TLR2 Antibody

Toll-like receptor 2, Mouse Anti Human
Shipped with Ice Packs
In Stock

Description

Definition and Biological Role of TLR2 Antibodies

TLR2 antibodies are monoclonal or polyclonal immunoglobulins that bind specifically to TLR2, a transmembrane protein expressed on immune cells (e.g., monocytes, dendritic cells) and non-immune cells (e.g., endothelial cells) . TLR2 recognizes pathogen-associated molecular patterns (PAMPs) from bacteria (e.g., Gram-positive lipopeptides), fungi, and viruses, initiating pro-inflammatory cytokine production (e.g., TNF-α, IL-6) . Antibodies against TLR2 block ligand-receptor interactions or downstream signaling, enabling mechanistic studies and therapeutic interventions .

Mechanism of Action

TLR2 antibodies function through two primary mechanisms:

  • Neutralization: Blocking ligand binding to TLR2’s extracellular domain (e.g., T2.5 antibody prevents Pam3_3CSK4_4 binding) .

  • Signal Inhibition: Disrupting TLR2 heterodimerization with co-receptors (TLR1/TLR6) or downstream adaptor proteins (MyD88, TIRAP) .

Table 1: Key TLR2 Antibodies and Their Mechanisms

Antibody NameClone/TargetMechanismApplication Model
T2.5Anti-TLR2 mAbBlocks Pam3_3CSK4_4 binding via SPR-confirmed inhibition Murine septic shock
MAB2616Clone 383936Neutralizes IL-8 secretion in HEK293 cells transfected with TLR2 In vitro cytokine assays
TL2.1IgG2a κImmunoprecipitates TLR2 (~90 kDa); inhibits microbial lipoprotein signaling Flow cytometry
OPN-305Humanized anti-TLR2Reduces myocardial infarct size in pigs by inhibiting TLR2-driven inflammation Porcine I/R injury

In Vitro Studies

  • Cytokine Modulation: Pre-treatment with TLR2 antibodies (e.g., CbTP) significantly reduces TNF-α, IL-6, and IL-1β production in human macrophages .

  • Pathogen Clearance: TLR2 antibodies enhance phagocytosis of Gram-positive bacteria by neutrophils .

In Vivo Models

  • Septic Shock: T2.5 antibody administration in mice inhibits lethal shock induced by Bacillus subtilis or Pam3_3CSK4_4, reducing TNF-α by >80% .

  • Cardioprotection: OPN-305 (humanized anti-TLR2) decreases infarct size by 40% and improves cardiac function in porcine myocardial ischemia-reperfusion models .

Clinical and Experimental Applications

Table 2: Applications of TLR2 Antibodies

ApplicationProtocol/OutcomeCitation
Western BlottingDetects TLR2 at 90–100 kDa in human lung tissue
Flow CytometryLabels TLR2 on monocytes (PE-conjugated TL2.1)
ImmunofluorescenceLocalizes TLR2 on HEK293 cell membranes
Therapeutic BlockadeReduces cytokine storm in sepsis

Challenges and Future Directions

  • Specificity: TLR2 antibodies may cross-react with other TLRs due to structural similarities .

  • Therapeutic Timing: Efficacy depends on early administration in acute inflammation (e.g., pre-reperfusion in myocardial injury) .

  • Biomarker Potential: TLR2 expression levels correlate with disease severity in sepsis and autoimmune disorders .

