IL-10 Anti-Human antibodies are immunoglobulin-based reagents designed to bind human IL-10 with high specificity. IL-10 is a 178-amino-acid homodimeric cytokine (class-2) encoded by the IL10 gene, functioning primarily as an anti-inflammatory mediator through JAK-STAT signaling pathways . Antibodies against IL-10 enable researchers to:
Neutralize IL-10 bioactivity in cellular assays
Detect IL-10 expression in tissues (e.g., immunohistochemistry)
IL-10’s functional domains have been mapped using antibody blocking studies and synthetic peptides:
C-terminal domain (residues 152–160): A synthetic nonapeptide (IT9302) mimicking this region replicates IL-10’s ability to:
N-terminal domain: Regulates mast cell proliferation but lacks cytokine-inhibitory properties .
Anti-human IL-10 antibodies exhibit differential recognition of viral IL-10 homologs:
Viral IL-10 | Antibody Cross-Reactivity | Functional Neutralization |
---|---|---|
EBV (BCRF1) | 6/7 tested antibodies | Yes (e.g., JES3-12G8) |
CMV | 0/7 tested antibodies | No |
Table 2: Cross-reactivity profile of anti-human IL-10 antibodies with viral homologs .
Immunohistochemistry: Detects IL-10 in human tonsil tissues with hematoxylin counterstaining .
Western Blot: Identifies IL-10 at ~16–23 kDa in transfected HEK293 cell lines .
Neutralizing Pathological IL-10: Antibodies like JES3-12G8 suppress IL-10’s immunosuppressive effects in chronic infections (e.g., EBV) .
Cancer Immunotherapy: PEGylated IL-10 analogs enhance CD8+ T cell cytotoxicity (Granzyme B/Perforin) while anti-IL-10 antibodies counteract tumor-induced immunosuppression .
Inflammatory Regulation: IL-10 antibodies reverse IL-10-mediated suppression of IFN-γ and TNF-α in Burkholderia pseudomallei infections, restoring protective immunity .
Species Specificity: Murine IL-10 shows no cross-reactivity with human-targeted antibodies, underscoring epitope divergence .
Cardiovascular Impact: Anti-IL-10 strategies mitigate vascular dysfunction in IL-10-deficient murine models .
IL-10 (Interleukin 10) is a natural endogenous immunosuppressive cytokine identified in humans, mice, and other organisms . It serves as a critical regulator of immune responses, often referred to initially as CSIF (cytokine synthesis inhibitory factor) due to its ability to suppress inflammatory cytokine production . Anti-human IL-10 antibodies are essential research tools that enable detection, quantification, and neutralization of IL-10 in experimental systems, allowing researchers to investigate IL-10's role in immune regulation, disease pathogenesis, and potential therapeutic interventions.
Different monoclonal antibody clones exhibit varying efficacy across applications. For instance, clone #23738 demonstrates effectiveness in neutralization assays, where at 2 μg/mL it neutralizes >60% of the bioactivity from 5 ng/mL recombinant human IL-10 . In contrast, clone #25209 has a considerably higher potency with a Neutralization Dose (ND50) of 0.005-0.03 μg/mL in the presence of 5 ng/mL recombinant human IL-10 . In Western blotting applications, certain antibodies like clone E10 consistently produce stronger bands compared to others such as JES3-19F1 and JES3-12G8 . These differences highlight the importance of selecting the appropriate clone for specific experimental needs.
Human IL-10 (hIL-10) contains distinct functional domains that contribute to its various biological activities. Research has identified a nonapeptide (IT9302) with complete homology to a sequence in the C-terminal portion (residues 152-160) of hIL-10 that mimics several of the cytokine's activities . Another nonapeptide (IT9403) near the N-terminal region lacks cytokine synthesis inhibitory properties but regulates mast cell proliferation . These findings suggest that relatively small segments of IL-10 are responsible for specific biological functions, which has implications for antibody design and epitope targeting.
