Interleukin-10, originally identified as Cytokine Synthesis Inhibitory Factor (CSIF), is an 18.5 kD protein that functions as a major immune regulatory cytokine . Recombinant human IL-10 (rhIL-10) is a laboratory-produced version of this naturally occurring protein, manufactured to maintain biological activity while ensuring consistency and purity for research and clinical applications. The protein shares over 80% sequence homology with the Epstein-Barr Virus protein BCRFI, suggesting evolutionary significance in immune regulation . As a critical immunomodulator, IL-10 acts on numerous cells within the immune system where it demonstrates profound anti-inflammatory functions, effectively limiting excessive tissue disruption caused by inflammatory processes .
The primary biological function of IL-10 is its potent anti-inflammatory activity. It achieves this effect through multiple mechanisms, including inhibition of macrophage-mediated cytokine synthesis, suppression of delayed-type hypersensitivity responses, and stimulation of TH2 cell responses, which collectively result in elevated antibody production . The net effect of IL-10 action appears to be the inhibition of proinflammatory T cell-mediated immunity, making it a critical regulator of immune homeostasis .
IL-10 primarily targets antigen-presenting cells (APCs) such as macrophages and monocytes, inhibiting their release of pro-inflammatory cytokines including granulocyte-macrophage colony-stimulating factor (GM-CSF), granulocyte colony-stimulating factor (G-CSF), IL-1 alpha, IL-1 beta, IL-6, IL-8, and tumor necrosis factor-alpha (TNF-alpha) . Additionally, IL-10 interferes with antigen presentation by reducing the expression of major histocompatibility complex class II (MHC-II) and co-stimulatory molecules, thereby inhibiting their ability to induce T cell activation .
A particularly significant aspect of IL-10 activity is its ability to control the inflammatory response of macrophages by reprogramming essential metabolic pathways, including mammalian target of rapamycin (mTOR) signaling . This metabolic reprogramming represents a fundamental mechanism by which IL-10 exerts its anti-inflammatory effects.
The mechanism of IL-10 action begins with binding to its heterotetrameric receptor complex comprising IL-10RA and IL-10RB subunits . This binding initiates a signaling cascade involving Janus kinase 1 (JAK1) and Signal Transducer and Activator of Transcription 2 (STAT2)-mediated phosphorylation of STAT3 . Upon phosphorylation, STAT3 translocates to the nucleus where it drives the expression of various anti-inflammatory mediators, establishing a coordinated anti-inflammatory program .
Contrary to the conventional understanding of IL-10 as purely immunosuppressive, recent research has revealed that IL-10 can directly activate and expand tumor-resident CD8+ T cells . This activation occurs without requiring lymph node involvement, suggesting that IL-10 can directly stimulate antitumor immune responses through CD8+ T cell activation . The discovery that IL-10 increases the frequency of interferon-gamma (IFNγ) producing CD8+ T cells in tumors represents a paradigm shift in understanding IL-10's role in cancer immunology .
Clinical research has demonstrated promising applications for recombinant human IL-10 in treating inflammatory conditions, particularly Crohn's disease. In a 24-week multicenter, prospective, randomized, double-blind, placebo-controlled trial, patients with mild to moderately active Crohn's disease received subcutaneous rhIL-10 at various dosages (1, 5, 10, or 20 μg/kg) or placebo daily for 28 consecutive days . The study found that 23.5% of patients receiving 5 μg/kg rhIL-10 experienced clinical remission and endoscopic improvement, compared to 0% in the placebo group . Interestingly, higher doses proved less effective than the 5 μg/kg dosage, suggesting a bell-shaped dose-response curve .
Table 1: Clinical Outcomes of rhIL-10 Treatment in Crohn's Disease Patients
Treatment Group | Clinical Remission Rate | Confidence Interval | Endoscopic Improvement |
---|---|---|---|
rhIL-10 (5 μg/kg) | 23.5% | 6.8%-49.9% | Yes |
Placebo | 0% | 0%-14.8% | No |
Higher doses (>5 μg/kg) | Less effective than 5 μg/kg | Not reported | Less effective |
Recent research has uncovered surprising immunostimulatory effects of IL-10 that may have significant implications for cancer treatment. Contrary to the traditional view of IL-10 as immunosuppressive, studies have shown that IL-10 can directly activate tumor-resident CD8+ T cells, leading to tumor rejection in various mouse tumor models . This effect occurs through direct stimulation of CD8+ T cells within the tumor microenvironment, without requiring T-cell trafficking from lymphoid organs .