Product Specs

Introduction
TLR2, a member of the Toll-like receptor (TLR) family, plays a crucial role in pathogen recognition and innate immunity activation. Exhibiting high conservation from Drosophila to humans, TLRs share structural and functional similarities. These receptors recognize pathogen-associated molecular patterns (PAMPs) present on infectious agents, triggering the production of cytokines essential for effective immune responses. Notably, TLRs display distinct expression patterns. TLR2, primarily expressed in peripheral blood leukocytes, mediates host responses to Gram-positive bacteria and yeast by stimulating NF-kappaB.
Physical Appearance
Clear, colorless solution, sterilized by filtration.
Formulation
The solution contains 1mg/ml of TLR2 antibody in a buffer of PBS at pH 7.4 with 0.1% sodium azide.
Storage Procedures
For storage up to 1 month, maintain the antibody at 4°C. For longer-term storage, keep it at -20°C. Avoid repeated freeze-thaw cycles.
Stability / Shelf Life
The antibody remains stable for 12 months when stored at -20°C and for 1 month at 4°C.
Applications
This TLR2 antibody has undergone testing by ELISA and Western blot analysis to confirm its specificity and reactivity. However, due to variations in applications, it is recommended to titrate the antibody for each experiment to achieve optimal results. For Western blot analysis, a dilution range of 1:500 to 1:1000 is suggested, with a starting dilution of 1:500.
Synonyms
Toll-like receptor 2, Toll/interleukin-1 receptor-like protein 4, CD282, TLR2, TIL4.
Purification Method
TLR2 antibody was purified from mouse ascitic fluids by protein-G affinity chromatography.
Type
Mouse Anti Human Monoclonal.
Clone
P1F10AT.
Immunogen
Anti-human TLR2 mAb, is derived from hybridization of mouse SP2/O myeloma cells with spleen cells from BALB/c mice immunized with recombinant human TLR2 amino acids 221-384 purified from E. coli.
Ig Subclass
Mouse IgG1 heavy chain and κ light chain.

Q&A

What is the TLR2 antibody and what epitopes does it recognize?

TLR2 antibody (TL2.1) is a mouse monoclonal IgG2a kappa light chain antibody that specifically recognizes and binds to the TLR2 protein of human origin. This antibody targets specific epitopes within the extracellular domain of TLR2, which contains the pattern recognition region responsible for identifying pathogen-associated molecular patterns (PAMPs). Various TLR2 antibodies have been developed that target different epitopes, including those in the extracellular domain that can block ligand binding. The specificity of these antibodies has been validated through multiple techniques, including immunoprecipitation of both overexpressed and endogenous TLR2 from cell lysates, confirming their utility in both human and murine model systems .

How does TLR2 function in the immune system and why is it an important research target?

TLR2 plays a crucial role in the innate immune system as a pattern recognition receptor that identifies pathogen-associated molecular patterns, particularly from Gram-positive bacteria and certain viruses. This functionality is essential for initiating innate immune responses, as TLR2 activation triggers the recruitment of immune cells and production of pro-inflammatory cytokines, thereby enhancing the host's ability to combat infections. TLR2 is primarily located on the cell surface of immune cells such as macrophages and dendritic cells, where it effectively detects extracellular pathogens .

The structure of TLR2 includes a highly conserved Toll/IL-1 receptor (TIR) domain essential for downstream signaling, which activates transcription factors like NFκB that orchestrate inflammatory responses. TLR2 has been identified as central to the innate immune response to lipoproteins of Gram-negative bacteria, several whole Gram-positive bacteria, as well as a receptor for peptidoglycan and lipoteichoic acid and other bacterial cell membrane products . Its involvement in numerous inflammatory and neurodegenerative conditions, including Parkinson's disease, makes it a significant target for both basic research and therapeutic development .

What are the available forms of TLR2 antibodies and their applications?

TLR2 antibodies are available in multiple forms for various research applications:

Antibody FormCatalog ExampleConcentrationPrimary Applications
Non-conjugatedsc-21759200 μg/mlWestern blotting, IP
Agarose conjugatedsc-21759 AC500 μg/ml, 25% agaroseImmunoprecipitation
HRP conjugatedsc-21759 HRP200 μg/mlWestern blotting, ELISA
FITC conjugatedsc-21759 FITC200 μg/mlFlow cytometry, IF
PE conjugatedsc-21759 PE200 μg/mlFlow cytometry
Alexa Fluor conjugatesVariousVariousFluorescence microscopy

These various conjugated forms allow researchers to detect TLR2 through multiple methodologies including western blotting (WB), immunoprecipitation (IP), immunofluorescence (IF), and flow cytometry (FCM). Functional blocking antibodies like TL2.1 are particularly useful for studying TLR2's role as a pattern recognition receptor in microbial product-induced cytokine production by TLR2-bearing cells such as human peripheral blood mononuclear cells .

How should researchers design experiments to evaluate TLR2 antibody specificity?

To properly evaluate TLR2 antibody specificity, researchers should implement a multi-method validation approach:

  • Genetic controls: Compare antibody binding between wild-type and TLR2-knockout cells or tissues. In published research, antibodies like T2.5 have been validated by demonstrating absent binding to TLR2^-/- macrophages while successfully binding to wild-type macrophages .

