TLR10 Antibody is a monoclonal antibody designed to target Toll-like receptor 10 (TLR10), a unique anti-inflammatory pattern-recognition receptor. Unlike other TLRs, TLR10 suppresses immune responses by inhibiting proinflammatory cytokine production and modulating signaling pathways . The antibody is used to study TLR10’s role in innate immunity, immune tolerance, and its potential therapeutic applications in inflammatory diseases .
Blocking TLR10: To study its suppressive effects on immune responses .
Engaging TLR10: To activate downstream anti-inflammatory pathways, such as inducing IL-1 receptor antagonist (IL-1Ra) .
Diagnostic/Therapeutic Target: Exploring TLR10 modulation in autoimmune diseases, cancer, and viral infections (e.g., SARS-CoV-2) .
TLR10 Antibody-mediated blocking enhances proinflammatory cytokine production (e.g., IL-1β, TNF-α) in human peripheral blood mononuclear cells (PBMCs) stimulated with TLR2 ligands like Pam3CSK4 . For example:
| Condition | IL-1β (pg/mL) | TNF-α (pg/mL) | IL-6 (pg/mL) |
|---|---|---|---|
| TLR10 Antibody Blocked | 2,065 ± 106 | 1,200 ± 50 | 1,500 ± 70 |
| Control (IgG) | 1,043 ± 51 | 800 ± 30 | 1,000 ± 40 |
| Data from PNAS study |
Mechanism: TLR10 forms heterodimers with TLR2 but fails to activate NF-κB or MAP kinase pathways, instead suppressing these signals .
Engagement of TLR10 on B-cells via antibody suppresses proliferation, cytokine secretion (e.g., MIP-1β), and activation markers . This B-cell intrinsic suppression impacts both T-dependent and T-independent immune responses .
Antigen-Specific Suppression: TLR10 transgenic mice show reduced antibody responses to antigens like ovalbumin .
Cytokine Downregulation: TLR10 engagement inhibits IL-6, IFN-β, and TNF-α production in B-cells .
TLR10 Antibody modulates immune responses through distinct pathways:
TLR10 overexpression in lung epithelial cells (A549) reduces proinflammatory cytokines (TNFα, IL8) and chemokines (CXCL10) during immunostimulation with viral proteins . This suggests TLR10 may mitigate cytokine storms in severe COVID-19.
| Cell Type | TLR10 Status | TNFα (pg/mL) | IL8 (pg/mL) |
|---|---|---|---|
| A549 (Control) | Wild-type | 120 ± 15 | 80 ± 10 |
| A549 (TLR10 OE) | Overexpressed | 60 ± 8 | 40 ± 5 |
| Data from Frontiers study |
TLR10’s anti-inflammatory properties make it a candidate for treating conditions like rheumatoid arthritis or lupus. Polymorphisms in TLR10 correlate with increased TLR2-mediated cytokine production, suggesting therapeutic targeting of TLR10 could reduce inflammation .
TLR10 functions as an inhibitory pattern-recognition receptor with anti-inflammatory properties, distinguishing it from other members of the Toll-like receptor family. Unlike other TLRs that primarily initiate pro-inflammatory responses, TLR10 has modulatory effects that dampen TLR2-mediated immune responses. Research has demonstrated that TLR10 acts through multiple mechanisms: it competes for ligands, forms inhibitory heterodimers with TLR2, and induces production of the anti-inflammatory cytokine IL-1Ra through PI3K/Akt-mediated pathways. When challenged with TLR2 ligands such as pam3CSK4 (Pam3Cys), human TLR10 transgenic mice exhibit significantly reduced inflammatory cytokine production compared to wild-type mice, confirming TLR10's inhibitory role in vivo .
TLR10 stands as the only inhibitory receptor within the TLR family, while all other TLRs predominantly trigger pro-inflammatory responses. This unique characteristic makes TLR10 particularly interesting for immunomodulatory research. Studies have shown that blocking TLR10 with specific antibodies significantly upregulates TLR2-mediated production of pro-inflammatory cytokines including IL-1β, IL-6, and TNF-α. Additionally, individuals carrying loss-of-function polymorphisms in the TLR10 gene demonstrate enhanced cytokine production in response to TLR2 ligands, further confirming TLR10's suppressive function. TLR10 is also distinctive in its ability to specifically induce IL-1Ra production without triggering pro-inflammatory cytokines like IL-1β, IL-6, or TNF-α .
