CLEC-2 is a type II transmembrane protein belonging to the C-type lectin-like domain (CTLD) superfamily. Key features include:
Domain: Homodimeric structure with a single hemITAM (hem-immunoreceptor tyrosine-based activation motif) in its cytoplasmic tail .
Expression: Primarily on platelets, megakaryocytes, and weakly on myeloid cells .
CLEC-2 binds ligands such as podoplanin (on tumor cells) and rhodocytin (a snake venom protein), triggering Syk- and PLCγ-dependent signaling pathways .
Multiple monoclonal and polyclonal antibodies have been developed for research and diagnostic applications. Below is a comparative analysis:
CLEC-2 deficiency in mice prolongs bleeding time and impairs thrombus stability .
Antibody-mediated depletion (e.g., INU1) reduces arterial occlusion in thrombosis models .
Humanized CLEC-2 mice show delayed vessel occlusion, suggesting interspecies functional differences .
Anti-CLEC-2 antibodies (e.g., AYP1) block tumor metastasis by inhibiting platelet-tumor cell interactions .
CLEC-2 immunodepletion does not induce thrombocytopenia, unlike GPVI-targeting therapies .
Flow cytometry: Detects CLEC-2 on platelets (e.g., APC-conjugated clones) .
Western blot: Identifies CLEC-2 in platelet lysates (~35 kDa band) .
Functional assays: Measures Syk phosphorylation and platelet aggregation .
CLEC2 (C-type lectin-like receptor 2) is a unique platelet activation receptor that signals through a single YXXL sequence, representing half of an immunoreceptor tyrosine-based activation motif (hemITAM) . The receptor is primarily expressed on platelets with approximately 2000 copies per cell, although murine CLEC2 has also been detected on peripheral blood neutrophils . CLEC2 antibodies are essential research tools because they allow for:
Identification and quantification of CLEC2 expression on platelets and other cell types
Investigation of CLEC2-mediated signaling pathways
Exploration of CLEC2's roles in thrombosis, tumor metastasis, and development
Development of potential antithrombotic therapeutics targeting CLEC2
The significance of these antibodies is highlighted by their use in demonstrating that CLEC2 and its endogenous ligand podoplanin are crucial for normal development, with mice deficient in either showing defective blood-lymphatic vessel separation .
CLEC2 antibodies target a receptor with unique regulatory characteristics that distinguish it from other platelet receptor antibodies:
Unlike antibodies against GPVI and FcγRIIa (which target receptors that undergo proteolytic regulation), CLEC2 antibodies recognize a receptor that is not regulated by proteolysis upon activation .
CLEC2 antibodies can both activate the receptor (when cross-linked) and block its function (as Fab fragments), allowing for versatile experimental approaches .
Some CLEC2 antibodies (such as AYP1) recognize conformational epitopes that are lost during the denaturing conditions of SDS-PAGE, making them suitable for specific applications like flow cytometry but not western blotting .
CLEC2 antibodies can induce proteolysis of other platelet receptors (GPVI and FcγRIIa) without affecting CLEC2 itself, revealing unique cross-regulatory mechanisms between platelet receptors .
This distinct profile makes CLEC2 antibodies particularly valuable for studying the integration of signaling pathways in platelets.
When working with CLEC2 antibodies, researchers should be aware of several nomenclature and variant considerations:
CLEC2 is also known as CLEC1B in humans, and antibodies may be labeled with either designation .
Related family members include CLEC2B (also known as AICL, Clrb, and CLECSF2), which represents a distinct target with different expression patterns and functions .
Species-specific variants exist with important differences:
Different antibody clones recognize distinct epitopes on CLEC2, affecting their functional properties:
Understanding these distinctions is essential for selecting the appropriate antibody and interpreting experimental results correctly.