The discovery that IL-10 treatment can circumvent the problem of restricted T cell migration into tumors represents a potentially transformative approach to immunotherapy. By reactivating tumor-resident CD8+ T cells, IL-10 may offer a novel strategy for enhancing antitumor immunity, particularly in cases where conventional approaches have failed .
Recombinant human IL-10 is typically supplied as a frozen liquid comprised of sterile-filtered aqueous buffered solution, containing glycerol and bovine serum albumin, without preservatives . Quality control measures include SDS-PAGE analysis for purity assessment and absorbance assays based on the Beers-Lambert law . Endotoxin levels are carefully monitored, with commercial preparations maintaining levels ≤0.1 ng per μg of human IL-10, as measured by chromogenic Limulus Amebocyte Lysate (LAL) assays .
These stringent quality control measures ensure that recombinant IL-10 preparations meet the requirements for research applications such as SDS-PAGE, functional studies, mass spectrometry, HPLC, and cell culture experiments .
Recombinant human IL10 is produced in E. coli using a plasmid containing the gene fragment encoding amino acids 19-178 of human IL10. The protein exhibits a purity exceeding 95%, as determined by SDS-PAGE analysis, and an endotoxin level below 1.0 EU/µg, as measured by the LAL method. This recombinant human IL10 protein has been validated for biological activity. Its ED50, determined using a cell proliferation assay with murine MC/9-2 cells, is less than 1 ng/ml, indicating a specific activity greater than 1.0x10^6 IU/mg.
Human IL10 is a pivotal anti-inflammatory cytokine that orchestrates immune responses and maintains homeostasis within the immune system. Primarily synthesized by monocytes, lymphocytes, and mast cells, IL10 is renowned for its ability to suppress the production of pro-inflammatory cytokines such as TNF-α, IL-1, and IL-6 [1][2]. This inhibitory function is essential for controlling excessive inflammatory responses that could lead to tissue damage and chronic inflammatory diseases.
IL10 exerts its effects by binding to the IL10 receptor, initiating a signaling cascade involving the JAK and STAT pathways. This activation leads to the transcription of anti-inflammatory genes and the suppression of pro-inflammatory mediators [3]. Research has demonstrated that IL10 deficiency can augment Th1 and Th17 immune responses, associated with various autoimmune conditions [4].
Furthermore, IL10 has been implicated in diverse pathological conditions, including chronic inflammatory diseases and cancer. Its role in modulating macrophage activation and promoting a regulatory T cell phenotype is crucial in conditions such as endometriosis and atherosclerosis, where inflammation is a central component [5][6]. IL10 can influence tumor microenvironments by modulating immune cell functions, potentially suppressing or promoting tumor growth depending on the specific context [7].
References:
[1] Jung, K., Lee, G., Park, C., Lee, T., Kim, J., Sung, E., et al. (2020). Mesenchymal stem cells decrease oxidative stress in the bowels of interleukin-10 knockout mice. Gut and Liver, 14(1), 100-107. https://doi.org/10.5009/gnl18438
[2] Ōnishi, A., Akimoto, T., Urabe, M., Hirahara, I., Muto, S., Ozawa, K., et al. (2015). Attenuation of methylglyoxal-induced peritoneal fibrosis: immunomodulation by interleukin-10. Laboratory Investigation, 95(12), 1353-1362. https://doi.org/10.1038/labinvest.2015.110
[3] Verma, R., Balakrishnan, L., Sharma, K., Khan, A., Advani, J., Gowda, H., et al. (2015). A network map of interleukin-10 signaling pathway. Journal of Cell Communication and Signaling, 10(1), 61-67. https://doi.org/10.1007/s12079-015-0302-x
[4] Wang, S., Gao, X., Shen, G., Wang, W., Li, J., Zhao, J., et al. (2016). Interleukin-10 deficiency impairs regulatory t cell-derived neuropilin-1 functions and promotes th1 and th17 immunity. Scientific Reports, 6(1). https://doi.org/10.1038/srep24249
[5] Idrissi, F., Fruchart, M., Bélarbi, K., Lamer, A., Dubois-Deruy, E., Lemdani, M., et al. (2022). Exploration of the core protein network under endometriosis symptomatology using a computational approach. Frontiers in Endocrinology, 13. https://doi.org/10.3389/fendo.2022.869053
[6] Xu, J., Zhang, X., Rong, S., Gao, H., Yang, W., Li, J., et al. (2018). Yirui capsules alleviate atherosclerosis by improving the lipid profile and reducing inflammation in apolipoprotein e-deficient mice. Nutrients, 10(2), 142. https://doi.org/10.3390/nu10020142
[7] Lima, M., Quintans-Júnior, L., Santana, W., Kaneto, C., Soares, M., & Villarreal, C. (2013). Anti-inflammatory effects of carvacrol: evidence for a key role of interleukin-10. European Journal of Pharmacology, 699(1-3), 112-117. https://doi.org/10.1016/j.ejphar.2012.11.040
IL10 is a major immune regulatory cytokine that exerts profound anti-inflammatory functions on numerous cells within the immune system. It limits excessive tissue disruption caused by inflammation. Mechanistically, IL10 binds to its heterotetrameric receptor comprising IL10RA and IL10RB, triggering JAK1 and STAT2-mediated phosphorylation of STAT3. Subsequently, STAT3 translocates to the nucleus where it drives the expression of anti-inflammatory mediators.