  • Recombinant protein validation: Test antibody binding to purified recombinant TLR2 protein using ELISA or surface plasmon resonance (SPR) analysis. SPR analysis has been used to demonstrate direct and specific interaction between TLR2 and immunostimulatory lipopeptides, which can be blocked by antibodies in a dose-dependent manner .

  • Immunoprecipitation studies: Verify that the antibody can precipitate both overexpressed and endogenous TLR2. For instance, T2.5 antibody has been shown to successfully immunoprecipitate native murine and human TLR2 from lysates of HEK293 cells overexpressing the receptors, as well as endogenous TLR2 from RAW264.7 macrophage lysates .

  • Cross-reactivity testing: Test against related TLRs (particularly TLR1 and TLR6, which form heterodimers with TLR2) to confirm specificity.

  • Functional blocking validation: Assess the antibody's ability to inhibit known TLR2-mediated cellular responses, such as inhibition of cytokine production in response to TLR2 agonists like peptidoglycan (PGN), lipoteichoic acid (LTA), and Pam3CSK4 .

Western Blotting Protocol:

  • Prepare cell/tissue lysates in RIPA buffer with protease inhibitors

  • Separate proteins by SDS-PAGE (10-12% gel recommended)

  • Transfer to PVDF membrane (wet transfer recommended for optimal results)

  • Block with 5% non-fat milk in TBST for 1 hour at room temperature

  • Incubate with primary TLR2 antibody (1:500-1:1000 dilution) overnight at 4°C

  • Wash 3× with TBST

  • Incubate with appropriate secondary antibody for 1 hour at room temperature

  • Develop using chemiluminescence or fluorescence detection systems

Immunofluorescence Protocol:

  • Fix cells with 4% paraformaldehyde in PBS

  • Permeabilize with 0.1% Triton X-100 (for intracellular detection)

  • Block with 5% normal serum in PBS for 1 hour

  • Incubate with TLR2 antibody (5-10 μg/ml) overnight at 4°C

  • Wash 3× with PBS

  • Incubate with fluorophore-conjugated secondary antibody

  • Counterstain nucleus with DAPI

  • Mount and visualize under fluorescence microscope

Flow Cytometry Protocol:

  • Harvest cells and wash in cold PBS with 2% FBS

  • For extracellular staining: incubate cells with fluorophore-conjugated TLR2 antibody (5 μg/ml) for 30 minutes on ice in the dark

  • For intracellular staining: fix and permeabilize cells before antibody incubation

  • Wash 2× in PBS with 2% FBS

  • Analyze by flow cytometry, comparing to appropriate isotype controls

How can TLR2 antibodies be used to investigate TLR2-mediated inflammatory responses?

TLR2 antibodies can be employed to study inflammatory responses through several approaches:

  • Functional blocking studies: Apply TLR2-blocking antibodies (like TL2.1 or T2.5) before stimulation with TLR2 agonists (PGN, LTA, or Pam3CSK4) to assess the specific contribution of TLR2 signaling to inflammatory responses. This approach has demonstrated significant inhibition of cytokine production (TNF-α and IL-6) in cellular models like RAW264.7 macrophages .

  • Signaling pathway analysis: Use TLR2 antibodies in combination with inhibitors of downstream signaling molecules to elucidate the TLR2-specific inflammatory pathways. TLR2 has been shown to signal through MyD88 and TRAF6, leading to NF-κB activation, cytokine secretion, and inflammatory responses .

  • In vivo inflammation models: Administer TLR2-blocking antibodies in animal models of inflammation or septic shock to assess therapeutic potential. For example, systemic application of T2.5 antibody upon lipopeptide challenge has been shown to inhibit the release of inflammatory mediators like TNF-α and prevent lethal shock-like syndrome in mice .

  • Co-receptor studies: Use TLR2 antibodies alongside antibodies against TLR1 or TLR6 to investigate the functional interactions between these receptors in response to various bacterial products. TLR2 cooperates with TLR1 to mediate immune responses to bacterial lipoproteins or lipopeptides, and with TLR6 for responses to other bacterial components .

How can TLR2 antibodies contribute to neuroinflammation and neurodegenerative disease research?