Optimizing flow cytometry for CLEC2 antibody detection requires specific methodological considerations:
Sample preparation protocol:
Use fresh blood samples collected in acid-citrate-dextrose (ACD) or sodium citrate anticoagulant
For platelets, prepare platelet-rich plasma (PRP) by centrifugation at 200g for 20 minutes
Fix samples with 1% paraformaldehyde if immediate analysis is not possible
Antibody selection and controls:
Gating strategy:
First gate on platelet population based on forward/side scatter characteristics
Use CD41 positivity to confirm platelet identity
Analyze CLEC2 expression on CD41+ events
Special considerations:
This methodology has successfully demonstrated that CLEC2 expression is restricted to platelets in human blood, with no detectable expression on monocytes, neutrophils, T cells, B cells, or dendritic cells .
CLEC2 immunoprecipitation experiments require specific protocols to effectively isolate and study this receptor:
Cell lysis procedure:
Wash platelets (3×10⁸) in modified Tyrode's buffer
Lyse in 1% NP-40 lysis buffer containing:
50 mM HEPES (pH 7.4)
150 mM NaCl
1% NP-40
Protease inhibitor cocktail
Phosphatase inhibitors (2 mM sodium orthovanadate, 5 mM sodium fluoride)
Immunoprecipitation steps:
Pre-clear lysates with protein G-Sepharose for 1 hour at 4°C
Incubate with anti-CLEC2 antibody (e.g., AYP1) at 2-5 μg per sample overnight at 4°C
Add protein G-Sepharose and incubate for 2 hours at 4°C
Wash beads 3× with lysis buffer
Elute bound proteins by boiling in SDS sample buffer
Detection methods:
Critical considerations:
This approach has been successfully used to investigate CLEC2 signaling pathways and interactions with other proteins in platelet activation cascades.
Studying CLEC2 internalization and trafficking using antibodies requires specialized approaches:
Time-course receptor tracking protocol:
Label surface CLEC2 with fluorescently tagged antibodies (e.g., AYP1-FITC)
Induce activation with CLEC2 agonists (e.g., rhodocytin)
Monitor receptor localization at fixed timepoints (0, 15, 30, 60, 120 minutes)
Quantify surface expression changes by flow cytometry
Distinguishing mechanisms of receptor regulation:
Use broad-spectrum metalloproteinase inhibitors (e.g., GM6001) to block potential shedding
Apply membrane-permeable calpain inhibitors (e.g., E64d) to prevent intracellular proteolysis
Employ endocytosis inhibitors to block internalization
Measure surface versus intracellular receptor pools
Comparative analysis with other platelet receptors:
Methodological considerations:
Combine flow cytometry with confocal microscopy for spatial resolution
Use fluorescence quenching techniques to distinguish surface from internalized receptors
Apply pH-sensitive fluorophores to track movement through acidic endosomal compartments
Research has demonstrated that unlike other platelet receptors, CLEC2 is neither shed nor internalized from the platelet surface upon autoactivation or in response to activation of GPVI and FcγRIIa , making it unique among platelet receptors.
Antibody-mediated depletion of CLEC2 represents a powerful approach for investigating receptor function in vivo:
Established depletion protocols:
For humanized CLEC2 mouse models (hCLEC-2 KI), intraperitoneal injection of anti-human CLEC2 antibodies:
Dose-dependent effects: 1-2 mg/kg typically achieves significant depletion
Monitoring: Flow cytometric assessment of platelet CLEC2 surface expression at regular intervals
Mechanism of antibody-mediated CLEC2 depletion:
Experimental applications:
Investigation of CLEC2's role in thrombosis models
Assessment of hemostatic function in the absence of CLEC2
Evaluation of potential antithrombotic strategies targeting CLEC2
Study of tumor metastasis and developmental processes
Advantages over genetic models:
These approaches have demonstrated that immunodepletion of human CLEC2 in humanized mouse models can be achieved, providing proof of principle for testing anti-human CLEC2 agents in vivo .
