting follicular CD4+ T cell differentiation. IL-6-deficient mice show impaired immune responses against various pathogens and reduced T-cell-dependent antibody responses, confirming its essential role in immune function .

How does IL-6 differ from other pro-inflammatory cytokines in structure and function?

Unlike many pro-inflammatory cytokines that primarily activate one signaling pathway, IL-6 exhibits unique aspects:

  • Utilizes both classical signaling (via membrane-bound IL-6 receptor) and trans-signaling (via soluble IL-6 receptor)

  • Activates multiple downstream pathways including JAK/STAT3, SHP-2, and MAPK cascades

  • Displays hormone-like properties affecting metabolic processes related to lipid metabolism, insulin resistance, and neuroendocrine function

  • Functions in both pro-inflammatory and anti-inflammatory contexts depending on the signaling mode and cellular environment

This functional versatility distinguishes IL-6 from cytokines like TNFα and IL-1β, which predominantly drive pro-inflammatory responses. The unique ability of IL-6 to utilize trans-signaling allows it to affect virtually all cells in the body through ubiquitously expressed gp130, even those that do not express membrane-bound IL-6R .

What cells primarily produce IL-6 during normal physiological conditions versus inflammatory states?

Normal physiological conditions:

  • Low levels of IL-6 are produced by:

    • Fibroblasts

    • Endothelial cells

    • Some immune cells at basal states

    • Adipose tissue

Inflammatory states:

  • Dramatically increased production by:

    • Activated macrophages and monocytes

    • T lymphocytes

    • Fibroblasts (particularly in rheumatoid arthritis)

    • Endothelial cells

    • Non-immune cells in tissues under stress conditions

During pathological conditions, IL-6 production can be triggered by various stimuli including lipopolysaccharide (LPS), damage-associated molecular patterns (DAMPs), and other pro-inflammatory cytokines like IL-1β and TNFα. Experimental evidence shows that breast cancer cell lines significantly upregulate IL-6 expression following stimulation with pathogen-associated molecular patterns (PAMPs) or DAMPs, demonstrating how non-immune cells can become major sources of IL-6 during inflammatory conditions .

What are the differences between classical IL-6 signaling and trans-signaling pathways?

Classical IL-6 Signaling:

  • Requires cells to express membrane-bound IL-6 receptor alpha (IL-6Rα)

  • IL-6 binds to membrane-bound IL-6Rα, which then associates with glycoprotein 130 (gp130)

  • Primarily mediates homeostatic and anti-inflammatory effects

  • Limited to cells expressing IL-6Rα (mainly hepatocytes, some leukocytes, and megakaryocytes)

Trans-Signaling:

  • Utilizes soluble IL-6 receptor alpha (sIL-6Rα)

  • IL-6 binds to sIL-6Rα in fluid phase, and this complex can activate any cell expressing gp130

  • Predominantly mediates pro-inflammatory responses

  • Can affect virtually all cell types in the body due to ubiquitous expression of gp130

  • Significantly elevated in inflammatory and autoimmune diseases

A third mechanism, known as trans-presentation, involves IL-6 bound to IL-6Rα on dendritic cells directly acting on CD4+ T cells through gp130 to induce Th17 cells. This differs from trans-signaling as it is not inhibited by soluble gp130, which efficiently blocks trans-signaling but not trans-presentation or classical signaling .

How do STAT3 and NF-κB interact in IL-6-mediated inflammation?

The interaction between STAT3 and NF-κB represents a critical synergistic mechanism in IL-6-mediated inflammation:

  • Molecular interaction: IL-6 primarily activates STAT3 through JAK kinases, while other inflammatory signals activate NF-κB.

  • Amplification loop: When both transcription factors are simultaneously activated, they form a positive feedback amplification mechanism called the "IL-6 amplifier" (IL-6 Amp):

    • NF-κB activation leads to IL-6 production

    • IL-6 activates STAT3

    • Activated STAT3 enhances NF-κB activation

    • This leads to further increased IL-6 production and additional pro-inflammatory cytokines

  • Disease relevance: This synergistic interaction has been demonstrated in models of rheumatoid arthritis, where F759 mice (which have enhanced STAT3 signaling due to lack of negative regulation by SOCS3) develop spontaneous arthritis that is further enhanced by NF-κB activators.

This amplification loop explains how localized inflammation can become chronic and systemic. The IL-6 Amp not only increases IL-6 production but also amplifies various pro-inflammatory cytokines and growth factors that are targets of the NF-κB pathway, creating a self-sustaining inflammatory environment .

What are the key regulatory mechanisms that control IL-6 signaling?