IL10 targets antigen-presenting cells (APCs) such as macrophages and monocytes, inhibiting their release of pro-inflammatory cytokines including granulocyte-macrophage colony-stimulating factor (GM-CSF), granulocyte colony-stimulating factor (G-CSF), IL-1 alpha, IL-1 beta, IL-6, IL-8, and TNF-alpha. It also interferes with antigen presentation by reducing the expression of MHC-class II and co-stimulatory molecules, thereby inhibiting their ability to induce T cell activation. Additionally, IL10 controls the inflammatory response of macrophages by reprogramming essential metabolic pathways including mTOR signaling.
Recombinant human Interleukin-10 (rhuIL-10) is a laboratory-synthesized version of the naturally occurring IL-10 cytokine, produced through genetic engineering techniques to match the structure and function of endogenous human IL-10. It is an anti-inflammatory, immunomodulatory cytokine that plays a crucial role in regulating mucosal inflammation and immune responses . The recombinant form (sometimes referred to commercially as SCH 52000 or Tenovil) is produced to enable research applications and therapeutic interventions that require pure, standardized IL-10 protein . RhuIL-10 functions by binding to its specific receptor complex and activating signaling pathways that ultimately lead to the suppression of pro-inflammatory responses in various cell types.
IL-10 production occurs across a diverse range of immune cells, though their relative contribution varies depending on context and stimulation. T helper 2 (Th2) cells represent a crucial source of antigen-specific IL-10 and exhibit epigenetic commitment to IL-10 production, ensuring that clonal progeny cells maintain high levels of IL-10 production over extended periods . Type 1 regulatory T cells (Tr1) have emerged as another significant source, with numerous animal models demonstrating their ability to permit pathogen persistence, ameliorate autoimmunity, and suppress graft or tumor rejection . Interestingly, both Th1 and Th17 cells can produce IL-10, particularly during infectious diseases, likely as a homeostatic mechanism to prevent uncontrolled T-cell activation . Human B cells also represent a potentially important source of IL-10 . Among antigen-presenting cells, macrophages typically produce higher levels of IL-10 than dendritic cells, with macrophage production often dependent on the specific stimulus, with TLR2 ligands generally being better inducers than TLR4 or TLR5 ligands .
The IL-10 receptor complex consists of two copies each of IL-10 receptor 1 (IL-10R1) and IL-10 receptor 2 (IL-10R2), forming a dimer of heterodimers that enables signal transduction following ligand binding . IL-10R1 binds IL-10 with relatively high affinity (50–200 pM), while IL-10R2 makes only a marginal contribution to ligand binding but is crucial for initiating signal transduction upon IL-10 binding . Most hematopoietic cells constitutively express low levels of IL-10R1, which can be dramatically upregulated in response to various stimuli . Non-hematopoietic cells, such as fibroblasts and epithelial cells, can also respond to stimuli by upregulating IL-10R1 . In contrast, IL-10R2 is expressed on most cell types, meaning that a diverse range of cells can potentially bind to and consume IL-10 . This widespread receptor distribution creates challenges for therapeutic administration of recombinant IL-10, as non-immune cells can divert the cytokine away from intended immunological targets, diminishing effective dosages at sites of inflammation .