Investigating CLEC2-mediated signaling requires sophisticated methodological approaches:
Antibody-based stimulation protocols:
Direct activation: Cross-linked whole antibodies (e.g., AYP1 IgG) can trigger CLEC2 signaling
Selective blockade: Fab fragments (e.g., AYP1 Fab) can block activation without inducing signaling
Controls: Both AYP1 and HEL1 antibodies stimulate platelet aggregation, but their Fab fragments do not, confirming that CLEC2 dimerization at either site is sufficient to trigger activation
Signaling pathway analysis methods:
Phosphorylation assays: Anti-phosphotyrosine western blotting to detect hemITAM phosphorylation
Kinase inhibition studies: Selective inhibition of Src (PP2) and Syk (PRT-060318) kinases to dissect pathway dependencies
Calcium flux measurements: Real-time monitoring of intracellular calcium release
Integrin activation assays: Flow cytometric measurement of αIIbβ3 activation
Cross-talk analysis with other platelet receptors:
Advanced techniques for detailed signaling analysis:
Proximity ligation assays to detect protein-protein interactions
Phosphoproteomic analysis for comprehensive pathway mapping
CRISPR-edited cell lines to confirm signaling dependencies
Live-cell imaging with fluorescent biosensors for real-time signaling visualization
These methodological approaches have revealed that CLEC2 signaling involves a unique Src- and Syk-dependent cascade that shares components with but is distinct from ITAM receptor pathways .
Working with CLEC2 antibodies in humanized mouse models requires specific experimental considerations:
Model system characteristics:
Humanized CLEC-2 (hCLEC-2 KI) mice replace mouse CLEC2 with the human variant
Surface expression of hCLEC-2 is approximately double that on human platelets
Other platelet glycoprotein receptors remain comparable to wild-type mice
Platelet activation and aggregation responses to G protein-coupled receptors and GPVI agonists are normal
Antibody selection criteria:
Human CLEC2-specific antibodies (e.g., AYP1, HEL1) are required
AYP1 and HEL1 act at different sites on CLEC2, providing complementary approaches
Both whole antibodies and Fab fragments should be prepared for different applications
Species-matched secondary antibodies are essential for detection protocols
Functional assay adaptations:
Thrombosis models: FeCl₃-induced carotid artery thrombosis with intravital microscopy
Hemostasis assessment: Tail bleeding time measurement
Platelet function: Aggregometry with CLEC2 agonists (rhodocytin) and other platelet activators
Flow chamber assays: Thrombus formation on collagen under defined shear conditions
Critical controls and validation steps:
Confirm human CLEC2 expression by flow cytometry
Verify antibody binding specificity in humanized versus wild-type mice
Include appropriate isotype controls for in vivo studies
Monitor potential immune responses against humanized proteins or administered antibodies
This model system has demonstrated that human CLEC2 can functionally replace mouse CLEC2 during development and in hemostasis, suggesting conserved interactions between CLEC2 and podoplanin in humanized mice .
Resolving contradictory findings about CLEC2 expression on immune cells requires methodological precision:
Reconciling contradictory expression data:
Murine studies demonstrate CLEC2 expression on peripheral blood neutrophils but only weakly on bone-marrow or elicited inflammatory neutrophils
This apparent contradiction can be explained by:
Species differences in CLEC2 expression patterns
Distinct antibody specificities and detection sensitivities
Sample preparation variables affecting epitope accessibility
Standardized detection protocol:
| Cell Type | Sample Preparation | Antibody Selection | Flow Cytometry Settings | Positive Control |
|---|---|---|---|---|
| Platelets | Minimal processing, gentle fixation | Direct fluorophore conjugates | High sensitivity for small cells | CD41+ gating |
| Neutrophils | Gradient isolation, avoid activation | Multiple epitope antibodies | Adjusted for larger cells | Ly6G+ (mouse) or CD66b+ (human) |
| Other Immune Cells | Minimal RBC lysis, maintain viability | Multiple clones and conjugates | Exclude platelet contamination | Lineage-specific markers |
Critical validation approaches:
Experimental factors affecting detection:
These approaches have helped clarify that murine CLEC2 has broader expression than human CLEC2, explaining some conflicting reports in the literature .
