Toll-like receptor 3 (TLR3) is a nucleotide-sensing TLR that recognizes double-stranded RNA, which is indicative of viral infection . Upon activation, TLR3 initiates an immune response via the adapter TRIF/TICAM1, leading to NF-kappa-B activation, IRF3 nuclear translocation, cytokine secretion, and inflammation . The TLR3 antibody, also known as Anti-TLR3 antibody [40C1285] (ab13915), is a mouse monoclonal antibody that targets TLR3 .
The TLR3 antibody, clone 40C1285, is a mouse monoclonal antibody . This antibody is suitable for various applications, including Western blotting (WB), immunohistochemistry (IHC-P), and flow cytometry (Intra) . It reacts with human and mouse samples . The immunogen corresponds to a synthetic peptide within human TLR3 aa 50-100 conjugated to Keyhole Limpet Haemocyanin . The antibody is of the IgG1 isotype .
The TLR3 antibody is utilized in several research applications to study immune responses and detect TLR3 expression in cells and tissues.
Western Blotting: The TLR3 antibody can be used to detect TLR3 protein in cell lysates and tissue samples .
Flow Cytometry: The TLR3 antibody can be used to analyze TLR3 protein expression in cell lines .
Immunohistochemistry: The TLR3 antibody can be used to stain formalin-fixed paraffin-embedded tissue sections .
The TLR3 antibody (ab13915) has been validated through various experimental techniques, demonstrating its specificity and efficacy in detecting TLR3. The data is summarized below.
| Lane | Sample | Result |
|---|---|---|
| 1 | Untransfected HEK293 cell lysate | TLR3 not detected |
| 2 | HEK293 cell lysate transfected with human TLR3 cDNA | TLR3 detected |
| 3 | Human intestine tissue lysate | TLR3 detected |
| 4 | Human placenta tissue lysate | Data not available |
| 5 | Human heart tissue lysate | Data not available |
| 6 | Human ovary tissue lysate | Data not available |
Figure 1: Immunohistochemical Analysis of TLR3 Antibody in Mouse Colon Tissue
Immunohistochemical analysis using ab13915 at 1/500 dilution showed intense signal in a subset of cells at the bases of the crypts in formalin-fixed paraffin-embedded mouse colon tissue sections .
The TLR3 antibody specifically recognizes the TLR3 protein, which is a key component of the innate and adaptive immune systems . TLR3 is activated by double-stranded RNA, a sign of viral infection, and acts via the adapter TRIF/TICAM1, leading to NF-kappa-B activation, IRF3 nuclear translocation, cytokine secretion, and the inflammatory response .
KEGG: ath:AT5G58010
STRING: 3702.AT5G58010.1
TLR3 functions as a pattern recognition receptor that specifically recognizes double-stranded RNA (dsRNA), a common viral molecular pattern. Upon recognition of dsRNA, TLR3 activates NF-kappaB and the IFN-beta promoter, initiating antiviral immune responses. TLR3 signaling is not triggered by either single-stranded RNA (ssRNA) or dsDNA, demonstrating its specificity for dsRNA molecular patterns .
LILRA3 acts as a soluble receptor for class I MHC antigens, binding both classical and non-classical HLA class I molecules, albeit with reduced affinities compared to related receptors LILRB1 or LILRB2. LILRA3 exhibits high-affinity binding to monocyte surfaces, resulting in the inhibition of LPS-induced TNF-alpha production, suggesting an immunomodulatory role in inflammatory responses .
TLR3 antibodies have demonstrated utility in studying virus-mediated immune responses, particularly those involving dsRNA recognition pathways. These antibodies can be used to assess TLR3-mediated signaling in experimental systems, including the suppression of poly(I):poly(C)-mediated IFN-beta production in human fibroblasts .
For LILRA3 antibodies such as ab235108, validated applications include Western Blotting (WB) and Immunohistochemistry on paraffin-embedded tissues (IHC-P), specifically with human samples. The antibody has been validated for detecting LILRA3 in human lung tissue at a dilution of 1/300 in immunohistochemical analysis following appropriate antigen retrieval in citrate buffer (pH 6.0) .