Researchers should employ multiple complementary techniques to comprehensively assess antibody specificity. Western blotting provides information about antibody recognition of denatured IL-10 protein. In this application, recombinant human IL-10 typically appears as a band at approximately 16-18 kDa under reducing conditions . ELISA testing evaluates antibody recognition of native protein conformation and can provide relative measurements of binding affinity . Most critically, functional neutralization assays should be performed to confirm that the antibody can inhibit IL-10's biological activity, such as its ability to suppress TNFα production in LPS-stimulated monocytes . These complementary approaches provide a comprehensive profile of antibody specificity and functionality.
Cross-reactivity with viral IL-10 homologs is a crucial consideration when selecting anti-human IL-10 antibodies. Many antibodies that recognize human IL-10 (hIL-10) also cross-react with Epstein-Barr virus IL-10 (ebvIL-10) but not with cytomegalovirus IL-10 (cmvIL-10) . For instance, while six out of seven tested anti-hIL-10 antibodies effectively neutralized both hIL-10 and ebvIL-10 activity, none affected cmvIL-10 function . This cross-reactivity has significant implications for research involving viral infections and could potentially impact clinical applications if these antibodies were to be developed for therapeutic use. Researchers should thoroughly test for cross-reactivity when precise specificity is required.
Binding affinity does not always correlate directly with neutralization capacity. Some antibodies that show relatively weak recognition in Western blot or ELISA can still effectively neutralize IL-10 activity . For example, antibody E10 exhibited high specificity for hIL-10 but could not fully neutralize cytokine suppression, suggesting its epitope may be distinct from receptor contact sites . Conversely, some antibodies with modest binding affinity can efficiently neutralize ebvIL-10, which has reduced affinity for the IL-10 receptor compared to hIL-10 . These observations underscore the importance of functional testing beyond simple binding assays when selecting antibodies for neutralization experiments.
For optimal Western blot detection of IL-10, PVDF membranes are recommended with antibody concentrations ranging from 2-5 μg/mL depending on the specific clone . Under reducing conditions using appropriate buffer systems (such as Immunoblot Buffer Group 1), human IL-10 typically appears as a specific band at approximately 16 kDa . For enhanced sensitivity, HRP-conjugated secondary antibodies with optimized chemiluminescent detection systems are preferred. When analyzing complex samples, appropriate positive controls (such as recombinant IL-10 or IL-10 transfected cell lysates) and negative controls should be included to validate specificity and establish detection limits.
To properly optimize neutralization assays, researchers should first establish a dose-response curve for recombinant IL-10 in their chosen biological system. For instance, using the MC/9-2 mouse mast cell proliferation model, IL-10 stimulates proliferation in a dose-dependent manner . From this baseline, researchers can introduce titrated concentrations of neutralizing antibodies to determine the concentration required for effective neutralization. For clone #23738, 2 μg/mL neutralizes >60% of the bioactivity from 5 ng/mL recombinant human IL-10 , while clone #25209 has an ND50 of approximately 0.005-0.03 μg/mL under similar conditions . Adequate pre-incubation time between antibody and IL-10 is crucial for effective neutralization, typically 30-60 minutes at room temperature.
A robust experimental design for studying IL-10-mediated immunosuppression involves stimulating a pro-inflammatory response (typically using LPS) in appropriate cells like THP-1 monocytes or peripheral blood mononuclear cells, which leads to TNFα production . When recombinant IL-10 is added to this system, TNFα levels are substantially reduced due to IL-10's cytokine synthesis inhibitory function . To evaluate anti-IL-10 antibody efficacy, researchers should add neutralizing antibodies at varying concentrations to determine their ability to restore TNFα production. Including appropriate controls is critical: an isotype control antibody to rule out non-specific effects, anti-IL-10 receptor antibodies as positive controls for neutralization, and controls for antibody cross-reactivity with viral IL-10 homologs when relevant .
Anti-IL-10 antibodies with known epitope specificities can be powerful tools for investigating functional domains of IL-10. By selecting antibodies that target different regions of IL-10, researchers can block specific functions while leaving others intact. This approach complements research that has identified distinct functional domains, such as the C-terminal nonapeptide (residues 152-160) that mimics several IL-10 activities, including inhibition of IL-8 production, induction of IL-1 receptor antagonist, and chemotactic effects on CD8+ T cells . Epitope mapping of neutralizing antibodies, combined with site-directed mutagenesis studies, can provide insights into structure-function relationships of IL-10 and potentially lead to the development of domain-specific inhibitors for therapeutic applications.
