IL-6 exerts its effects through three distinct pathways :
Classic Signaling: Binding to membrane-bound IL-6 receptor (IL-6R) and gp130 co-receptor.
Trans-Signaling: Interaction with soluble IL-6R (sIL-6R) and gp130, driving chronic inflammation.
Cluster Signaling: Membrane-bound IL-6:IL-6R complexes activate gp130 on adjacent cells.
Activation of these pathways triggers downstream JAK/STAT3, MAPK, and PI3K/AKT cascades, regulating immune responses, angiogenesis, and metabolism .
Induces acute-phase proteins (e.g., C-reactive protein) during infection or injury .
Promotes differentiation of B cells into antibody-producing plasmablasts and Th17/Tfh T-cell subsets .
Elevated levels correlate with autoimmune diseases (e.g., rheumatoid arthritis), cancers, and atherosclerosis .
Acts as a myokine during exercise, enhancing lipolysis and insulin sensitivity .
Supports liver regeneration and epithelial repair via IL6ST-YAP-NOTCH signaling .
Recombinant IL-6 is widely used in:
Drug Development: Screening IL-6 inhibitors (e.g., Tocilizumab) using HEK-Blue™ IL-6 reporter cells .
Biomarker Studies: Quantifying IL-6 in ELISA assays with sensitivity down to 15 pg/mL .
Mechanistic Studies: Investigating mutations (e.g., R167A, E171A) that enhance STAT3 activation or receptor binding .
A phase I trial evaluated subcutaneous IL-6 (3–30 µg/kg) in cancer patients :
Dose (µg/kg) | Key Observations |
---|---|
3–10 | Tolerable; increased CRP, fibrinogen, and platelets. |
30 | Dose-limiting hepatotoxicity/cardiac arrhythmia; no antitumor responses observed. |
Computational and experimental studies identified critical residues for IL-6:IL-6R binding :
R167A: Increased affinity for IL-6R:gp130 complex, enhancing STAT3 phosphorylation by 2-fold.
E171A: Reduced IL-6R binding but stronger gp130 interaction, boosting biological activity.
R181A/V: Abolished IL-6R binding but retained gp130 affinity, suggesting alternative signaling routes.
Mutant | IL-6R Binding (K<sub>D</sub>) | gp130 Complex Binding (K<sub>D</sub>) | STAT3 Activation |
---|---|---|---|
Wild-Type | 3.2 nM | 1.8 nM | Baseline |
R167A | 2.9 nM | 0.9 nM | ↑ 200% |
E171A | 5.1 nM | 1.2 nM | ↑ 150% |
IL-6 blockade (e.g., Tocilizumab) is FDA-approved for rheumatoid arthritis and juvenile idiopathic arthritis . Emerging research explores IL-6 inhibitors for obesity-linked inflammation and cancer metastasis .
Recombinant Human IL6 is a laboratory-produced version of the naturally occurring cytokine, designed to mimic the biological activities of endogenous IL6. The recombinant protein typically contains the 30-212aa sequence of human IL6 with an N-terminal 6xHis-tag to facilitate purification. In terms of biological activity, properly produced recombinant IL6 maintains the functional properties of the endogenous protein while offering advantages in purity, consistency, and availability for research purposes. High-quality recombinant IL6 preparations typically demonstrate >95% purity and endotoxin levels below 1.0 EU/μg, making them suitable for in vitro and in vivo research applications .
IL6 serves as a pleiomorphic cytokine with diverse functions in normal physiology. It is primarily produced by immune cells such as macrophages, T cells, and B cells, as well as non-immune cells like fibroblasts and endothelial cells. IL6 plays crucial roles in the acute-phase response as part of the body's defense mechanism against infections and injuries, during which IL6 levels can rise significantly . Beyond inflammation, IL6 demonstrates anti-inflammatory, pro-resolution, and regenerative properties important for pathogen clearance and tissue repair. In the central nervous system, IL6 regulates energy and glucose homeostasis and mediates crosstalk between insulin-sensitive tissues, intestinal L cells, and pancreatic islets to adapt to changes in insulin demand . IL6 also serves as a myokine during exercise and plays a protective role during liver injury by maintaining tissue regeneration capabilities .