Antibody specificity for TLR3 can be validated through functional assays examining the antibody's ability to suppress TLR3-mediated responses. For example, researchers have confirmed specificity by demonstrating that monoclonal antibodies against TLR3 specifically suppress poly(I):poly(C)-mediated IFN-beta production in human fibroblasts naturally expressing TLR3 .
For LILRA3 antibodies, specificity can be validated through immunohistochemistry on known LILRA3-expressing tissues like human lung tissue. Proper controls should include antigen retrieval optimization and blocking with appropriate sera (e.g., 10% normal goat serum) to minimize non-specific binding .
TLR3 agonists can significantly enhance monoclonal antibody-based immunotherapies through multiple mechanisms. Research demonstrates that TLR3 stimulation via poly-ICLC enhances cetuximab-mediated antibody-dependent cellular cytotoxicity (ADCC) against head and neck cancer cells . This synergistic effect occurs through several pathways:
Increased NK cell cytotoxicity: TLR3-stimulated peripheral blood mononuclear cells (PBMCs) show robust enhancement of cetuximab-dependent ADCC, which is abrogated by NK cell depletion .
Enhanced NK cell activation: NK cells exposed to both TLR3 agonists and cetuximab express higher levels of activation markers CD107a and granzyme B than cells exposed to either stimulus alone .
Improved dendritic cell maturation: The combination of poly-ICLC and cetuximab induces substantial upregulation of costimulatory molecules CD80, CD83, and CD86 on dendritic cells, a process partially dependent on NK cells .
Enhanced cross-priming: Dendritic cells matured under these conditions demonstrate improved cross-priming abilities, generating higher numbers of antigen-specific CD8+ T cells .
The cytolytic activity of TLR3-stimulated NK cells varies significantly based on polymorphic variants of FcγRIIIa (at codon 158), which impacts antibody-dependent cellular cytotoxicity (ADCC). When designing experiments to investigate these effects, researchers should:
Genotype test subjects for FcγRIIIa polymorphisms prior to experimental design to account for this variable.
Use NK cells isolated from donors with different FcγRIIIa polymorphic variants to properly assess variation in ADCC potential.
Include appropriate controls for each polymorphic variant group when evaluating antibody efficacy.
Consider how TLR3 stimulation might differentially affect NK cells based on their FcγRIIIa genotype, as research has shown that cytolytic activity differs among cells expressing different polymorphic variants of FcγRIIIa when stimulated with TLR3 agonists .
Recent advances in artificial intelligence offer promising methodologies for antibody development. AI-based technologies can generate de novo antigen-specific antibody CDRH3 sequences using germline-based templates, effectively mimicking natural antibody generation processes like germline gene recombination and somatic hypermutation .
This approach has been successfully validated through the generation of antibodies against SARS-CoV-2, demonstrating that AI-based processes can bypass the complexity of traditional antibody generation methods while maintaining efficacy . This methodology offers several advantages:
Increased efficiency compared to traditional experimental approaches
Ability to rapidly generate candidate sequences for testing
Potential to optimize binding affinity through in silico methods
Reduced reliance on animal immunization protocols
To study LILRA3 binding interactions with class I MHC antigens, researchers can employ several methodological approaches:
Flow cytometry: Researchers can assess binding potency by analyzing LILRA3 interaction with cells expressing class I MHC antigens.
Competitive binding assays: These can determine the relative binding affinities compared to other receptors like LILRB1 or LILRB2.
Functional inhibition studies: Researchers can measure LILRA3's ability to inhibit LPS-induced TNF-alpha production by monocytes, which serves as a functional readout of its binding activity .
Immunoprecipitation assays: These can identify protein-protein interactions between LILRA3 and its binding partners.