TLR10 expression has been definitively demonstrated in B-cells, where it plays a significant role in modulating adaptive immune responses. When designing experiments involving TLR10, researchers should consider that its expression and function appear to be particularly important within the B-cell lineage. In primary human B-cells, TLR10 engagement suppresses proliferation induced by various stimuli, including pairwise engagement of BCR, TLR7, and CD40 receptor, as well as Staphylococcus aureus Cowan strain (SAC). This cell type-specific expression pattern necessitates careful selection of appropriate cell models for TLR10 studies. Additionally, TLR10 has been identified in trophoblasts during gestation, where it has been associated with apoptosis through caspase-3 activation. When designing experiments, researchers should account for these tissue-specific expression patterns and functional variations .
Generating specific TLR10 antibodies requires careful consideration of antigen design and validation protocols. Based on published research, an effective approach involves using the extracellular domain of TLR10 (amino acids 20-474) as an immunogen. After immunization, hybridoma clones should undergo rigorous counter-screening against closely related TLRs (particularly TLR1) to eliminate cross-reactive antibodies. Validation should include flow cytometry analysis of TLR10-transfected cells versus empty vector controls to confirm specificity. For example, the development of monoclonal antibodies 3C10C5 and 5C2C5 followed this methodology, resulting in highly specific reagents for TLR10 research. Additionally, functional validation through B-cell stimulation assays can confirm the antibody's biological activity, as demonstrated by the suppressive effects observed when these antibodies engage TLR10 on primary human B-cells .
When designing experiments to investigate TLR10's inhibitory functions, researchers should employ both gain-of-function and loss-of-function approaches. For antibody-mediated engagement studies, a protocol involving pre-incubation of cells with anti-TLR10 antibody (e.g., 3C10C5 at 20μg/mL) followed by cross-linking with a secondary antibody (anti-Mouse IgG1 F(ab)'2 fragment at 20μg/mL) for 30 minutes before stimulation has proven effective. This approach prevents potential interference from IgG FcR-mediated inhibition. Appropriate controls include isotype-matched antibodies at equivalent concentrations. For stimulation, researchers should consider using multiple activators to assess the breadth of TLR10's inhibitory effects, including anti-IgM (20μg/mL), anti-CD40 (0.1μg/mL), TLR7/8 agonist R848 (100ng/mL), and TLR9 agonist CpG (2μg/mL). This comprehensive stimulation panel allows assessment of TLR10's effects on both T-dependent and T-independent B-cell activation pathways .
Multiple complementary techniques should be employed to comprehensively evaluate TLR10-mediated suppression of B-cell responses. Proliferation assays using CFSE-labeled B-cells provide quantitative assessment of division rates in the presence of TLR10 antibodies versus controls. Cytokine/chemokine production measurements (particularly MIP-1β/CCL4) via ELISA offer insights into functional outcomes of TLR10 engagement. RNA analysis through PCR arrays focused on B-cell activation genes can reveal broader transcriptional effects, as demonstrated in studies showing widespread suppression of activation-induced genes following TLR10 engagement. Signal transduction analysis should examine phosphorylation states of key mediators in B-cell activation pathways. For in vivo assessment, researchers can utilize transgenic mice expressing human TLR10, challenging them with either T-dependent or T-independent antigens and measuring antibody responses. Adoptive transfer experiments with TLR10-expressing B-cells into B-cell-deficient mice provide definitive evidence of B-cell intrinsic effects .