TLR2 antibodies offer significant potential for investigating neuroinflammatory mechanisms in neurodegenerative conditions like Parkinson's disease:

  • Microglial activation studies: TLR2 is implicated in microglial activation, which plays a central role in neuroinflammation. Blocking antibodies can help determine the specific contribution of TLR2 to microglial activation in various disease models. Research has shown that Parkinson's is associated with an inflammatory response largely mediated by microglia, with TLR2 receptors being responsible for microglial activation and release of toxic factors .

  • Neuroprotection assessment: Researchers can evaluate whether TLR2-blocking antibodies provide neuroprotective effects in neurodegenerative disease models. This approach is exemplified by studies investigating whether TLR2 receptor antibodies can prevent the development of Parkinson's-like behavior in animal models and protect neurons from dying .

  • Inflammatory cascade characterization: TLR2 antibodies can help delineate the inflammatory cascade in the CNS by blocking this specific receptor while measuring various inflammatory mediators. This helps establish the position of TLR2 in the neuroinflammatory cascade.

  • Therapeutic development pipeline: Studies using TLR2 antibodies in animal models of neurodegeneration can serve as the foundation for therapeutic development. For example, Opsona has developed an antibody that blocks the TLR2 receptor for potential treatment of diseases like rheumatoid arthritis, which is being investigated for Parkinson's disease treatment. After demonstrating efficacy in direct brain administration, the antibody could be redesigned to more easily enter the brain, potentially preventing further neuronal loss in Parkinson's patients .

What strategies should be employed when investigating TLR2 heterodimer formation with TLR1 or TLR6?

Investigating TLR2 heterodimer formation requires specialized approaches:

  • Co-immunoprecipitation assays: Use TLR2 antibodies to pull down protein complexes, then probe for TLR1 or TLR6 to confirm heterodimer formation. This technique can reveal which ligands promote specific heterodimer formations.

  • Proximity ligation assays (PLA): This technique can detect protein-protein interactions in situ, allowing visualization of TLR2-TLR1 or TLR2-TLR6 heterodimers in their native cellular environment.

  • FRET/BRET analysis: Fluorescence or bioluminescence resonance energy transfer techniques can detect close proximity between fluorescently tagged TLR2 and its partners, providing evidence of heterodimer formation.

  • Selective agonist studies: Use TLR2 antibodies alongside selective agonists known to activate specific heterodimers (TLR2/1 or TLR2/6) to dissect the functional consequences of different heterodimer formations. Research has demonstrated that TLR2 cooperates with LY96 to mediate immune responses to bacterial lipoproteins and other microbial components, and with TLR1 specifically for responses to bacterial lipoproteins or lipopeptides .

  • Knockout/knockdown comparison: Compare responses in cells with normal TLR expression versus those with TLR1 or TLR6 knocked out/down to determine which heterodimer is responsible for specific responses to TLR2 agonists.

How can researchers determine the efficacy of TLR2-blocking antibodies in therapeutic applications?

Evaluating TLR2-blocking antibody efficacy for therapeutic applications involves several key experimental approaches:

  • Surface plasmon resonance (SPR) analysis: This technique can demonstrate direct interactions between TLR2 and its ligands, and quantify how effectively antibodies block these interactions. Studies have shown that antibodies like T2.5 can block TLR2-lipopeptide interactions in a dose-dependent manner, as measured by SPR .

  • In vitro cytokine inhibition assays: Measure the ability of TLR2 antibodies to inhibit cytokine production (particularly TNF-α and IL-6) in response to TLR2 agonists in relevant cell types. For example, anti-T20 antibody has been shown to significantly inhibit PGN, LTA, and Pam3CSK4-driven TNF-α and IL-6 production by RAW264.7 cells .

  • Animal model efficacy studies:

    • Septic shock models: Test antibody protection against lethal shock-like syndromes induced by TLR2 agonists or Gram-positive bacteria. T2.5 antibody has demonstrated protection against lethal shock induced by P3CSK4 or Gram-positive bacteria in mice .

    • Neuroinflammation models: Evaluate antibody effects on microglial activation, neuroinflammation, and neuroprotection in models of Parkinson's disease or other neurodegenerative conditions .

    • Allergic response models: Assess antibody effects in models of TLR2-driven allergic reactions. Anti-T20 antibody has protected OVA allergic mice from PGN-induced lethal anaphylaxis and reduced serum levels of TNF-α, IL-6, and LTC4 compared to isotype control-treated mice .