IL6 signaling occurs through two primary mechanisms: classical signaling and trans-signaling. For signal transduction to occur, IL6 must first bind to the IL6 receptor (IL6R), after which the resultant complex associates with glycoprotein 130 (gp130) . In classical signaling, IL6 binds to membrane-bound IL6R (mbIL6R), which is expressed on only a few cell types, most notably hepatocytes and some leukocytes including macrophages and T-cell subsets. This binding forms a complex that then associates with gp130, which is expressed by all cell types, leading to intracellular signaling .
In trans-signaling, IL6 binds to a soluble form of IL6R (sIL6R), which is generated through cleavage of membrane-bound IL6R by disintegrin and metalloprotease domain-containing protein 17 (ADAM-17) during inflammation. The IL6-sIL6R complex can then bind to gp130 on cells that do not express IL6R, allowing IL6 to affect a broader range of cell types under inflammatory conditions . The anti-inflammatory and antibacterial activities of IL6 are predominantly mediated by classical signaling, whereas its pro-inflammatory effects typically occur through trans-signaling .
Production of high-quality recombinant human IL6 involves several critical steps. Initially, the target gene sequence (30-212aa) is amplified using specific primers and cloned into an expression plasmid that includes an N-terminal 6xHis-tag. This recombinant plasmid is then transfected into yeast cells, which are selected using appropriate antibiotics 24 hours post-transfection to isolate successfully transfected cells .
The selected cells are cultured under optimized conditions to promote protein expression, then lysed to release the IL6 protein. Purification is typically achieved using Ni-NTA affinity chromatography, which selectively binds the His-tagged IL6 protein. Following purification, quality assessment involves evaluating protein purity through SDS-PAGE and Western blotting. Endotoxin testing using the Limulus Amebocyte Lysate (LAL) assay ensures levels remain below 1.0 EU/μg, which is critical for preventing experimental artifacts. Functional validation through ELISA demonstrates binding capacity to IL6 antibodies, with typical EC50 values ranging from 35.80-41.82 ng/mL .
Researchers can assess the functional activity of recombinant IL6 through several complementary approaches:
Binding Assays: ELISA-based methods measuring binding to IL6 recombinant antibodies or IL6R provide initial confirmation of correct protein folding and receptor recognition .
Cell-Based Functional Assays: Experiments using hepatoma cell lines like Fao can assess IL6's ability to induce acute-phase protein expression. In the presence of dexamethasone, properly functioning IL6 should induce a 6-fold increase in β-fibrinogen mRNA levels .
Signaling Pathway Activation: Measuring phosphorylation of STAT3 and activation of the JAK-STAT pathway in responsive cells provides direct evidence of functional activity. This can be assessed through Western blotting or flow cytometry techniques .
Time-Course Experiments: Functional IL6 should induce β-fibrinogen mRNA production immediately after addition, reaching maximum levels between 12-18 hours in appropriate cell models .
Dose-Response Analysis: Testing multiple concentrations helps establish the EC50 (effective concentration at 50% maximal response), which should be compared with established standards .
Critical quality control parameters for recombinant IL6 include:
Parameter | Acceptable Range | Significance | Testing Method |
---|---|---|---|
Purity | >95% | Prevents interference from contaminants | SDS-PAGE, HPLC |
Endotoxin Content | <1.0 EU/μg | Prevents inflammatory artifacts | LAL test |
Biological Activity | EC50: 35-45 ng/mL | Confirms functional capacity | ELISA, cell-based assays |
Protein Concentration | Batch-dependent | Ensures accurate dosing | Bradford/BCA assay |
Aggregation State | Predominantly monomeric | Affects bioavailability | Size exclusion chromatography |
Post-translational Modifications | Consistent with human IL6 | Affects receptor binding | Mass spectrometry |
Researchers should also verify batch-to-batch consistency and storage stability to ensure experimental reproducibility. Reconstituted IL6 typically maintains activity for 1-2 weeks at 4°C and should be stored at -80°C for longer-term preservation .
Recombinant human IL6 significantly impacts gene expression in hepatocytes through activation of the JAK-STAT signaling pathway. In the rat hepatoma cell line Fao, treatment with IL6, particularly in the presence of dexamethasone (10⁻⁶ M), induces a 6-fold increase in β-fibrinogen mRNA levels. This induction begins immediately after IL6 addition and reaches maximum levels between 12-18 hours post-treatment .