TLR10 genetic variations significantly impact immune responses, with loss-of-function polymorphisms leading to enhanced pro-inflammatory cytokine production in response to TLR2 ligands. To study these impacts, researchers should implement a multi-faceted approach. First, genotyping of study populations should identify relevant SNPs in the TLR10 gene. PBMCs isolated from individuals with different TLR10 genotypes can then be challenged with TLR2 ligands like Pam3Cys or viable microorganisms such as Borrelia burgdorferi. Researchers should measure both pro-inflammatory cytokines (IL-1β, IL-6, TNF-α) and anti-inflammatory mediators (IL-1Ra) to comprehensively assess the impact of polymorphisms. Data analysis should consider gene-dose effects, as research has demonstrated that cytokine production correlates with the number of dysfunctional TLR10 alleles carried by individuals. To establish causality, complementation studies using exogenous expression of wild-type TLR10 in cells from individuals with polymorphisms can determine whether normal TLR10 function restores appropriate immune regulation .
Studying TLR10 presents unique challenges since conventional mouse models lack functional TLR10 due to sequence gaps and retroviral insertions in the gene. To overcome this limitation, several approaches have proven effective. Human TLR10 transgenic mice represent a valuable tool, created by inserting the human TLR10 open reading frame into the Rosa26 locus using recombination-mediated cassette exchange (RMCE). These transgenic mice should be backcrossed to C57BL/6 background for at least 10 generations to ensure genetic homogeneity. When working with these models, researchers should confirm transgene expression using PCR with specific primers (e.g., TLR10 cond-forward: 5′-GACAGCAGAGGGTGATGCTC and TLR10 cond-reverse: 5′-CTTCCTCACAGATAGGCATGG). Functional assessment can include challenging mice with TLR2 ligands like Pam3Cys (50μg intraperitoneally) and measuring cytokine responses after approximately 4 hours. Ex vivo studies with splenocytes or peritoneal macrophages from these transgenic mice provide additional insights into cell-specific responses. For human-focused research, primary human B-cells represent the most physiologically relevant model, though humanized mouse models could potentially bridge the gap between in vitro studies and human applications .
Distinguishing direct from indirect TLR10-mediated effects requires sophisticated experimental approaches. Researchers should implement cotransfection studies in human cell lines (such as HEK293 cells) expressing TLR10 alongside other TLRs to assess direct competitive interactions or heterodimer formation. These studies can be complemented with co-immunoprecipitation experiments to physically demonstrate protein-protein interactions. For signaling studies, researchers should examine the kinetics of activation, with early timepoints (5-15 minutes) more likely to reveal direct effects while later timepoints may reflect secondary consequences. The PI3K/Akt pathway appears particularly important for TLR10 signaling, so researchers should employ specific inhibitors (such as wortmannin or LY294002) to determine which effects depend on this pathway. For instance, studies have shown that PI3K/Akt inhibition completely blocks TLR10-induced IL-1Ra production at both mRNA and protein levels, suggesting this is a direct signaling consequence. Cross-linking experiments with anti-TLR10 antibodies can isolate TLR10-specific effects, while supernatant transfer experiments can identify effects mediated by soluble factors like IL-1Ra .
When interpreting TLR10 antibody cross-linking experiments, several factors require careful consideration. First, researchers must distinguish between blocking effects (where antibodies prevent ligand binding) and agonistic effects (where antibodies mimic ligand binding). This distinction can be addressed by comparing Fab fragments (which block but typically don't signal) with whole antibodies or cross-linked antibodies. Cross-linking with secondary antibodies enhances receptor clustering but may introduce artifacts if Fc receptors are engaged; using F(ab)'2 fragments of secondary antibodies can mitigate this concern. Appropriate isotype controls at equivalent concentrations are essential for accurate interpretation. Timing is also critical—immediate signaling events (minutes) likely represent direct TLR10 effects, while later changes (hours) may involve secondary mediators. For instance, cross-linking experiments with anti-TLR10 antibodies specifically induced IL-1Ra without producing IL-1β, IL-6, TNF-α, or IL-8, revealing a selective anti-inflammatory signaling pathway. Blocking experiments with PI3K/Akt inhibitors demonstrated this pathway's requirement for IL-1Ra induction, providing mechanistic insights into TLR10 signaling .