The existence of viral IL-10 homologs (particularly ebvIL-10 and cmvIL-10) has significant implications for research using anti-human IL-10 antibodies. Many antibodies cross-react between human IL-10 and ebvIL-10, but not with cmvIL-10 . This cross-reactivity pattern provides a unique opportunity to study how viruses have evolved to mimic host immunoregulatory molecules. Researchers can exploit differential reactivity patterns to distinguish viral from host cytokines in infection models. Additionally, understanding the structural basis for cross-reactivity (or lack thereof) can provide insights into IL-10 evolution and structure-function relationships. For clinical applications, awareness of potential cross-reactivity with viral homologs is essential when developing therapeutic antibodies, as noted with the humanized JES3-9D7 antibody that has potential for clinical use .
Anti-IL-10 antibodies provide valuable tools for dissecting IL-10 signaling networks and their cross-talk with other pathways. In experimental designs, researchers can use neutralizing antibodies to block IL-10 activity and examine downstream effects on gene expression and cellular functions. For example, studies have shown that IL-10 blockade partially abrogates S100A8 and S100A9 mRNA expression increases in models of endotoxin tolerance . Additionally, by combining IL-10 neutralization with inhibitors of other pathways, researchers can investigate signaling cross-talk. IL-10 has been shown to interact with mTORC1 signaling in the regulation of late-phase TNF responses in human macrophages . Careful experimental design with appropriate controls and time-course analyses can reveal how IL-10 signaling integrates with other inflammatory and immune regulatory networks.
Several technical challenges can arise when detecting IL-10 by Western blot. IL-10 exists as a homodimer in its native state but runs as a monomer (approximately 16-18 kDa) under reducing conditions . A common issue is detection sensitivity, as IL-10 is often present at low concentrations in biological samples. This can be addressed by using high-affinity antibodies (clone E10 consistently gives darker bands than other clones like JES3-19F1) and optimized detection systems. Sample preparation is also critical; for recombinant IL-10, researchers observe differences in band intensity even when equal amounts are loaded, as evidenced by Ponceau staining . For biological samples, concentration steps may be necessary. Non-specific bands can be minimized by optimizing blocking conditions (PBS-Tween with 5% milk is commonly used) and carefully titrating primary and secondary antibody concentrations.
A comprehensive validation strategy employs multiple complementary techniques. For Western blot validation, comparing reactivity between mock-transfected and IL-10-transfected cell lines provides a stringent specificity test . For ELISA, comparing signal with recombinant IL-10 versus irrelevant proteins (like IFNγ) can confirm specificity . Most importantly, functional validation through neutralization assays confirms that the antibody blocks IL-10's biological activity. The gold standard approach uses multiple applications: if an antibody specifically recognizes IL-10 in Western blot, ELISA, and effectively neutralizes IL-10 activity in functional assays, its specificity is strongly supported. Additionally, knockout or knockdown controls, though not mentioned in the provided search results, would provide definitive validation.
When designing experiments to investigate IL-10's immunoregulatory functions, researchers must consider several factors. Cell type specificity is critical as IL-10 has differential effects on various immune cells - suppressing CD4+ T cell responses to IL-8 while acting as a chemotactic factor for CD8+ T cells . Timing is also crucial since IL-10 can have different effects during early versus late immune responses, as evidenced by its role in mTORC1-regulated late-phase TNF responses . Concentration dependence must be established through dose-response curves, as biological effects may vary at different IL-10 concentrations. When using neutralizing antibodies, titration is essential to ensure complete neutralization without non-specific effects. Finally, appropriate readouts should be selected based on the specific IL-10 function being studied, such as cytokine production, cell proliferation, chemotaxis, or gene expression changes.
Emerging antibody engineering technologies will likely enhance the utility of anti-IL-10 research tools in several ways. Domain-specific antibodies targeting discrete functional regions of IL-10 could enable selective blockade of specific IL-10 activities while preserving others, based on the identification of functionally distinct domains like the C-terminal nonapeptide (IT9302) . Bispecific antibodies could simultaneously target IL-10 and other immune regulatory molecules to investigate pathway crosstalk. Humanized versions of existing antibodies, such as the humanized JES3-12G8 , will facilitate translation from preclinical to clinical applications. Additionally, antibody fragments (Fab, scFv) with improved tissue penetration could enhance in vivo applications, while recombinant antibodies with site-specific modifications could improve consistency and reduce batch-to-batch variation that currently necessitates laboratory-specific antibody titration .