IL6 also affects other acute-phase proteins, though to varying degrees. While IL6 is a potent inducer of β-fibrinogen, it demonstrates relatively weaker stimulation of α1-acid glycoprotein mRNA synthesis compared to other inflammatory mediators like IL-1β and TNFα. Conversely, IL6 (along with IL-1β and TNFα) reduces albumin mRNA concentrations to approximately 30% of control levels, reflecting its role in downregulating negative acute-phase proteins during inflammatory responses .
These differential effects on gene expression reflect IL6's complex role in orchestrating the hepatic acute-phase response, with some genes being primary IL6 targets and others responding more strongly to alternative inflammatory mediators or combinations thereof .
When using recombinant IL6 in cell culture experiments, researchers should consider several important factors:
Cell Type Selection: Remember that classical IL6 signaling only occurs in cells expressing membrane-bound IL6R (primarily hepatocytes, macrophages, and specific T-cell subsets), while trans-signaling can affect a broader range of cells through the soluble IL6R mechanism .
Dosage Determination: Establish dose-response relationships for your specific cell type and readout. For hepatocytes, concentrations sufficient to induce acute-phase protein expression typically range from 10-100 ng/mL, though this varies by experimental system .
Co-factors and Synergistic Effects: Consider the addition of dexamethasone (typically at 10⁻⁶ M), which potentiates IL6-induced β-fibrinogen mRNA expression and is an absolute requirement for stimulation of α1-acid glycoprotein mRNA .
Temporal Dynamics: Account for the time-dependent nature of IL6 responses. β-fibrinogen mRNA induction begins immediately after IL6 addition, reaching maximum levels between 12-18 hours, while other responses may show different kinetics .
Combinatorial Effects: When studying acute-phase responses, consider that IL6 often works in concert with other inflammatory mediators. While no synergistic effect occurs for β-fibrinogen mRNA induction, combinations of IL6/IL-1β, IL6/TNFα, or IL-1β/TNFα regulate α1-acid glycoprotein and albumin mRNA synergistically .
Soluble Receptor Considerations: In some experimental systems, adding exogenous soluble IL6R may be necessary to study trans-signaling mechanisms in cells lacking membrane-bound IL6R .
Researchers can differentiate between classical and trans-signaling effects of IL6 through several methodological approaches:
Cell Type Selection: Using cells that express membrane-bound IL6R (e.g., hepatocytes, macrophages) versus those that do not (e.g., endothelial cells, epithelial cells) allows for initial distinction between potential signaling mechanisms .
Selective Inhibition: Employing sgp130Fc, a fusion protein that specifically inhibits trans-signaling without affecting classical signaling, helps distinguish between these pathways. If a response is inhibited by sgp130Fc, it suggests involvement of trans-signaling .
Soluble Receptor Manipulation: Adding exogenous soluble IL6R to experimental systems can enhance trans-signaling. If cellular responses increase with sIL6R addition in cells lacking membrane IL6R, this indicates trans-signaling .
ADAM-17 Inhibition: Using inhibitors of ADAM-17 (the enzyme that cleaves membrane-bound IL6R to generate sIL6R) can reduce trans-signaling without affecting classical signaling .
Readout Selection: Certain biological responses have been associated predominantly with one pathway. Anti-inflammatory and antibacterial activities typically result from classical signaling, while pro-inflammatory effects generally occur through trans-signaling .
Genetic Approaches: Utilizing cells from IL6R knockout models reconstituted with either membrane-bound (non-cleavable) or soluble IL6R allows for clean separation of the two pathways .
The rat hepatoma cell line Fao demonstrates that different acute-phase proteins respond differently to various inflammatory mediators. While IL6 strongly induces β-fibrinogen, other inflammatory mediators like IL-1β and TNFα preferentially induce α1-acid glycoprotein. This suggests that the acute-phase response involves coordinated action of multiple cytokines, each regulating specific subsets of genes .
Synergistic effects occur between inflammatory mediators for certain responses. The combinations of IL6/IL-1β, IL6/TNFα, and IL-1β/TNFα synergistically regulate α1-acid glycoprotein and albumin mRNA, even though no synergistic effect is observed for β-fibrinogen mRNA induction. This indicates pathway convergence for some target genes but not others .
IL6 signaling impacts other inflammatory pathways through its effects on T-cell differentiation. IL6 is essential for the development of T follicular helper cells and for driving naive CD4+ T cells toward the Th17 lineage, thus influencing adaptive immune responses beyond innate inflammation .