Several regulatory mechanisms tightly control IL-6 signaling to prevent excessive inflammation:

  • SOCS3 (Suppressor of Cytokine Signaling 3):

    • Primary negative regulator induced by STAT3

    • Binds to phosphorylated tyrosine residues on gp130 and inhibits JAK activity

    • Creates a negative feedback loop to limit IL-6 signaling duration

  • Protein inhibitors of activated STATs (PIAS):

    • Directly inhibit STAT3 DNA-binding and transcriptional activity

    • Function as E3-SUMO ligases that modify STAT3

  • Protein tyrosine phosphatases (PTPs):

    • Dephosphorylate JAKs and activated STATs

    • Include SHP-2, PTP1B, and TC-PTP

  • TRAF3 (TNF receptor-associated factor 3):

    • Inhibits IL-6–STAT3 signaling in B cells

    • Loss-of-function mutations in TRAF3 are observed in B-cell lymphoma and multiple myeloma

  • Soluble gp130 (sgp130):

    • Acts as a natural inhibitor of IL-6 trans-signaling

    • Selectively blocks the pro-inflammatory trans-signaling pathway without affecting classical signaling

Dysregulation of these control mechanisms, particularly SOCS3, has been implicated in chronic inflammatory diseases. Mice with the F759 mutation in gp130, which prevents SOCS3 binding, develop spontaneous arthritis with increased STAT3 activation, demonstrating the importance of these regulatory pathways .

How does IL-6 contribute to the pathogenesis of autoimmune diseases?

IL-6 contributes to autoimmune disease pathogenesis through multiple mechanisms:

  • Immune cell dysregulation:

    • Promotes differentiation of pathogenic Th17 cells while inhibiting regulatory T cell development

    • Enhances B-cell differentiation into antibody-producing plasma cells, contributing to autoantibody production

    • Drives the development of follicular helper T cells (Tfh) that support germinal center reactions

  • Tissue-specific effects:

    • Activates synovial fibroblasts in rheumatoid arthritis, creating a hyperplastic synovium

    • Promotes osteoclast differentiation, contributing to bone erosion

    • Induces acute phase proteins from the liver that perpetuate systemic inflammation

  • IL-6 amplifier activation:

    • Synergistic interaction between STAT3 and NF-κB creates a positive feedback loop

    • Local tissue inflammation becomes self-sustaining and chronic

    • Non-immune cells become significant sources of inflammatory mediators

  • Trans-signaling predominance:

    • Increased levels of sIL-6R in autoimmune conditions expand IL-6 effects to all gp130-expressing cells

    • Shifts IL-6 activity from homeostatic to predominantly pro-inflammatory

Clinical and experimental evidence strongly supports IL-6's role in autoimmunity. IL-6-deficient mice are resistant to several experimental autoimmune disease models including collagen-induced arthritis, antigen-induced arthritis, and experimental autoimmune encephalomyelitis. Human studies show correlation between IL-6 levels and disease severity in rheumatoid arthritis and other inflammatory autoimmune diseases .

What is the role of IL-6 in cancer development and progression?

IL-6 influences multiple aspects of cancer biology:

  • Tumor cell proliferation and survival:

    • Activates STAT3, which regulates genes involved in proliferation, survival, and resistance to apoptosis

    • Functions as a direct growth factor for certain cancers (e.g., multiple myeloma, hepatocellular carcinoma)

  • Tumor microenvironment modulation:

    • Promotes angiogenesis through VEGF induction

    • Recruits immunosuppressive cells including myeloid-derived suppressor cells

    • Inhibits dendritic cell maturation, reducing anti-tumor immune responses

    • Shifts CD4+ T cell differentiation away from anti-tumor Th1 cells toward Th2 and Th17 phenotypes

  • Metastatic capacity enhancement:

    • Induces epithelial-to-mesenchymal transition (EMT)

    • Increases cancer stem cell-like properties

    • Promotes tissue invasion and metastatic colonization

  • Therapeutic resistance:

    • Activates survival pathways that confer resistance to chemotherapy and targeted therapies

    • Creates a pro-inflammatory environment that supports tumor recurrence

Studies show IL-6 production by both tumor cells and stromal cells within the tumor microenvironment. Early evidence of IL-6's role in cancer came from observations that cardiac myxoma cells produce IL-6, and IL-6 transgenic mice develop plasmacytomas. More recent genomic analyses have found gain-of-function mutations in IL-6 signaling components (particularly gp130) in inflammatory hepatocellular tumors, while loss-of-function mutations in negative regulators like TRAF3 are found in B-cell lymphomas and multiple myeloma .