Systems biology approaches can leverage anti-IL-10 antibodies to comprehensively map IL-10's role in immune networks. High-dimensional analyses combining IL-10 neutralization with single-cell technologies (scRNA-seq, CyTOF) can reveal cell type-specific responses to IL-10 across the immune system. Temporal studies using precisely timed IL-10 neutralization can elucidate its role in different phases of immune responses, building on observations of its involvement in late-phase TNF regulation . Network analyses integrating transcriptomic, proteomic, and functional data can map signaling cascades downstream of IL-10, including its intersection with mTORC1 signaling . Computational modeling incorporating data from IL-10 neutralization experiments can predict system-wide consequences of IL-10 modulation and identify potential compensatory mechanisms. These integrative approaches will provide a more complete understanding of IL-10's complex regulatory functions across different physiological and pathological contexts.
Antibody Clone | Western Blot Detection | ELISA Recognition | Neutralization Capacity | Cross-reactivity with ebvIL-10 | Cross-reactivity with cmvIL-10 |
---|---|---|---|---|---|
23738 | Positive | Positive | >60% at 2 μg/mL | Yes | No |
25209 | Positive | Not specified | ND50: 0.005-0.03 μg/mL | Not specified | Not specified |
E10 | Strong positive | Positive | Partial | No in blot/ELISA, modest in function | No |
JES3-19F1 | Weak positive | Not specified | Near complete | Yes | No |
JES3-12G8 | Weak positive | Not specified | Near complete | Yes | No |
3C12C12 | Not specified | Positive | Not specified | Not specified | Not specified |
Interleukin-10 (IL-10) is a crucial anti-inflammatory cytokine that plays a significant role in regulating immune responses. It is also known as human cytokine synthesis inhibitory factor (CSIF). In humans, IL-10 is encoded by the IL10 gene located on chromosome 1 . This cytokine is primarily produced by monocytes and, to a lesser extent, by lymphocytes, including type-II T helper cells (TH2), mast cells, CD4+ CD25+ Foxp3+ regulatory T cells, and certain subsets of activated T cells and B cells .
IL-10 is a homodimer, with each subunit consisting of 178 amino acids . It belongs to the class-2 cytokine family, which includes other cytokines such as IL-19, IL-20, IL-22, IL-24, and various interferons . The IL-10 protein signals through a receptor complex composed of two IL-10 receptor-1 and two IL-10 receptor-2 proteins . This receptor complex activates the STAT3 signaling pathway via the phosphorylation of the cytoplasmic tails of IL-10 receptor 1 and IL-10 receptor 2 by JAK1 and Tyk2, respectively .
The expression of IL-10 is tightly regulated at both the transcriptional and post-transcriptional levels. It is minimally expressed in unstimulated tissues and requires triggering by commensal or pathogenic flora . IL-10 expression involves extensive locus remodeling in monocytes upon stimulation of TLR or Fc receptor pathways . The induction of IL-10 involves ERK1/2, p38, and NF-κB signaling, with transcriptional activation via promoter binding of transcription factors NF-κB and AP-1 . Additionally, IL-10 expression is regulated at the post-transcriptional level, involving control of mRNA stability via AU-rich elements and microRNAs such as let-7 and miR-106 .
While IL-10 is a well-studied cytokine in both mice and humans, there are notable differences in its function and regulation between the two species. In mice, IL-10 is rigidly associated with TH2 clones, whereas in humans, both TH1 and TH2 clones can secrete IL-10 . This difference highlights the complexity of IL-10’s role in immune regulation and its varying effects across species.
IL-10 has significant therapeutic potential due to its anti-inflammatory properties. It has been explored in various clinical settings to modulate immune responses and treat inflammatory diseases . For instance, IL-10 has been shown to inhibit the activation of human myeloid cells in response to TLR activation . Additionally, IL-10 suppression has been found to enhance T-cell antitumor immunity, making it a potential target for cancer immunotherapy .