When designing dose-escalation studies with rhuIL-10, researchers must consider several critical factors to ensure valid and translatable results. Based on clinical trials, a sequential-escalating-dose approach is recommended to properly evaluate both safety and efficacy parameters . In previous studies, doses ranging from 1 to 20 microg/kg have been utilized, with an important observation that the dose-response relationship is not linear—the 5 microg/kg dose demonstrated greater efficacy than higher doses of 10 and 20 microg/kg in Crohn's disease patients . Researchers should implement a treatment duration of sufficient length (typically 28 consecutive days in clinical studies) with an appropriate follow-up period (20 weeks has been used) to capture both immediate effects and delayed responses . Careful monitoring for dose-related adverse effects is essential, with special attention to hematological parameters as asymptomatic and reversible anemia and thrombocytopenia have been observed at higher doses . Additionally, researchers should consider that subcutaneous administration may result in different pharmacokinetics compared to intravenous delivery, with potential implications for bioavailability at inflammatory sites .
A comprehensive assessment of IL-10 efficacy in inflammatory disease models requires multiple complementary endpoints. Clinical indices specific to the disease being studied should be employed—for example, in Crohn's disease studies, the Crohn's Disease Activity Index (CDAI) with scores ranging from 200-350 indicating moderate activity has been used . Researchers should clearly define remission criteria, such as complete remission (CDAI < 150 with a 100-point decrease from baseline) versus clinical remission (CDAI < 150) . Endoscopic evaluation provides crucial objective evidence of mucosal healing that complements symptomatic improvement and should be included whenever possible . Biomarker analysis, including inflammatory mediators and immune cell populations, can provide mechanistic insights into treatment response . For animal models, histopathological examination of relevant tissues is essential to confirm anti-inflammatory effects at the cellular level . Safety parameters must be monitored in parallel with efficacy measures, with special attention to dose-dependent adverse events such as thrombocytopenia . Finally, investigators should include appropriate placebo controls and consider intention-to-treat analysis to account for study dropouts, as demonstrated in rigorous clinical trials of rhuIL-10 .
Accurately measuring IL-10 production in different cell populations requires specialized techniques that can detect both protein expression and identify cellular sources. Flow cytometry with intracellular cytokine staining remains the gold standard for single-cell resolution of IL-10 production, allowing simultaneous identification of cell lineage markers and cytokine expression . For researchers interested in tracking IL-10 production in vivo, reporter systems such as the IL-10/green fluorescence protein (GFP) reporter mice offer powerful tools to visualize IL-10-producing cells in real-time within tissues . Enzyme-linked immunosorbent assay (ELISA) of culture supernatants provides quantitative measurement of secreted IL-10 but lacks information on cellular sources unless performed with purified cell populations . For gene expression analysis, quantitative PCR can detect IL-10 mRNA levels, while RNA-sequencing offers comprehensive transcriptomic profiling that places IL-10 expression in broader genetic context . When studying epigenetic regulation of IL-10, techniques examining chromatin accessibility and histone modifications at the IL-10 locus can provide insights into cell-type-specific regulation patterns . Cell sorting prior to analysis is recommended when examining mixed cell populations to distinguish production sources with precision .
The contradictory outcomes observed in clinical trials of rhuIL-10 for inflammatory bowel disease stem from multiple factors that researchers should consider when designing studies. A striking observation from dose-finding studies is the non-linear dose-response relationship, where the 5 microg/kg dose demonstrated superior efficacy compared to both lower (1 microg/kg) and higher doses (10 and 20 microg/kg) . This counterintuitive finding suggests that IL-10 may exhibit a narrow therapeutic window, with potential inhibitory mechanisms activated at higher concentrations . Meta-analysis of multiple trials shows significant heterogeneity in response rates, with an I² statistic of 40% for complete remission outcomes, indicating substantial variability between studies . Patient selection criteria may contribute to these discrepancies, as trials typically enrolled therapy-refractory patients with established disease rather than treatment-naïve individuals . The route of administration presents another variable, as subcutaneous delivery used in many trials results in systemic distribution with potential cytokine diversion to non-target tissues expressing IL-10 receptors . Furthermore, the short treatment duration (typically 28 days) may be insufficient to achieve maximal therapeutic effect in a chronic condition like Crohn's disease . The statistical power of many studies was limited by small sample sizes, particularly in subgroup analyses, resulting in wide confidence intervals (e.g., 6.8%-49.9% for clinical remission in the 5 microg/kg group) . Safety concerns, particularly higher withdrawal rates due to adverse events in IL-10-treated patients (RR=13.50; 95% CI 3.89 to 46.79), have also complicated the risk-benefit assessment of this therapeutic approach .