How did IL-6 contribute to COVID-19 pathophysiology, and what methodological approaches were used to study this?

IL-6 played a significant role in COVID-19 pathophysiology, particularly in severe cases:

  • Contribution to cytokine storm:

    • Elevated IL-6 levels correlated with disease severity and mortality

    • Participated in the hyperinflammatory state leading to acute respiratory distress syndrome (ARDS)

    • Promoted recruitment of inflammatory cells to the lungs

  • Trans-signaling activation:

    • Increased levels of soluble IL-6 receptor were observed in severe COVID-19

    • Trans-signaling expanded IL-6 effects beyond cells expressing membrane-bound IL-6R

    • This contributed to widespread endothelial activation and coagulopathy

  • IL-6 amplifier activation:

    • SARS-CoV-2 infection activated both NF-κB and STAT3 pathways

    • Created synergistic feedback loop further enhancing cytokine production

    • Non-immune pulmonary cells became significant sources of inflammatory mediators

Methodological approaches used to study IL-6 in COVID-19:

  • Clinical biomarker studies:

    • Quantitative ELISA measurements of serum IL-6 and sIL-6R in patient cohorts

    • Correlation analyses between IL-6 levels and clinical outcomes

    • Longitudinal monitoring of IL-6 during disease progression

  • Cellular models:

    • In vitro infection of human airway epithelial cells

    • Analysis of IL-6 production following SARS-CoV-2 infection

    • Assessment of IL-6 signaling pathway activation using phospho-flow cytometry

  • Therapeutic intervention studies:

    • Clinical trials of IL-6 receptor antagonists (e.g., tocilizumab, sarilumab)

    • Stratification of patients based on inflammatory markers

    • Evaluation of timing for intervention in relation to disease course

The involvement of IL-6 in COVID-19 pathophysiology built upon previous understanding of IL-6 in acute and chronic inflammation, demonstrating how fundamental cytokine biology research can inform clinical responses to emerging diseases .

What are the most reliable methods for measuring human IL-6 in different biological samples?

Different experimental contexts require specific approaches for measuring IL-6:

1. Enzyme-Linked Immunosorbent Assay (ELISA):

  • Applications: Gold standard for quantifying IL-6 protein in serum, plasma, and cell culture supernatants

  • Methodology: Typically uses sandwich ELISA format with capture and detection antibodies

  • Advantages: High specificity, good sensitivity (typically 0.5-5 pg/mL), well-established commercial kits

  • Limitations: Cannot distinguish between free IL-6 and IL-6 bound to soluble receptors

2. Multiplex Bead-Based Assays:

  • Applications: Simultaneous measurement of IL-6 alongside other cytokines in limited sample volumes

  • Methodology: Uses antibody-coated beads with distinct fluorescent signatures

  • Advantages: Allows assessment of cytokine networks, reduces sample requirement

  • Limitations: Potential cross-reactivity, sometimes less sensitive than dedicated ELISA

3. Quantitative Real-Time PCR (qRT-PCR):

  • Applications: Measurement of IL-6 mRNA expression in cells and tissues

  • Methodology: RNA extraction, reverse transcription, and qPCR with specific primers

  • Advantages: Detects transcriptional regulation, applicable to tissue samples

  • Limitations: Does not reflect post-transcriptional regulation or protein secretion

4. Flow Cytometry:

  • Applications: Identification of IL-6-producing cells and intracellular IL-6

  • Methodology: Cell permeabilization and staining with fluorochrome-conjugated anti-IL-6 antibodies

  • Advantages: Single-cell resolution, can be combined with cell phenotyping

  • Limitations: Requires protein transport inhibitors (like Brefeldin A), semi-quantitative

5. Immunohistochemistry/Immunofluorescence:

  • Applications: Localization of IL-6 production within tissue architecture

  • Methodology: Tissue fixation, sectioning, and staining with anti-IL-6 antibodies

  • Advantages: Preserves spatial information, can identify cellular sources

  • Limitations: Limited quantification, potential background staining

The choice of method depends on research questions and sample types. For clinical studies, standardized ELISAs with established reference ranges are preferred. Research contexts may benefit from combining methods, such as using qRT-PCR to assess transcriptional regulation alongside ELISA to confirm protein production .

How can researchers effectively study IL-6 signaling pathways in experimental models?