During exercise, IL6 acts as a myokine with anti-inflammatory properties, potentially through inhibition of TNFα production and stimulation of anti-inflammatory cytokines like IL-10 and IL-1 receptor antagonist. This demonstrates context-dependent interactions with other inflammatory mediators .
IL6 plays a pivotal role in exercise physiology as a myokine released from working muscle fibers, with increased production correlating with exercise duration, intensity, and muscle glycogen depletion. Researchers investigating IL6 in exercise physiology should consider the following approaches:
Temporal Sampling Protocols: IL6 is acutely released during exercise, necessitating careful timing of sample collection. Researchers should design protocols with multiple sampling points during and after exercise to capture the dynamic changes in IL6 levels .
Exercise Modality Considerations: Different exercise types (endurance vs. resistance) and intensities produce varying IL6 responses. Study designs should incorporate standardized exercise protocols with clearly defined parameters to enable meaningful comparisons between studies .
Muscle-Organ Crosstalk Analysis: IL6 facilitates communication between working muscles and other organs during exercise. Researchers can investigate this crosstalk using arteriovenous difference measurements across exercising limbs and target organs, or through tissue-specific receptor expression analysis .
Signaling Pathway Differentiation: During exercise, IL6 predominantly activates classical signaling pathways that contribute to both performance-related adaptations and anti-inflammatory benefits. Researchers should measure both IL6 and soluble IL6R levels to understand the balance between classical and trans-signaling .
Genetic Influence Assessment: The rs2228145 SNP affects the balance between membrane-bound and soluble IL6R, potentially influencing exercise responses. Genotyping study participants for this polymorphism can help explain individual variations in training adaptations and physical activity patterns .
The rs2228145 SNP (Asp358Ala) in the IL6R gene significantly impacts IL6 signaling dynamics with important implications for research methodologies and interpretation:
This polymorphism affects the balance between classical and trans-signaling by increasing ADAM-17-mediated shedding of membrane-bound IL6R. Individuals carrying the C allele (coding for Alanine) demonstrate higher circulating levels of soluble IL6R compared to those with the AA genotype .
The increased shedding of IL6R in C allele carriers disrupts the classical/trans-signaling balance, potentially enhancing trans-signaling-mediated pro-inflammatory effects while reducing classical signaling responses. Researchers should consider genotyping study participants for this polymorphism when investigating IL6-related phenotypes or treatment responses .
Recent research has identified an association between self-selected physical activity levels and rs2228145 genotype, with the AA genotype associated with greater activity levels. This suggests that altered IL6 signaling dynamics may influence exercise behaviors and adaptations, with potential implications for exercise physiology research and personalized exercise prescriptions .
When studying inflammatory conditions where IL6 plays a role, accounting for this genetic variation may help explain heterogeneity in disease manifestation or treatment response. The polymorphism has been associated with various diseases, possibly through its effects on IL6 signaling balance .
Accurate measurement and interpretation of IL6 levels in biological samples are influenced by multiple factors that researchers must consider:
Assay Selection: Different immunoassay platforms (ELISA, multiplex, chemiluminescence) may yield varying absolute values for the same sample. Researchers should maintain consistency in assay methodology throughout a study and be cautious when comparing IL6 values across studies using different assay platforms .
Sampling Timing: IL6 levels fluctuate rapidly in response to various stimuli. The time of sample collection relative to immune challenges, exercise, or circadian rhythms significantly impacts measured values. Standardized sampling protocols with carefully documented timing are essential for meaningful comparisons .
Sample Processing: Pre-analytical variables including time to processing, temperature, centrifugation parameters, and freeze-thaw cycles can affect IL6 stability and measured concentrations. Platelet activation during blood collection can release IL6, potentially causing artifactual elevation .
Concomitant Pharmacotherapy: Many medications, particularly glucocorticoids and immunomodulators, affect IL6 production or signaling. Detailed documentation of participant medication use is crucial for proper interpretation .
Biological Context: The net biological effect of IL6 depends on multiple factors beyond its absolute concentration, including:
Reference Range Considerations: IL6 reference ranges vary by population, age, sex, and assay methodology. Study-specific reference ranges may be necessary for accurate interpretation .