Studying human TLR10 presents unique challenges due to its absence in common laboratory mice. To address this species barrier, researchers can implement several strategies. First, human TLR10 transgenic mice provide a valuable in vivo model, though researchers must recognize that human TLR10 may interact differently with murine signaling components. Cell-specific transgenic expression (e.g., B-cell-restricted TLR10) might provide more precise insights into tissue-specific functions. For mechanistic studies, chimeric receptors combining human TLR10 extracellular domains with murine intracellular domains might enhance signaling compatibility in murine cells. Humanized mouse models with reconstituted human immune systems offer another approach, potentially allowing study of human TLR10 in a more native cellular context. Complementary in vitro studies with human primary cells remain essential, particularly focusing on B-cells where TLR10 function is well-established. When interpreting results across these models, researchers should carefully consider species-specific differences in signaling pathways, cytokine responses, and receptor expression patterns. Ultimately, multiple complementary approaches—transgenic mice, primary human cells, and adoptive transfer experiments—provide the most robust framework for understanding human TLR10 biology .
TLR10's unique inhibitory functions within the TLR family suggest several promising therapeutic directions. Given its ability to suppress B-cell activation and proliferation, TLR10 agonists might be developed to treat B-cell-mediated autoimmune diseases or antibody-driven pathologies. The induction of IL-1Ra without pro-inflammatory cytokines makes TLR10 particularly attractive as a targeted approach for inflammatory conditions where IL-1 plays a significant role. Research suggests that TLR10 engagement specifically dampens TLR2-driven inflammation, indicating potential applications in diseases with TLR2-mediated pathology. Conversely, TLR10 antagonists might enhance immune responses during vaccination or immunotherapy. To advance these applications, researchers should focus on identifying natural TLR10 ligands and developing synthetic agonists/antagonists with high specificity. Since genetic variation in TLR10 alters inflammatory responses, pharmacogenomic approaches might identify patient populations most likely to benefit from TLR10-targeted therapies. Future research should also explore the effects of TLR10 modulation in disease-specific contexts using relevant preclinical models and primary cells from patients with the targeted conditions .
Despite significant advances in understanding TLR10 function, its natural ligands remain unidentified. To discover these ligands, researchers should pursue several complementary approaches. Candidate-based screening could examine known TLR2 ligands, given TLR10's interaction with TLR2 pathways. This might include lipopeptides, lipoteichoic acids, and other microbial components. Unbiased approaches using purified TLR10 extracellular domain as "bait" in pull-down assays with microbial lysates could identify novel binding partners. Reporter cell systems expressing TLR10 and readouts for inhibitory signaling (such as suppression of NF-κB activation) might detect ligand activity in complex mixtures. Crosslinking studies with photoactivatable probes followed by mass spectrometry could identify molecules that physically interact with TLR10. Crystallographic studies of the TLR10 extracellular domain might reveal structural features that predict ligand specificity. Since TLR10 appears to modulate TLR2 responses, competitive binding assays could determine whether TLR10 directly competes with TLR1 or TLR6 for TLR2 heterodimerization or for TLR2 ligands. Finally, examining microorganisms that induce TLR10-dependent responses might narrow the search for relevant ligands .
Advanced "-omics" technologies offer powerful tools to expand our understanding of TLR10 biology beyond current knowledge boundaries. Transcriptomics approaches using RNA-seq could provide comprehensive profiles of gene expression changes following TLR10 engagement, building upon the PCR array findings that showed suppression of multiple activation-induced genes in B-cells. Proteomics studies could identify TLR10-interacting partners through approaches like proximity labeling or immunoprecipitation followed by mass spectrometry, potentially revealing novel components of TLR10 signaling complexes. Phosphoproteomics would be particularly valuable for mapping TLR10 signaling cascades, especially focusing on the PI3K/Akt pathway known to be involved in IL-1Ra induction. Single-cell approaches could reveal cell-specific responses and heterogeneity within populations, addressing whether all B-cells respond uniformly to TLR10 engagement. Epigenomic profiling might identify TLR10-mediated chromatin modifications that contribute to its sustained suppressive effects. Systems biology integration of these datasets could generate comprehensive models of TLR10 signaling networks and their cross-regulation with other immune pathways. Finally, population-level genomics studies could expand our understanding of how TLR10 polymorphisms impact disease susceptibility and inflammatory responses across diverse human populations .