Studying IL-6 signaling requires multi-faceted approaches:

1. Cell-Based Signaling Assays:

  • Phospho-flow cytometry: Measures phosphorylation of STAT3 (Y705) at single-cell level

  • Western blotting: Detects activation of JAK/STAT3, SHP-2, MAPK, and AKT pathways

  • Luciferase reporter assays: Monitors STAT3-dependent transcriptional activity

  • Proximity ligation assays: Visualizes protein-protein interactions in signaling complexes

2. Receptor Dynamics Assessment:

  • Surface receptor quantification: Flow cytometry with fluorescently-labeled anti-IL-6R antibodies

  • Receptor shedding measurement: ELISA for soluble IL-6R in culture supernatants

  • Receptor internalization studies: Fluorescence microscopy with labeled IL-6

3. Pathway Dissection Strategies:

  • Small molecule inhibitors: JAK inhibitors (e.g., ruxolitinib), STAT3 inhibitors

  • siRNA/shRNA knockdown: Targeting specific pathway components

  • CRISPR/Cas9 gene editing: Creating signaling component knockouts or mutations

  • Dominant-negative constructs: Overexpression of signaling-deficient mutants

4. Classical vs. Trans-Signaling Discrimination:

  • Selective pathway activation: Using IL-6/sIL-6R complex vs. IL-6 alone

  • Selective pathway inhibition: Using sgp130-Fc to block trans-signaling only

  • Cell-type specific responses: Comparing cells with/without membrane IL-6R expression

5. In Vivo Signaling Analysis:

  • Conditional knockout models: Cell-type specific deletion of gp130 or STAT3

  • Pathway-reporter mice: Expressing luciferase under STAT3-responsive promoters

  • Phospho-protein analysis in tissues: Immunohistochemistry for phospho-STAT3

  • Adoptive transfer experiments: Using cells with modified signaling components

An integrated approach combining multiple techniques provides the most comprehensive understanding of IL-6 signaling. For example, researchers studying rheumatoid arthritis models have combined conditional STAT3 knockout in specific cell populations with phospho-flow cytometry and RNA-sequencing to map the cell type-specific contributions to disease pathology .

What experimental design considerations are important when studying IL-6 in disease models?

Critical experimental design considerations include:

1. Model Selection and Validation:

  • Disease relevance: Ensure the model recapitulates key aspects of human pathology

  • IL-6 dependency validation: Confirm IL-6 involvement using neutralizing antibodies or knockout approaches

  • Species differences awareness: Human and mouse IL-6 biology have important distinctions

  • Genetic background consideration: Strain-dependent differences affect IL-6 responses (e.g., C57BL/6 vs. BALB/c)

2. IL-6 Source and Stimulation Parameters:

  • Recombinant protein quality: Use low-endotoxin preparations to avoid TLR activation

  • Concentration range determination: Establish dose-response relationships (physiological vs. pathological)

  • Temporal dynamics assessment: Acute vs. chronic exposure affects outcomes

  • Combined cytokine effects: Consider IL-6 in context of other inflammatory mediators

3. Signaling Pathway Dissection:

  • Classical vs. trans-signaling distinction: Use selective tools (sgp130-Fc, IL-6/sIL-6R complex)

  • Downstream pathway inhibition: Apply specific JAK/STAT inhibitors

  • Cell type-specific analysis: Use conditional knockouts or cell sorting

  • Feedback mechanism evaluation: Assess SOCS3 induction and function

4. Readout Selection and Timing:

  • Acute response markers: Phosphorylation events (minutes to hours)

  • Transcriptional changes: Gene expression profiling (hours)

  • Phenotypic consequences: Cell differentiation, proliferation (days)

  • Disease-specific outcomes: Tissue pathology, clinical parameters (days to weeks)

5. Controls and Reproducibility Measures:

  • Pathway activation controls: Include IL-6 stimulation time course

  • Inhibitor specificity controls: Test effects on related cytokine pathways

  • Genetic model validation: Confirm knockout efficiency at protein level

  • Biological replicates: Account for variability in primary cell responses

6. Translational Considerations:

  • Human sample correlation: Compare findings with patient material when possible

  • Therapeutic intervention timing: Test preventive vs. established disease treatment

  • Biomarker identification: Develop measures to track IL-6 activity in vivo

The F759 mouse model provides an excellent example of rigorous experimental design. These mice, which have enhanced STAT3 activation due to impaired negative regulation, spontaneously develop age-dependent arthritis that resembles human rheumatoid arthritis. Studies with this model systematically examined cell type-specific contributions, interactions with environmental factors, and synergistic effects with other inflammatory pathways .

How do post-translational modifications affect IL-6 function and signaling outcomes?