Detecting CLEC2 post-translational modifications presents several technical challenges that require specific approaches:
Phosphorylation detection challenges:
The single YXXL motif (hemITAM) of CLEC2 has lower stoichiometry of phosphorylation than dual ITAM receptors
Solution approach: Enhance sensitivity using:
Phosphatase inhibitor cocktails during cell lysis
Anti-phosphotyrosine antibodies for immunoprecipitation followed by CLEC2 detection
Phos-tag™ SDS-PAGE for mobility shift detection of phosphorylated species
Mass spectrometry for precise phosphosite mapping
Glycosylation analysis difficulties:
CLEC2 contains multiple potential N-glycosylation sites
Solution approach: Systematic glycoproteomic analysis:
| Method | Application | Advantages | Limitations |
|---|---|---|---|
| PNGase F treatment | Removes N-linked glycans | Simple, effective | Loses glycan information |
| Lectin binding assays | Identifies glycan types | Non-destructive | Limited specificity |
| Mass spectrometry | Detailed glycan structure | Comprehensive | Complex analysis |
| Site-directed mutagenesis | Functional significance | Direct causality | Potential structural effects |
Conformational epitope preservation:
Dimerization and complex formation detection:
CLEC2 dimerization is critical for signaling but challenging to detect
Solution approach: Advanced biophysical techniques:
Chemical crosslinking prior to SDS-PAGE
Blue native PAGE for complex detection
Förster resonance energy transfer (FRET) for live-cell analysis
Single-molecule tracking in membrane microdomains
These technical approaches have revealed that CLEC2 undergoes significant post-translational regulation, including dimerization-dependent signaling that is distinct from proteolytic regulation seen with other platelet receptors .
Interpreting conflicting results between different CLEC2 antibody clones requires systematic analysis:
Epitope-dependent functional differences:
AYP1 and HEL1 antibodies act at different sites on CLEC2
HEL1 Fab fragments neither block rhodocytin-induced platelet aggregation nor block AYP1 IgG-induced aggregation
This indicates distinct binding sites with different functional consequences
Resolution approach: Map epitopes using:
Competition binding assays
Deletion/mutation constructs
Hydrogen-deuterium exchange mass spectrometry
X-ray crystallography of antibody-antigen complexes
Systematic antibody validation framework:
| Validation Parameter | Method | Expected Outcome | Troubleshooting |
|---|---|---|---|
| Specificity | Western blot with KO controls | Single band at expected MW or no signal with conformational epitopes | Test multiple antibody concentrations |
| Sensitivity | Titration on recombinant protein | Consistent detection limit | Optimize signal amplification |
| Functional effect | Platelet aggregation assays | Clone-specific activation or inhibition | Test both whole IgG and Fab fragments |
| Cross-reactivity | Testing on related proteins | No binding to other family members | Confirm with genetic knockouts |
Application-specific selection criteria:
Data integration approaches:
Triangulate results using multiple antibody clones
Correlate antibody-based findings with genetic approaches
Consider species differences in epitope conservation
Evaluate potential steric hindrance between different antibodies
This analytical framework has revealed that differences between antibody clones can provide valuable insights into receptor function, as demonstrated by the distinct effects of AYP1 and HEL1 on CLEC2 signaling .
The development of CLEC2 antibodies as therapeutic agents represents an emerging research direction:
Therapeutic rationale:
CLEC2 deficiency reduces vessel occlusion in thrombosis models with minimal effects on hemostasis
Occlusion is unaltered in CLEC2 Y7A signaling-null mice, suggesting receptor presence rather than signaling affects thrombus stability
Immunodepletion of CLEC2 using antibodies has similar effects on thrombus formation
These findings position CLEC2 as a potential antithrombotic target with a favorable risk-benefit profile
Current development approaches:
Humanized mouse models (hCLEC-2 KI) to evaluate human CLEC2-targeted antibodies in vivo
Testing of different antibody formats:
Whole IgG for immunodepletion strategies
Fab fragments for functional blocking without depletion
F(ab')2 fragments for receptor dimerization without Fc effects
Evaluation of administration routes and pharmacokinetic profiles
Efficacy and safety assessment strategy:
| Parameter | Methodology | Key Findings | Implications |
|---|---|---|---|
| Antithrombotic efficacy | FeCl₃-induced carotid artery thrombosis | Reduced vessel occlusion | Potential therapeutic benefit |
| Bleeding risk | Tail bleeding time | Minimal prolongation | Favorable safety profile |
| Immunogenicity | Anti-drug antibody detection | Model-dependent | Requires humanization strategies |
| Duration of effect | Flow cytometric monitoring | 11-24 days for depletion | Dosing interval guidance |
Translational research directions:
Optimizing humanized antibodies to minimize immunogenicity
Exploring bispecific antibodies targeting both CLEC2 and complementary pathways
Developing site-specific antibody-drug conjugates
Investigating small-molecule CLEC2 inhibitors as alternatives to antibody therapies
The humanized CLEC2 mouse model has provided proof of principle that anti-human CLEC2 agents can be tested in vivo, paving the way for preclinical evaluation of potential therapeutics .