  • Biomarker measurement: Monitor relevant biomarkers of inflammation (cytokines, chemokines, acute phase proteins) to quantify therapeutic efficacy in animal models:

BiomarkerControl (pg/ml)TLR2 Agonist (pg/ml)TLR2 Antibody + Agonist (pg/ml)Inhibition (%)
TNF-α10-30800-1200100-30075-90%
IL-65-20500-90080-25070-85%
LTC42-10300-70040-15075-88%
  • CNS penetration studies: For neurological applications, evaluate the antibody's ability to cross the blood-brain barrier or develop modified delivery strategies. Current research indicates that some TLR2 antibodies do not readily cross into the brain, necessitating direct brain administration or redesign for improved CNS penetration .

How should researchers address inconsistent results when using TLR2 antibodies?

When facing inconsistent results with TLR2 antibodies, researchers should systematically troubleshoot:

  • Antibody validation: Re-validate antibody specificity using positive and negative controls (TLR2-knockout cells/tissues). Research has shown that TLR2 surface expression by murine macrophages can be surprisingly weak, while both intra- and extracellular expression increases upon systemic microbial challenge, potentially causing detection inconsistencies .

  • Expression level considerations: Account for variable TLR2 expression levels across different cell types and conditions. TLR2 is expressed on macrophages, smooth muscle, lung, spleen, thymus, brain, and adipose tissue, with particularly prominent expression in peripheral blood leukocytes, macrophages, and monocytes .

  • Proper controls: Include isotype controls to account for non-specific binding, particularly in flow cytometry and immunofluorescence applications.

  • Epitope accessibility: Consider that TLR2's conformation may change upon ligand binding or heterodimerization, potentially affecting epitope accessibility. Different antibodies targeting different epitopes may yield varying results depending on receptor activation state.

  • Protocol optimization checklist:

    • Adjust antibody concentration (titration experiments)

    • Modify incubation times and temperatures

    • Try different blocking reagents to reduce background

    • For intracellular detection, ensure proper fixation and permeabilization

    • For flow cytometry, verify viability staining to exclude dead cells

  • Heterodimer considerations: TLR2 functions through heterodimer formation with TLR1 or TLR6. Different experimental conditions may favor different heterodimer formations, potentially affecting antibody binding or functional studies .

How can researchers differentiate between TLR2-specific effects and off-target effects of TLR2 antibodies?

Differentiating TLR2-specific from off-target effects requires rigorous controls:

  • Genetic validation: Compare antibody effects in wild-type versus TLR2-knockout cells or animals. Functional studies have validated antibody specificity by showing absent effects in TLR2^-/- systems while maintaining effects in wild-type systems .

  • Multiple antibody approach: Use different TLR2 antibodies targeting distinct epitopes to confirm consistent results. If different antibodies targeting different TLR2 epitopes produce similar results, TLR2-specificity is more likely.

  • Isotype control comparison: Always compare results with matched isotype control antibodies to account for Fc receptor-mediated or other non-specific effects.

  • Complementary approaches: Complement antibody studies with genetic approaches (siRNA knockdown, CRISPR knockout) or small-molecule inhibitors when available.

  • Reconstitution experiments: In TLR2-deficient systems, reconstitute TLR2 expression and demonstrate restoration of antibody effects.

  • Dose-response relationships: Establish clear dose-response relationships for antibody effects, which typically follow predictable patterns for target-specific interactions.

  • Ligand competition assays: Demonstrate that antibody effects can be competed away by excess TLR2 ligands in a manner consistent with specific binding.

What are the critical considerations when interpreting changes in TLR2 expression levels using antibody-based detection methods?

When interpreting TLR2 expression data from antibody-based detection methods, researchers should consider:

  • Subcellular localization dynamics: TLR2 may be expressed both on the cell surface and intracellularly, with expression patterns changing upon activation. Research has shown that endogenous TLR2 is detectable both on the surface of primary murine human macrophages and within the cytoplasmic space, with expression patterns potentially shifting during inflammatory responses .

  • Reference gene/protein selection: Choose appropriate housekeeping genes/proteins that remain stable under your experimental conditions for normalization.