Post-translational modifications (PTMs) of IL-6 and its signaling components create complex regulatory mechanisms:

1. IL-6 Protein Modifications:

  • N-glycosylation at Asn73: Affects protein stability and receptor binding affinity

  • Disulfide bonds: Critical for proper protein folding and bioactivity

  • Proteolytic processing: Can generate variants with altered receptor interactions

  • Oxidation: Reactive oxygen species can modify IL-6 structure and function

2. Receptor Component Modifications:

  • IL-6Rα shedding: ADAM17-mediated proteolytic cleavage generates sIL-6R

  • gp130 phosphorylation patterns: Different tyrosine residues activate distinct pathways

  • Receptor glycosylation: Affects protein stability and ligand binding properties

  • Receptor ubiquitination: Regulates internalization and degradation kinetics

3. Signaling Intermediate Modifications:

  • STAT3 acetylation: Enhances transcriptional activity

  • STAT3 serine phosphorylation (S727): Modulates transcriptional output

  • JAK SUMOylation: Affects kinase activity and stability

  • NF-κB post-translational modifications: Acetylation and phosphorylation alter interactions with STAT3

4. Methodological Approaches to Study PTMs:

  • Mass spectrometry: Identifies specific modification sites and stoichiometry

  • Phospho-specific antibodies: Detect activation status of signaling components

  • Site-directed mutagenesis: Determines functional consequences of specific modifications

  • Inhibitors of PTM enzymes: Reveals the role of specific modifications in signaling outcomes

5. Disease Relevance:

  • Altered glycosylation patterns: Observed in inflammatory conditions

  • Increased ADAM17 activity: Enhances sIL-6R generation in inflammatory diseases

  • Dysregulated phosphatase activity: Affects STAT3 activation duration

  • Oxidative stress effects: Modifies IL-6 and signaling components in inflammatory environments

Understanding the complex interplay between different PTMs requires integrated approaches combining proteomic analysis with functional studies. For example, researchers have mapped the differential tyrosine phosphorylation patterns on gp130 to distinct downstream signaling events, with STAT3 activation primarily mediated through phosphorylation at Y705, while Y759 phosphorylation creates a binding site for the negative regulator SOCS3 .

How does the microbiome influence IL-6 production and signaling in health and disease?

The microbiome-IL-6 relationship represents an emerging frontier in understanding inflammatory regulation:

1. Microbiome-Mediated IL-6 Induction:

  • Pattern recognition receptor activation: Microbial components trigger IL-6 production via TLRs and NOD-like receptors

  • Metabolite-mediated effects: Short-chain fatty acids and other bacterial metabolites modulate IL-6 production

  • Intestinal barrier function: Microbiome composition affects gut permeability and subsequent systemic inflammation

  • Trained immunity effects: Prior microbial exposure conditions innate immune cells' IL-6 response

2. IL-6 Effects on Microbiome Composition:

  • Intestinal epithelial regeneration: IL-6 promotes epithelial proliferation after injury

  • Antimicrobial peptide regulation: IL-6 signaling affects defensin production

  • Mucus layer composition: IL-6 influences goblet cell function and mucin production

  • Immune surveillance modulation: IL-6 shapes mucosal immune responses to commensals

3. Disease-Specific Interactions:

  • Inflammatory bowel disease: Dysbiosis correlates with altered IL-6 production

  • Rheumatoid arthritis: Specific microbiome signatures associated with IL-6-driven inflammation

  • Cancer: Microbiome influences IL-6 production in the tumor microenvironment

  • Metabolic disorders: Microbiome-derived signals affect IL-6 production by adipose tissue

4. Experimental Approaches:

  • Gnotobiotic models: Comparing germ-free and colonized animals for IL-6 responses

  • Selective bacterial colonization: Using defined bacterial communities to identify species-specific effects

  • Fecal microbiota transplantation: Transferring microbiomes between diseased and healthy subjects

  • Metagenomic analysis with cytokine profiling: Correlating microbiome composition with IL-6 levels

5. Therapeutic Implications:

  • Probiotic interventions: Specific bacteria may modulate IL-6 production

  • Prebiotic approaches: Dietary components that shape microbiome and influence IL-6

  • Fecal transplantation: Potential to normalize IL-6 responses through microbiome restoration

  • Combined approaches: Targeting microbiome alongside direct IL-6 inhibition

This bidirectional relationship creates complex feedback loops, where IL-6-driven inflammation can alter microbiome composition, which in turn affects IL-6 production. Research in this area requires integration of microbiome sequencing technologies with immunological methods to assess IL-6 signaling and functional outcomes in relevant tissue environments .

How do genetic variants in the IL-6 signaling pathway contribute to disease susceptibility and therapeutic responses?