For studying TLR3-ligand interactions and antibody blocking efficacy, several methodological approaches have proven effective:
Reporter gene assays: Using HEK293 cells transfected with human TLR3 expression vectors to measure NF-kappaB activation and IFN-beta promoter activity in response to dsRNA. This system can assess the specificity of TLR3 for different nucleic acid ligands (dsRNA vs. ssRNA or dsDNA) and the blocking capacity of anti-TLR3 antibodies .
Cytokine production assays: Measuring IFN-beta production by human fibroblasts naturally expressing TLR3 on their surface following poly(I):poly(C) stimulation, with and without anti-TLR3 antibodies .
Biolayer interferometry (BLI): Similar to the technique used for LAG-3 binding studies, BLI can be adapted to study TLR3-ligand interactions and antibody blocking, using streptavidin capture biosensors and purified components .
Flow cytometry: To assess antibody binding to cells expressing TLR3 and the competitive inhibition of ligand binding.
Based on the preclinical characterization of relatlimab (an anti-LAG-3 antibody), several methodological approaches can be applied to evaluate blocking antibodies against immune checkpoint receptors:
Biolayer interferometry (BLI): This technique effectively assesses the ability of antibodies to block receptor-ligand interactions. For example, researchers used BLI with streptavidin capture biosensors to evaluate how relatlimab blocked the interaction between LAG-3 and its ligands MHC II and FGL1 .
Binding affinity determination: The binding kinetics of antibodies to their targets can be characterized using surface plasmon resonance or BLI, determining association and dissociation rate constants (kon and koff) and equilibrium dissociation constants (KD) .
Functional cellular assays: These assess the ability of blocking antibodies to reverse receptor-mediated inhibition of T-cell function, measuring outcomes such as cytokine production, proliferation, or cytotoxicity .
Combination studies: For checkpoint inhibitors that might work synergistically (like LAG-3 and PD-1 blockade), combination studies can reveal nonredundant mechanisms and potential synergistic therapeutic effects .
When using LILRA3 antibodies for immunohistochemistry on paraffin-embedded tissues (IHC-P), researchers commonly encounter several technical challenges:
Insufficient antigen retrieval: For LILRA3 detection, high-pressure antigen retrieval in citrate buffer (pH 6.0) has been validated. Insufficient retrieval can result in false negatives. Optimization of retrieval conditions (time, temperature, buffer composition) is crucial for specific staining .
Background staining: Effective blocking is essential to prevent non-specific binding. Using 10% normal goat serum for blocking has been validated for LILRA3 antibody (ab235108) in IHC-P applications .
Antibody concentration: The optimal dilution for LILRA3 antibody (ab235108) in IHC-P has been determined to be 1/300. Using too high or too low antibody concentrations can result in non-specific staining or insufficient signal, respectively .
Tissue fixation variables: Overfixation can mask epitopes, while underfixation can result in poor tissue morphology. Standardizing fixation protocols can help ensure consistent results.
To optimize TLR3 antibody-based suppression of dsRNA-mediated signaling, researchers should consider:
Antibody concentration titration: Determine the optimal antibody concentration for maximum suppression of poly(I):poly(C)-mediated IFN-beta production.
Pre-incubation timing: Optimize the timing of antibody pre-incubation before dsRNA stimulation to achieve maximum blocking effect.
Cell type considerations: Different cell types may express varying levels of TLR3 or have different accessibility of TLR3 to antibodies. Human fibroblast cell lines naturally expressing TLR3 on the cell surface have been validated as effective models .
Readout selection: IFN-beta production has been validated as an effective readout for TLR3-mediated signaling in response to dsRNA. NF-kappaB activation can serve as an alternative readout in reporter systems .
Specificity controls: Include controls with ssRNA and dsDNA stimulation to confirm the specificity of the observed suppression to TLR3-mediated dsRNA recognition .
When encountering inconsistent results in ADCC assays using antibodies and TLR3 stimulation, researchers should implement these troubleshooting strategies:
FcγRIIIa polymorphism analysis: The cytolytic activity of TLR3-stimulated NK cells differs significantly among cells expressing different polymorphic variants of FcγRIIIa. Genotyping donor samples for FcγRIIIa polymorphisms and stratifying results accordingly can help explain variability .