  • Quantification methods: For western blotting or flow cytometry, use appropriate quantification methods:

    • Flow cytometry: Report mean/median fluorescence intensity with appropriate statistics

    • Western blot: Use proper densitometry with linear range validation

  • Detection method limitations:

    • Western blotting detects total protein but not subcellular localization

    • Flow cytometry typically measures surface expression unless permeabilization is performed

    • Immunofluorescence provides localization information but can be less quantitative

  • Activation-induced changes: TLR2 expression can change rapidly upon activation. Studies have demonstrated that both intra- and extracellular TLR2 expression increases upon systemic microbial challenge, potentially affecting detection sensitivity and interpretation .

  • Heterodimer considerations: TLR2 functions through heterodimers with TLR1 or TLR6, which may affect epitope accessibility depending on the antibody used .

  • Species differences: Be aware of potential differences in TLR2 expression patterns between species (e.g., human vs. mouse) when translating findings across models.

How are TLR2 antibodies being applied in neurodegenerative disease research?

TLR2 antibodies are emerging as important tools in neurodegenerative disease research:

  • Parkinson's disease investigations: Research groups are developing and testing anti-TLR2 monoclonal antibodies as potential therapeutic agents for Parkinson's disease. These studies are based on findings that TLR2 receptors are responsible for the activation of microglia and release of toxic factors, with TLR2 levels increased in Parkinson's disease .

  • Blood-brain barrier challenges: Current research acknowledges that many TLR2 antibodies do not readily cross the blood-brain barrier. "Proof of concept" studies involve direct brain administration while researchers work to develop modified antibodies with improved CNS penetration .

  • Neuroprotection assessment: TLR2 antibodies are being tested for their ability to prevent neuronal death in animal models of neurodegenerative disease. Researchers are examining whether these antibodies can prevent the development of Parkinson's-like behavior in models and protect neurons from dying .

  • Microglial modulation: Rather than complete microglial suppression, researchers are using TLR2 antibodies to selectively modulate microglial activation, potentially allowing beneficial microglial functions while blocking harmful inflammatory responses.

  • Future therapeutic potential: If TLR2 receptor antibodies prove neuroprotective in animal models, they could be redesigned for improved brain penetration and tested clinically to potentially prevent further neuronal loss in Parkinson's patients. Future applications might even extend to preventing Parkinson's development in high-risk individuals .

What are the latest developments in using TLR2 antibodies for inflammatory and allergic disease research?

Recent developments in TLR2 antibody applications for inflammatory and allergic disease research include:

  • Anaphylaxis protection: Novel antibodies like anti-T20, which targets a specific 20-mer peptide in the extracellular domain of TLR2, have demonstrated protective effects against lethal anaphylaxis in animal models. This antibody protected OVA allergic mice from PGN-induced lethal anaphylaxis and reduced serum levels of inflammatory mediators .

  • Cytokine inhibition profiles: Researchers have characterized the inhibitory effects of various TLR2 antibodies on specific cytokine production patterns. For instance, anti-T20 significantly inhibited PGN, LTA, and Pam3CSK4-driven TNF-α and IL-6 production by RAW264.7 cells .

  • Domain-specific targeting: Rather than targeting the entire TLR2 extracellular domain, researchers are developing antibodies against specific epitopes or domains that may offer more selective blocking effects. Anti-T20 antibody targets a specific 20-mer peptide located in the extracellular domain of mouse TLR2 .

  • Septic shock intervention: TLR2 antibodies like T2.5 have shown therapeutic potential in models of septic shock. T2.5 prevents lethal shock-like syndromes induced by P3CSK4 or Gram-positive bacteria in mice, suggesting potential clinical applications in severe sepsis .

  • Dual-targeting approaches: Some researchers are exploring combinations of antibodies targeting both TLR2 and its co-receptors (TLR1, TLR6) to achieve more complete blocking of inflammatory pathways. This approach recognizes that TLR2 cooperates with TLR1 to mediate immune responses to bacterial lipoproteins/lipopeptides and with TLR6 for responses to other bacterial components .

How might advanced imaging techniques enhance TLR2 antibody applications in research?

Advanced imaging techniques are expanding the utility of TLR2 antibodies in research:

  • Super-resolution microscopy: Techniques like STORM or PALM combined with TLR2 antibodies can reveal nanoscale organization of TLR2 receptors on the cell surface, potentially identifying signaling clusters or reorganization upon activation.

  • Live-cell imaging: Using minimally disruptive fluorescently-tagged Fab fragments of TLR2 antibodies allows tracking of receptor trafficking and clustering dynamics in real-time in living cells.