Genetic variation in IL-6 pathway components has significant implications for personalized medicine:

1. Promoter and Regulatory Region Variants:

  • IL-6 -174 G>C polymorphism: Affects transcriptional regulation and baseline IL-6 levels

  • IL-6R promoter variants: Influence receptor expression levels

  • Enhancer region polymorphisms: Alter tissue-specific and stimulus-responsive IL-6 production

  • microRNA binding site variations: Affect post-transcriptional regulation of IL-6 and pathway components

2. Receptor and Signaling Component Variants:

  • IL-6R Asp358Ala variant: Increases IL-6R shedding, affecting trans-signaling/classical signaling balance

  • gp130 Gly148Arg polymorphism: Associated with reduced IL-6 responsiveness and decreased myocardial infarction risk

  • JAK and STAT3 variants: Alter signaling efficiency and duration

  • SOCS3 polymorphisms: Affect negative regulation of the pathway

3. Disease Associations:

  • Rheumatoid arthritis: IL-6 pathway variants contribute to disease susceptibility and severity

  • Cardiovascular disease: IL-6R variants modify inflammation-related cardiovascular risk

  • Cancer susceptibility: Polymorphisms affecting IL-6 production linked to certain cancers

  • Infectious disease severity: Genetic variants influence cytokine storm risk in infections like COVID-19

4. Therapeutic Response Prediction:

  • Anti-IL-6 therapy response: Genetic variants predict differential response to IL-6 blockade

  • Biomarker development: Genetic profiles combined with protein measurements improve prediction

  • Adverse event susceptibility: Variants associated with infection risk during IL-6 targeted therapy

  • Drug dosing considerations: Pharmacogenetic factors affecting drug metabolism and IL-6 pathway

5. Research Methodologies:

  • Genome-wide association studies (GWAS): Identify IL-6 pathway variants associated with diseases

  • Targeted sequencing approaches: Deep characterization of pathway components

  • Functional genomics: CRISPR-based screening to assess variant effects

  • Ex vivo patient sample analysis: Testing variant effects on cellular responses

  • Systems biology integration: Combining genetic data with other omics for comprehensive modeling

Experimental evidence has linked several IL-6 pathway genetic variations to disease outcomes. For example, genome-wide association studies, meta-analyses, and SNP mapping have associated IL-6, IL-6R, and gp130 variants with inflammatory diseases such as rheumatoid arthritis and coronary artery disease. The frequency of a polymorphism in the IL-6 gene promoter that correlates with lower plasma IL-6 levels is reduced in systemic-onset juvenile chronic arthritis, suggesting a protective effect of reduced IL-6 production .

What are the critical considerations for developing and validating IL-6 assays for research applications?

Developing robust IL-6 assays requires attention to several factors:

1. Antibody Selection and Validation:

  • Epitope considerations: Choose antibodies recognizing stable, accessible epitopes

  • Cross-reactivity testing: Validate specificity against related cytokines (IL-11, LIF, OSM)

  • Affinity determination: Select antibodies with appropriate affinity for desired sensitivity

  • Paired antibody compatibility: For sandwich assays, ensure epitopes don't interfere

  • Lot-to-lot consistency: Establish quality control procedures for antibody performance

2. Standard Preparation and Calibration:

  • Recombinant protein source: Use fully characterized, bioactive recombinant IL-6

  • Reference material alignment: Calibrate against international standards when available

  • Matrix effects assessment: Evaluate potential interference from sample components

  • Stability testing: Determine optimal storage conditions and freeze-thaw stability

  • Dilution linearity verification: Confirm proportional response across dilution range

3. Sample Handling Protocols:

  • Collection standardization: Establish consistent procedures (anticoagulants, processing times)

  • Pre-analytical variables control: Minimize ex vivo activation during processing

  • Storage condition optimization: Determine temperature and preservative requirements

  • Potential interferent identification: Test effects of hemolysis, lipemia, and medication

  • Freeze-thaw cycles limitation: Validate maximum acceptable number

4. Assay Performance Characteristics:

  • Sensitivity requirements: Define lower limit of quantification needed for application

  • Dynamic range determination: Ensure coverage of physiological and pathological ranges

  • Precision assessment: Evaluate intra-assay and inter-assay variability

  • Accuracy verification: Compare with reference methods or spike-recovery experiments

  • Robustness testing: Evaluate performance across operators and laboratory conditions

5. Validation for Specific Applications:

  • Matrix-specific validation: Different requirements for serum, plasma, cell culture, tissue lysates

  • Disease-state testing: Verify performance in samples with potential interfering factors

  • Clinical correlation studies: Relate measurements to clinical parameters when relevant

  • Reference range establishment: Define normal values for relevant populations

  • Reproducibility across laboratories: For multi-center studies

The Quantikine Human IL-6 Immunoassay represents a well-validated example, designed as a 4.5-hour solid-phase ELISA for measuring IL-6 in cell culture supernatants, serum, and plasma. Such standardized assays undergo rigorous validation to ensure reliable performance across different sample types and experimental conditions .