When studying IL6 signaling, researchers should be aware of several common experimental design pitfalls:
Oversimplification of Signaling Pathways: Failing to distinguish between classical and trans-signaling can lead to misinterpretation of results. Experimental designs should incorporate methods to differentiate these pathways, particularly when studying complex inflammatory responses .
Neglecting Soluble Receptor Dynamics: Many studies focus solely on IL6 concentrations without measuring corresponding soluble IL6R levels. Since sIL6R is critical for trans-signaling and often exists in excess of IL6, this oversight can limit mechanistic insights .
Inappropriate Cell Model Selection: Using cell types that lack membrane-bound IL6R to study classical signaling, or failing to account for endogenous sIL6R production in culture systems, can yield misleading results .
Inadequate Temporal Sampling: IL6-induced responses follow distinct time courses, with some effects (like β-fibrinogen induction) occurring immediately and others (like α1-acid glycoprotein induction) showing lag phases of 8+ hours. Single-timepoint measurements may miss critical dynamics .
Disregarding Synergistic Effects: IL6 often works in concert with other inflammatory mediators like IL-1β and TNFα. Studying IL6 in isolation may fail to capture physiologically relevant synergistic effects that occur in vivo .
Inadequate Consideration of Genetic Variants: Polymorphisms like rs2228145 significantly impact IL6 signaling dynamics. Failure to account for genetic variation can increase unexplained variance and reduce experimental power .
Overlooking Post-translational Modifications: Variations in glycosylation patterns between recombinant and endogenous IL6, or between different recombinant preparations, can affect bioactivity and receptor interactions .
Researchers seeking to develop targeted approaches to modulate specific IL6 signaling pathways can employ several strategic methods:
Pathway-Selective Inhibitors: To specifically target trans-signaling while preserving beneficial classical signaling, researchers can use sgp130Fc, a fusion protein that selectively inhibits the IL6-sIL6R complex without affecting IL6 binding to membrane-bound IL6R. This approach preserves anti-inflammatory and antibacterial activities while reducing pro-inflammatory effects .
ADAM-17 Modulation: Since ADAM-17 is the key enzyme responsible for cleaving membrane-bound IL6R to generate sIL6R, inhibitors of this protease can reduce trans-signaling while preserving classical signaling. This approach is particularly relevant in inflammatory conditions where excessive trans-signaling drives pathology .
JAK-STAT Pathway Modulation: Targeting downstream components of the IL6 signaling cascade offers another level of specificity. JAK inhibitors like tofacitinib or STAT3 inhibitors can modulate IL6 signaling outcomes while potentially affecting other cytokine pathways that converge on these signaling molecules .
Engineered IL6 Variants: Developing IL6 muteins with altered receptor binding properties can create molecules that preferentially activate either classical or trans-signaling. For example, variants with enhanced binding to membrane-bound IL6R but reduced affinity for sIL6R could promote beneficial regenerative effects while minimizing inflammatory responses .
Cell-Type Specific Approaches: Since different cell populations express varying levels of IL6R and gp130, targeted delivery systems can direct IL6-modulating agents to specific cell types. This approach helps restrict intervention effects to the most relevant cellular compartments .
Genetic Strategies: For research purposes, CRISPR/Cas9-mediated generation of cells or organisms with modified IL6 signaling components (e.g., non-cleavable IL6R variants) can provide valuable models for studying pathway-specific effects and developing targeted therapeutic approaches .
Several emerging areas of IL6 research warrant further investigation:
Tissue-Specific IL6 Functions: Beyond its well-characterized roles in inflammation and immunity, IL6 demonstrates tissue-specific functions that require deeper exploration. In the central nervous system, IL6 regulates energy and glucose homeostasis, while in the liver, it plays a protective role during injury by maintaining tissue regeneration capabilities. Further research is needed to fully characterize these tissue-specific actions and their therapeutic implications .
IL6 in Exercise Biology: While IL6 is known to function as a myokine during exercise, facilitating muscle-organ crosstalk and contributing to both performance-related adaptations and anti-inflammatory benefits, the precise mechanisms and long-term implications remain incompletely understood. Investigating how genetic variants like rs2228145 affect exercise responses and adaptation could provide insights into personalized exercise prescriptions .
Cluster Signaling Mechanisms: Beyond classical and trans-signaling, a third mechanism called "cluster signaling" has been identified, wherein membrane-bound IL6:IL6R complexes on transmitter cells activate IL6ST receptors on neighboring receiver cells. The physiological relevance and regulatory mechanisms of this signaling modality require further investigation .