The clinical trials of rhuIL-10 in Crohn's disease provide valuable lessons for translational researchers designing future cytokine-based therapies. A key finding is the bell-shaped dose-response curve, where the intermediate dose of 5 microg/kg demonstrated superior efficacy (23.5% clinical remission rate) compared to both lower and higher doses, suggesting complex dose-dependent mechanisms that must be carefully optimized . The observation that higher doses (10 and 20 microg/kg) were less effective challenges the conventional "more is better" approach often applied to biologics development . Safety data revealed dose-related, mild-to-moderate adverse effects that were generally reversible, with asymptomatic anemia and thrombocytopenia observed primarily at higher doses, establishing the importance of comprehensive hematological monitoring during IL-10 therapy . Meta-analysis of multiple trials showed no statistically significant benefit for IL-10 versus placebo in inducing complete remission (RR=1.43; 95% CI 0.62 to 3.29) or clinical remission (RR=1.29; 95% CI 0.79 to 2.11), highlighting the challenges of translating promising preclinical results to clinical benefit . The significantly higher withdrawal rate due to adverse events in IL-10-treated patients (RR=13.50; 95% CI 3.89 to 46.79) underscores the importance of tolerability in chronic therapy . Route of administration emerged as a crucial consideration, as subcutaneous delivery may result in cytokine diversion to non-target tissues expressing IL-10 receptors, potentially diminishing effective concentrations at inflammatory sites . These findings collectively suggest that future cytokine therapies may benefit from targeted delivery systems, combination approaches, or modified cytokines with improved pharmacokinetic profiles.
Enhancing IL-10's therapeutic potential requires innovative approaches that address the limitations identified in previous clinical studies. Based on the observation that subcutaneous administration may result in cytokine diversion to non-target tissues, researchers should explore targeted delivery systems that concentrate IL-10 at sites of inflammation while minimizing systemic exposure . The development of IL-10 variants with modified receptor binding properties could potentially enhance specificity for immune cells while reducing interaction with non-immune tissues expressing IL-10 receptors . Combination therapies represent another promising avenue, with preclinical studies suggesting that antibodies to IL-10 used alongside chemotherapy may be effective in treating certain diseases, though this approach requires careful evaluation in inflammatory conditions . Cell-based delivery systems, such as engineered regulatory T cells or mesenchymal stem cells programmed to produce IL-10 at inflammatory sites, could provide more sustained and localized cytokine delivery . Understanding the epigenetic and transcriptional regulation of endogenous IL-10 production could lead to small molecule approaches that enhance IL-10 expression in specific cell populations rather than administering the recombinant protein . The complex dose-response relationship observed in clinical trials (with 5 microg/kg showing better efficacy than higher doses) suggests that precisely controlled dosing regimens, possibly with adaptive dosing based on biomarker response, may optimize therapeutic outcomes . Finally, patient stratification approaches that identify individuals most likely to respond to IL-10 therapy could significantly improve success rates in future clinical trials.
Research on recombinant human IL-10 has provided valuable insights that extend to the broader field of anti-inflammatory cytokine therapeutics. The complex dose-response relationship observed with IL-10, where intermediate doses showed greater efficacy than higher doses, challenges the linear dose-escalation paradigm often applied to biological therapeutics and suggests similar considerations may be relevant for other immunomodulatory cytokines . The finding that IL-10 signaling exhibits cell type-specific effects—suppressing function in most immune cells while enhancing activity in B cells and NK cells—highlights the importance of comprehensive cellular profiling when evaluating novel cytokine therapies . The observation that IL-10 can inhibit various inflammatory pathways through multiple mechanisms, including NF-κB inhibition, MAPK suppression, and PI3K/AKT activation, demonstrates how anti-inflammatory cytokines can exert pleiotropic effects that may vary depending on cellular context . The challenges encountered with systemic administration of IL-10, where widespread receptor expression led to cytokine diversion away from target tissues, has prompted development of more sophisticated delivery approaches that could benefit other cytokine therapies . Epigenetic studies of IL-10 expression have revealed how T cell subsets maintain stable cytokine production profiles, providing a framework for understanding transcriptional regulation of other immunomodulatory cytokines . Finally, the recognition that IL-10 plays complex roles in infectious diseases, autoimmunity, and cancer has underscored the context-dependent nature of cytokine function and the need for disease-specific therapeutic strategies when targeting inflammatory pathways .