How can researchers accurately distinguish between classical and trans-signaling effects of IL-6 in experimental systems?

Distinguishing between these signaling modes requires specialized experimental approaches:

1. Selective Pathway Activation:

  • IL-6 alone vs. IL-6/sIL-6R complex: IL-6 activates only classical signaling in cells lacking membrane IL-6R, while the complex activates both pathways

  • Engineered fusion proteins: Hyper-IL-6 (covalently linked IL-6/sIL-6R) selectively activates trans-signaling

  • Cell-type specific responses: Compare effects on cells known to express or lack membrane IL-6R

  • Dose-response relationships: Classical and trans-signaling may have different concentration thresholds

2. Selective Pathway Inhibition:

  • sgp130-Fc: Selectively blocks trans-signaling without affecting classical signaling

  • Anti-IL-6R antibodies: Block both pathways by preventing IL-6 binding to both membrane and soluble IL-6R

  • Anti-IL-6 antibodies: Inhibit both pathways by neutralizing the cytokine

  • Membrane IL-6R shedding inhibitors: ADAM17 inhibitors reduce sIL-6R generation

3. Genetic Manipulation Approaches:

  • IL-6R knockout cells/animals: Assess trans-signaling by adding IL-6/sIL-6R complex

  • Non-sheddable IL-6R mutants: Express IL-6R resistant to proteolytic cleavage

  • Cell-type specific IL-6R deletion: Tissue-specific knockout of IL-6R

  • gp130 signaling mutants: Selectively disrupt specific downstream pathways

4. Analytical and Visualization Methods:

  • Phospho-flow cytometry: Compare STAT3 phosphorylation in membrane IL-6R+ vs. IL-6R- cells

  • Immunofluorescence microscopy: Visualize receptor localization and internalization

  • Transcriptomic profiling: Identify gene signatures specific to each signaling mode

  • Reporter cell lines: Develop specific readouts for classical vs. trans-signaling

5. In Vivo Approaches:

  • sgp130-Fc transgenic mice: Selective blockade of trans-signaling in vivo

  • Conditional IL-6R knockout models: Cell-type specific deletion of membrane IL-6R

  • sIL-6R measurement: Quantify soluble receptor levels in biological fluids

  • Tissue-specific response assessment: Compare responses in tissues with different IL-6R expression

The differential regulation of these pathways has significant implications for disease mechanisms. For example, in COVID-19, increased sIL-6R levels in severe disease suggest enhanced trans-signaling, which explains the systemic inflammatory effects in tissues that don't express membrane IL-6R. Selective inhibition of trans-signaling using sgp130-Fc can provide targeted anti-inflammatory effects while preserving the homeostatic functions of classical signaling .

What methodological approaches help resolve contradictory findings in IL-6 research literature?

Addressing contradictions requires systematic investigation:

1. Experimental System Standardization:

  • Cell/animal model selection: Different models may have inherently different IL-6 responses

  • Reagent consistency: Use validated, consistent sources of recombinant proteins and antibodies

  • Protocol harmonization: Standardize experimental conditions, timing, and concentrations

  • Positive and negative controls: Include well-characterized controls in all experiments

  • Blinding procedures: Implement where appropriate to reduce bias

2. Context-Dependent Effects Analysis:

  • Cell type specificity: Systematically compare effects across relevant cell types

  • Microenvironment reconstruction: Consider the influence of other cytokines, growth factors

  • Temporal dynamics assessment: Evaluate both acute and chronic effects

  • Concentration-response relationships: Test across physiological and pathological ranges

  • Species differences consideration: Compare human and mouse systems directly

3. Signaling Pathway Dissection:

  • Classical vs. trans-signaling discrimination: Use specific tools to distinguish pathways

  • Downstream pathway inhibitors: Apply selective JAK/STAT3 vs. SHP2/MAPK inhibitors

  • Genetic models: Use pathway component knockouts or mutations

  • Single-cell analysis: Account for heterogeneous responses within populations

4. Technical Validation Approaches:

  • Orthogonal methods: Confirm findings using independent techniques

  • Antibody validation: Verify specificity using knockout controls

  • Detection method comparison: ELISA vs. bioassay vs. mass spectrometry for IL-6 quantification