CLEC2 antibody applications in microparticle and extracellular vesicle research require specialized methodological considerations:
Isolation protocols for CLEC2-bearing microparticles:
Differential centrifugation: 1,500g (15 min) → 13,000g (2 min) → 20,000g (20 min) for microparticle enrichment
Size-exclusion chromatography for separation based on particle size
Immunoaffinity capture using anti-CLEC2 antibodies (e.g., AYP1)
Flow cytometry-based sorting of CLEC2+ microparticles
Detection and characterization strategies:
High-sensitivity flow cytometry using:
Calibrated size beads for size determination
Fluoresceinated anti-CLEC2 antibodies (e.g., AYP1)
Co-staining for platelet markers (CD41) to confirm origin
Annexin V binding to detect phosphatidylserine exposure
Nanoparticle tracking analysis with fluorescent antibody labeling
Electron microscopy with immunogold labeling using anti-CLEC2 antibodies
Functional assessment methods:
Binding assays to podoplanin-expressing cells
Signaling capacity in recipient cells
Procoagulant activity measurements
Immunomodulatory effects on target cells
Critical experimental considerations:
CLEC2 is expressed on microparticles derived from activated platelets, while GPVI is not
Pre-analytical variables (collection, processing, storage) significantly affect results
Standardization using reference materials improves inter-study comparability
Combination of multiple techniques provides more comprehensive characterization
This research area has revealed that CLEC2 and GPVI have distinct patterns of distribution on microparticles, with CLEC2 but not GPVI being expressed on microparticles derived from activated platelets , suggesting different functional roles in intercellular communication.
Innovative approaches for investigating CLEC2-podoplanin interactions using antibodies are advancing our understanding of this important receptor-ligand pair:
Advanced imaging techniques:
Super-resolution microscopy (STORM/PALM) with dual-labeled antibodies
Intravital microscopy with fluorescently tagged antibodies for in vivo visualization
Correlative light and electron microscopy (CLEM) for ultrastructural context
Live-cell single-molecule tracking of receptor-ligand interactions
Biosensor development for real-time interaction studies:
FRET-based biosensors using antibody-conjugated fluorophore pairs
Bioluminescence resonance energy transfer (BRET) systems
Surface plasmon resonance with immobilized antibodies or recombinant proteins
Quartz crystal microbalance with dissipation monitoring (QCM-D) for binding kinetics
Engineered antibody formats for specific applications:
| Antibody Format | Application | Advantages | Limitations |
|---|---|---|---|
| Bispecific (CLEC2/podoplanin) | Co-localization studies | Simultaneous targeting | Complex production |
| Nanobodies | In vivo imaging, therapeutics | Small size, tissue penetration | Potentially immunogenic |
| scFv fragments | Biosensors, targeting | Monovalent binding | Reduced stability |
| Photoswitchable antibodies | Controlled activation | Spatiotemporal control | Requires specialized equipment |
Experimental models for interaction studies:
These approaches have demonstrated that human CLEC2 can compensate for mouse CLEC2 during development and in hemostasis, suggesting a conserved interaction between CLEC2 and podoplanin in humanized mice and providing new opportunities for studying this critical receptor-ligand pair in health and disease.