IL6 in Epithelial Regeneration: Through activation of the IL6ST-YAP-NOTCH pathway, IL6 induces inflammation-induced epithelial regeneration. This regenerative capacity could have significant implications for wound healing and tissue repair, but the underlying mechanisms and therapeutic potential require additional research .
Metabolic Functions of IL6: IL6 mediates crosstalk between insulin-sensitive tissues, intestinal L cells, and pancreatic islets to adapt to changes in insulin demand, suggesting a more complex role in metabolic regulation than previously appreciated. Further research is needed to fully characterize these functions and their implications for metabolic diseases .
Advanced technologies are poised to significantly enhance our understanding of IL6 biology across multiple dimensions:
Single-Cell Technologies: Single-cell RNA sequencing and mass cytometry can reveal cell-type-specific responses to IL6 stimulation, helping to map the heterogeneity of IL6 effects across diverse cell populations. These approaches can identify previously unrecognized IL6-responsive cell types and characterize cell-specific signaling dynamics, providing a more nuanced understanding of IL6's role in complex tissues .
CRISPR/Cas9 Genome Editing: Precise genetic manipulation enables creation of modified cell lines and animal models with alterations to specific components of the IL6 signaling pathway. Examples include generating non-cleavable IL6R variants to isolate classical signaling, or introducing specific SNPs like rs2228145 to study their functional consequences in controlled genetic backgrounds .
Intravital Imaging: Real-time visualization of IL6 signaling in living organisms using fluorescent reporters can track the spatiotemporal dynamics of IL6 production, receptor engagement, and downstream signaling activation. This approach offers insights into how IL6 signals propagate through tissues during physiological processes like exercise or pathological conditions like inflammation .
Systems Biology Approaches: Integrating multi-omics data (transcriptomics, proteomics, metabolomics) with computational modeling can help decipher the complex networks through which IL6 influences cellular function. These approaches are particularly valuable for understanding how IL6 interacts with other inflammatory mediators and signaling pathways in different physiological contexts .
Structural Biology Techniques: Advanced structural methods including cryo-electron microscopy and X-ray crystallography can provide atomic-level insights into the interactions between IL6, its receptors, and signaling components. Such structural information facilitates rational design of pathway-selective modulators with enhanced specificity for therapeutic applications .
IL6 research has significant implications for therapeutic development across multiple disease areas:
Selective Pathway Modulation: Understanding the dichotomous signaling of IL6 (anti-inflammatory effects via classical signaling versus pro-inflammatory effects via trans-signaling) enables development of pathway-specific interventions. Selective inhibition of trans-signaling using agents like sgp130Fc could potentially preserve beneficial IL6 functions while blocking detrimental inflammatory processes, offering advantages over current approaches that block both pathways .
Personalized Medicine Approaches: Genetic variations like the rs2228145 SNP significantly impact IL6 signaling dynamics and potentially treatment responses. Incorporating genotyping for such polymorphisms could enable stratification of patients for clinical trials and personalized therapeutic regimens, potentially improving treatment outcomes in diseases where IL6 plays a significant role .
Metabolic Disease Applications: The recognition of IL6's roles in energy homeostasis, glucose metabolism, and cross-talk between insulin-sensitive tissues opens new avenues for metabolic disease therapeutics. Modulating specific aspects of IL6 signaling could potentially address metabolic dysregulation in conditions like type 2 diabetes or obesity .
Exercise Mimetics: Understanding how exercise-induced IL6 production contributes to beneficial metabolic and anti-inflammatory effects could inform development of "exercise mimetics" that replicate these effects pharmacologically. Such approaches might be particularly valuable for patients unable to engage in physical activity due to disability or severe disease .
Tissue Regeneration Applications: IL6's role in tissue regeneration, particularly through the IL6ST-YAP-NOTCH pathway in epithelial tissues, suggests potential applications in regenerative medicine. Targeted activation of these pathways could potentially enhance healing in conditions characterized by impaired tissue repair .
Combined Biomarker/Therapeutic Approaches: Integration of IL6 pathway biomarkers (including soluble receptor levels and genetic variants) with therapeutic interventions could enable more precise treatment selection and monitoring, potentially improving outcomes in complex inflammatory conditions like rheumatoid arthritis, cardiovascular diseases, and various cancers .