NK cell isolation quality: Variations in NK cell purity and viability can affect ADCC potency. Using standardized isolation protocols and assessing cell viability and purity before experiments is crucial.
Timing of TLR3 stimulation: Optimize the timing of TLR3 agonist administration relative to antibody treatment, as this can affect NK cell activation status and subsequent cytotoxic function.
Standardized readouts: For consistent results, standardize cytotoxicity measurements using 51Cr release assays or flow cytometry-based methods measuring CD107a and granzyme B expression .
Target cell variability: Variations in target cell receptor expression can affect ADCC efficiency. Regular monitoring of target cell receptor expression levels is recommended.
AI-based technologies present revolutionary potential for antibody design through several mechanisms:
De novo generation of antigen-specific antibody CDRH3 sequences: AI algorithms can generate novel antibody sequences using germline-based templates, effectively mimicking natural B cell processes such as germline gene recombination and somatic hypermutation .
Efficiency improvements: AI-based processes bypass the complexity and time requirements of natural antibody generation, offering more efficient alternatives to traditional experimental approaches for antibody discovery .
Cross-reactivity prediction: AI models can potentially predict antibody cross-reactivity with non-target antigens, allowing researchers to screen out problematic candidates before experimental validation.
Structure-based optimization: AI can leverage structural information to optimize antibody-antigen interactions, potentially improving binding affinity and specificity.
Response prediction: Future AI systems might predict how specific antibodies will perform in diverse immune contexts, potentially forecasting efficacy in different patient populations.
The successful validation of AI-generated antibodies against SARS-CoV-2 demonstrates the practical application of these approaches and suggests broader potential for accelerating therapeutic antibody development .
Based on recent research, several promising applications for TLR3 agonists in combination with immunotherapeutic antibodies are emerging:
Enhanced ADCC in cancer immunotherapy: TLR3 agonists like poly-ICLC significantly improve cetuximab-mediated ADCC against head and neck cancer cells, suggesting broader applications with other therapeutic antibodies targeting different tumor types .
Improved dendritic cell maturation: The combination of TLR3 agonists and therapeutic antibodies induces robust upregulation of costimulatory molecules on dendritic cells, enhancing their ability to prime anti-tumor immune responses .
Cross-priming enhancement: TLR3 agonists combined with antibodies improve dendritic cell-mediated cross-priming of antigen-specific CD8+ T cells, potentially broadening the immune response beyond the initial antibody target .
Overcoming resistance mechanisms: TLR3 stimulation might help overcome resistance to antibody monotherapies by engaging multiple immune effector mechanisms simultaneously.
Vaccine adjuvant potential: TLR3 agonists could serve as effective adjuvants for therapeutic cancer vaccines, potentially synergizing with concurrent antibody therapies.
The molecular understanding of LILRA3 and TLR3 has several implications for future therapeutic antibody development:
Novel checkpoint targets: LILRA3's role as a soluble receptor for class I MHC antigens and its ability to bind monocytes and inhibit TNF-alpha production suggests potential applications in inflammatory disorders. Therapeutic approaches might involve LILRA3 mimetics or antagonists depending on the disease context .
Antiviral strategies: Understanding TLR3's role in recognizing viral dsRNA and initiating antiviral immune responses could inform the development of therapeutic antibodies that modulate rather than block TLR3 function, potentially enhancing beneficial antiviral responses while limiting inflammatory damage .
Combination therapy design: The demonstrated synergy between TLR3 stimulation and antibody therapies like cetuximab suggests a framework for designing combination approaches targeting other receptor-ligand pairs .
Personalized medicine applications: Knowledge of how FcγRIIIa polymorphisms affect TLR3-stimulated NK cell responses could inform patient selection strategies for antibody therapies, identifying individuals most likely to benefit from specific approaches .
Bispecific antibody development: Understanding the molecular interactions of these receptors could facilitate the development of bispecific antibodies targeting multiple pathways simultaneously, potentially achieving synergistic therapeutic effects with a single molecule.