Ensuring consistent quality of recombinant IL-10 is crucial for research reliability and reproducibility. Researchers should verify protein purity through techniques such as SDS-PAGE and high-performance liquid chromatography (HPLC), with expectations of >95% purity for most research applications . Bioactivity testing using functional assays is essential, as protein concentration alone does not guarantee biological activity—typical assays include measuring suppression of pro-inflammatory cytokine production by lipopolysaccharide-stimulated monocytes or macrophages . Endotoxin contamination can significantly confound experimental results by independently activating immune pathways, so researchers should confirm endotoxin levels below 0.1 EU/μg protein using Limulus Amebocyte Lysate (LAL) assays . Stability assessment under various storage conditions should be performed to establish appropriate handling guidelines, as protein degradation can impact both concentration and activity . Batch-to-batch consistency should be verified through standardized quality control testing, particularly when transitioning between lots during long-term studies . Clinical trial data showed no antibody formation against IL-10 after 4 weeks of treatment, suggesting good immunological tolerance, but researchers should still monitor for potential immunogenicity in extended studies . Finally, researchers should confirm the absence of rhuIL-10 serum accumulation during multiple-dose administration protocols, as was demonstrated in clinical studies with daily subcutaneous dosing over 28 days .
Interpreting contradictory findings between IL-10 knockout and supplementation studies requires careful consideration of several methodological factors. Developmental compensation in knockout models represents a significant confounder—lifelong absence of IL-10 may trigger adaptive changes in related cytokine pathways that modify phenotypic outcomes compared to acute IL-10 blockade or supplementation . The timing of IL-10 administration is critical, as its effects may differ substantially depending on the phase of the immune response (initiation, effector, or resolution phases) . Local versus systemic IL-10 effects should be distinguished, as knockout models eliminate IL-10 throughout the body, while supplementation studies often deliver IL-10 systemically, potentially missing critical tissue-specific effects . Dose-response relationships should be carefully examined, as clinical data demonstrate non-linear effects where intermediate doses (5 microg/kg) showed greater efficacy than both lower and higher doses . Cell-specific IL-10 production and responsiveness vary widely, so the absence of IL-10 may impact different cell populations to varying degrees that cannot be fully recapitulated by systemic supplementation . Compensatory changes in IL-10 receptor expression may occur in knockout models, potentially altering sensitivity to exogenous IL-10 in supplementation studies . Finally, context dependency is crucial—IL-10's effects vary dramatically depending on the inflammatory stimulus, tissue microenvironment, and concurrent immune activation, explaining why identical IL-10 manipulations may yield different outcomes in different disease models .
Optimizing experimental conditions for studying IL-10's effects on specific immune cell subsets requires careful attention to multiple parameters. Cell isolation techniques should preserve physiological receptor expression and signaling capacity—enzymatic digestion methods can cleave surface receptors, potentially altering IL-10 responsiveness, so mechanical dissociation is preferred when possible . For in vitro studies, serum selection is critical as serum components can influence baseline activation states and may contain bovine IL-10 with partial cross-reactivity to human IL-10 receptors—researchers should consider using defined serum-free media for the most controlled conditions . The timing of IL-10 addition to cultures significantly impacts outcomes, as pre-treatment generally enhances anti-inflammatory effects compared to addition after inflammatory stimulus . Researchers should implement dose-ranging experiments that include both low (1 microg/kg) and high (up to 20 microg/kg) concentrations to capture potential biphasic responses observed in clinical trials . The choice of activation stimuli is crucial—TLR ligand selection can dramatically impact IL-10 responsiveness, with TLR2 ligands generally being better inducers of IL-10 than TLR4 or TLR5 ligands in certain cell types . For studying epigenetic regulation of IL-10 production, extended culture periods may be necessary to allow stable epigenetic modifications to emerge, particularly when examining T cell subsets with epigenetic commitment to IL-10 production . Co-culture systems incorporating multiple cell types can provide more physiologically relevant data than isolated cell cultures, particularly when studying the regulatory effects of IL-10 on antigen presentation and T cell activation . Finally, researchers should confirm IL-10 receptor expression on target cells prior to functional studies, as receptor levels vary substantially between cell types and can be dramatically upregulated by various stimuli .