  • Multi-color imaging: Simultaneous visualization of TLR2 (using specific antibodies) along with TLR1, TLR6, and downstream signaling components can elucidate the spatiotemporal dynamics of heterodimer formation and signal propagation.

  • Intravital microscopy: TLR2 antibodies conjugated to appropriate fluorophores enable visualization of TLR2 expression and trafficking in live animal models during inflammatory responses or disease progression.

  • Correlative light-electron microscopy (CLEM): This technique combines the specificity of fluorescently-labeled TLR2 antibodies with the ultrastructural context provided by electron microscopy, giving insights into the precise subcellular localization of TLR2.

  • PET imaging with radiolabeled antibodies: For in vivo applications, particularly in neurodegenerative disease research, radiolabeled TLR2 antibodies could potentially allow non-invasive monitoring of neuroinflammation and therapeutic efficacy.

  • Functional imaging combinations: Combining TLR2 antibody staining with calcium imaging or other functional readouts can link receptor engagement to immediate downstream signaling events.

What are the most critical considerations for researchers selecting TLR2 antibodies for their studies?

When selecting TLR2 antibodies, researchers should prioritize:

  • Validation status: Choose antibodies with comprehensive validation data, including demonstrated specificity in both positive controls (wild-type cells/tissues) and negative controls (TLR2-knockout systems) .

  • Application suitability: Select antibodies specifically validated for your intended application (western blotting, immunofluorescence, flow cytometry, functional blocking). Different applications may require different antibody characteristics .

  • Species reactivity: Ensure the antibody recognizes TLR2 from your species of interest. Some antibodies like T2.5 have been validated for both murine and human TLR2, which is valuable for translational research .

  • Epitope location: Consider whether the epitope location matters for your research question. Antibodies targeting functional domains may be more effective for blocking studies, while antibodies against well-conserved regions may be better for detection across species .

  • Functional vs. detection applications: For functional studies, select antibodies with demonstrated blocking activity (e.g., TL2.1 or T2.5). For detection studies, prioritize sensitivity and specificity in your application of interest .

  • Clone information and reproducibility: Prefer antibodies with transparent information about the clone, allowing for reproducibility across studies and laboratories.

  • Technical support: Choose suppliers that provide detailed protocols and technical support for optimization in specific applications.

Product Science Overview

Introduction

Toll-like receptors (TLRs) are a critical component of the innate immune system, acting as the first line of defense against pathogens. Among these, Toll-like receptor 2 (TLR2) is one of the most studied pattern recognition receptors (PRRs). TLR2 is known for its ability to recognize a wide range of microbial components by forming heterodimers with other TLRs, such as TLR1, TLR4, TLR6, and TLR10 .

Structure and Function

TLR2 is a membrane-bound receptor that plays a pivotal role in the immune response by recognizing pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs). Upon ligand binding, TLR2 activates intracellular signaling pathways that lead to the production of pro-inflammatory cytokines and type I interferons . This activation is crucial for initiating and modulating the immune response to infections and tissue damage.

Dimerization and Ligand Recognition

One of the unique features of TLR2 is its ability to form heterodimers with other TLRs. This dimerization expands the range of ligands that TLR2 can recognize. For instance, TLR2/TLR1 heterodimers are known to recognize triacylated lipopeptides, while TLR2/TLR6 heterodimers recognize diacylated lipopeptides . This versatility allows TLR2 to detect a broad spectrum of microbial components, including those from Gram-positive bacteria, fungi, and parasites .

Role in Inflammation and Disease

TLR2 is involved in both pro-inflammatory and anti-inflammatory processes. While it is essential for mounting an effective immune response, overactivation of TLR2 can lead to chronic inflammation and contribute to various inflammatory and autoimmune diseases . Recent studies have also highlighted the role of TLR2 in neuroinflammation and neurodegenerative diseases, such as Alzheimer’s disease .

Mouse Anti Human TLR2 Antibodies

Mouse anti-human TLR2 antibodies are widely used in research to study the expression and function of TLR2 in human cells. These antibodies are typically monoclonal, meaning they are derived from a single clone of immune cells and recognize a specific epitope on the TLR2 protein. They are invaluable tools for investigating the role of TLR2 in various physiological and pathological processes.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.