  • Internal replication: Repeat critical experiments independently

5. Integration with Clinical Data:

  • Human sample validation: Test hypotheses in primary patient samples

  • Biomarker correlation studies: Link experimental findings to clinical parameters

  • Pharmacological validation: Test effects of clinically used IL-6 inhibitors

  • Genetic association studies: Connect to human genetic variants in IL-6 pathway

6. Meta-Analysis and Systematic Review:

  • Literature curation: Comprehensive assessment of published findings

  • Quality assessment: Evaluate methodology of contradictory studies

  • Statistical power analysis: Consider sample size and variability

  • Publication bias consideration: Account for unpublished negative findings

A concrete example comes from contradictory findings regarding IL-6's role in T helper cell differentiation. While initial studies suggested IL-6 primarily promotes Th2 differentiation and inhibits Th1 responses, later work found that IL-6 induced by repeated inflammation can actually drive Th1 responses in certain contexts like peritoneal fibrosis. Systematic investigation revealed that the effect depends on the cytokine microenvironment, timing of exposure, and differentiation stage of the T cells, reconciling seemingly contradictory observations .

What are the current frontiers in IL-6 research and promising future directions?

IL-6 research continues to evolve along several exciting frontiers:

  • Single-cell technologies: Advances in single-cell RNA sequencing and proteomics are revealing previously unappreciated heterogeneity in IL-6 responses across cell populations and identifying novel IL-6-responsive cell types. This approach is particularly valuable for understanding IL-6's role in complex tissue environments like the tumor microenvironment and inflamed synovium.

  • Spatial biology integration: New technologies combining single-cell resolution with spatial information are mapping IL-6 production and signaling within tissue architecture, providing insights into local cytokine gradients and cell-cell interactions that regulate IL-6 responses.

  • Systems biology approaches: Computational modeling of IL-6 signaling networks is helping to predict emergent properties and explain complex phenotypes. These models integrate genomic, transcriptomic, and proteomic data to simulate pathway dynamics under different conditions.

  • Targeted therapeutic approaches: Beyond current monoclonal antibody therapies, research is exploring selective inhibition of trans-signaling, development of small molecule inhibitors with improved tissue penetration, and cell type-specific delivery systems to reduce side effects.

  • Biomarker development: Multi-parameter biomarker panels are being developed to identify patients most likely to benefit from IL-6-targeted therapies, predict treatment responses, and monitor disease activity with greater precision.

  • Metabolic interface exploration: The relationship between IL-6 signaling and cellular metabolism is emerging as a crucial regulatory axis, with implications for understanding IL-6's role in metabolic diseases and cancer.

  • Structural biology advances: Cryo-electron microscopy is providing unprecedented insights into the three-dimensional structure of IL-6 receptor complexes, facilitating structure-based drug design and understanding of signaling mechanisms.

These frontiers are expanding our understanding of IL-6 biology beyond traditional immunology into areas such as neurobiology, metabolism, and aging. As research progresses, we can anticipate more nuanced and targeted approaches to modulating IL-6 activity in disease, moving beyond complete blockade to selective pathway modulation with improved therapeutic indices .

Product Science Overview

Structure and Production

IL-6 is produced by a variety of cell types, including monocytes, fibroblasts, endothelial cells, and T cells . The recombinant form of IL-6 is typically produced in E. coli and is used in various research and clinical applications .

Functions

IL-6 has several key functions:

  • Immune Response: It promotes the differentiation of B and T cells, particularly Th17 cells .
  • Inflammation: IL-6 is involved in the acute phase response and can induce fever in individuals with autoimmune diseases or infections .
  • Hematopoiesis: It supports the proliferation of hematopoietic progenitor cells and hybridoma cells .
Clinical Significance

Disturbed IL-6 production has been associated with various pathological processes, including inflammatory autoimmune diseases such as rheumatoid arthritis and certain cancers . Due to its significant role in these conditions, IL-6 is a target for therapeutic interventions.

Applications

Recombinant human IL-6 is used in:

  • Cell Culture: It is used to promote the differentiation and proliferation of various cell types .
  • Functional Assays: IL-6 is employed in assays to study its biological activity and effects on cells .
  • Research: It is used in studies related to immune response, inflammation, and hematopoiesis .
Quality and Specifications

Recombinant IL-6 is available in different grades, such as research grade and premium grade, with varying levels of biological activity . It is crucial to use the appropriate grade and concentration for specific experimental needs to ensure reproducible results.

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.