Melanoma: Layilin is highly expressed on clonally expanded, exhausted CD8+ tumor-infiltrating lymphocytes (TILs). It colocalizes with integrin αLβ2 (LFA-1) to enhance cellular adhesion, promoting cytotoxic T cell retention in tumors. Genetic deletion of LAYN reduces LFA-1-dependent adhesion and impairs tumor cell killing .
Hepatocellular Carcinoma (HCC): LAYN+ CD8+ T cells exhibit exhaustion markers (e.g., CD39, TIM3) and reduced cytotoxic activity. Blocking LAYN partially restores their function, suggesting therapeutic potential .
Integrin Activation: Layilin enhances LFA-1 adhesiveness by colocalizing with αLβ2 integrins. This interaction is critical for immune synapse formation in cytotoxic T cells .
Immune Regulation:
LAYN Antagonism: In HCC, LAYN blockade reverses CD8+ T cell exhaustion, restoring cytotoxic activity .
Biomarker Potential: High LAYN expression is associated with immune-hot tumors, making it a candidate for predicting immunotherapy response .
Layilin (LAYN) is a C-type lectin domain-containing membrane glycoprotein that functions as a receptor for hyaluronate and interacts with several proteins including NF2, RDX, and TLN1 . LAYN has emerged as a significant molecule in immunological research due to its selective expression on highly activated, clonally expanded CD8+ T cells in various human cancers, including melanoma and hepatocellular carcinoma (HCC) . Its importance stems from its role in augmenting integrin-mediated cellular adhesion, which enhances antitumor immunity . Recent studies have identified LAYN as a valuable prognostic biomarker across various tumor types and as an indicator of dysfunctional or exhausted T cells .
Polyclonal LAYN antibodies:
Recognize multiple epitopes on the LAYN protein
Examples include rabbit polyclonal antibodies (20535-1-AP) that detect LAYN in WB, IHC, and IF applications
Advantages: Higher sensitivity due to recognition of multiple epitopes, more tolerant to minor protein changes
Best used for: Initial protein detection, applications requiring high sensitivity
Monoclonal LAYN antibodies:
Target a single epitope on the LAYN protein
Examples include mouse monoclonal antibodies like clone PAT20G8AT and rabbit recombinant monoclonal antibodies (EPR11875(2))
Advantages: Higher specificity, more consistent lot-to-lot performance, reduced background
Best used for: Quantitative applications, experiments requiring high reproducibility
The choice between polyclonal and monoclonal depends on experimental needs. For novel research where LAYN detection may be challenging, polyclonals offer better sensitivity. For precise quantification or reproducible experiments, monoclonals are preferable .
LAYN is highly expressed on clonally expanded but phenotypically exhausted CD8+ T cells in human cancers, making it a valuable marker for studying T cell exhaustion . A methodological approach includes:
Sample preparation: Isolate tumor-infiltrating lymphocytes (TILs) from tumor samples using tissue dissociation and gradient centrifugation.
Multi-parameter flow cytometry analysis:
Include LAYN antibody in panels with other exhaustion markers (PD-1, TIM-3, LAG-3)
Use proper compensation controls and FMO (fluorescence minus one) to establish gating strategies
Analyze LAYN expression specifically within clonally expanded T cell populations
Functional assessment:
Correlative analyses:
This approach has revealed that CD8+ T cells overexpressing LAYN exhibit characteristics of exhaustion and diminished antitumor effects, but treatment with LAYN antagonists can partially restore their functionality .
Proper antibody validation is critical for reproducible research . For LAYN antibodies, include:
Essential controls:
Genetic knockout/knockdown validation:
Expression system validation:
Overexpression of tagged LAYN in appropriate cell lines
Parallel detection with anti-tag antibody and LAYN antibody
Peptide competition/blocking:
Pre-incubate antibody with the immunizing peptide/recombinant protein
Signal should be significantly reduced or eliminated
Application-specific controls:
For WB: Include molecular weight markers to confirm band at expected size (43 kDa)
For IHC/IF: Include isotype control antibodies at matching concentration
For flow cytometry: Include fluorescence minus one (FMO) controls
Tissue/sample validation:
Test in tissues known to express LAYN (e.g., liver cancer tissue, A549 cells, mouse brain tissue)
Test in negative control tissues with low/no LAYN expression
Following recent recommendations from antibody characterization initiatives like YCharOS and Only Good Antibodies (OGA), comprehensive validation should be conducted and documented before proceeding with experimental applications .
Optimizing immunoprecipitation (IP) with LAYN antibodies requires careful consideration of several factors:
Antibody selection and preparation:
Cell lysis optimization:
Use buffer containing 1% NP-40 or Triton X-100, 150 mM NaCl, 50 mM Tris pH 7.5, and protease/phosphatase inhibitors
For membrane proteins like LAYN, consider specialized membrane protein extraction kits
Perform lysis on ice and process samples quickly to preserve protein interactions
Co-IP protocol refinements:
To study LAYN interaction with integrins like LFA-1, use gentler crosslinking approaches (e.g., DSP or DTSSP crosslinkers at 0.5-2 mM)
Adjust salt concentration (125-150 mM) to optimize stringency
Incubate antibody-lysate mixture overnight at 4°C with gentle rotation
Controls and validation:
Analysis considerations:
Analyze by western blot using antibodies against suspected interaction partners
Consider mass spectrometry analysis for unbiased identification of interaction partners
This approach has been successfully used to demonstrate LAYN's colocalization and functional relationship with LFA-1, revealing its role in enhancing T cell adhesion and antitumor immunity .
Several technical challenges may arise when using LAYN antibodies for flow cytometry:
Solution: Optimize antibody concentration by titration (typically starting at 1:10-1:100 for IF/flow applications)
Approach: Prepare a dilution series (e.g., 1:10, 1:50, 1:100, 1:200) and determine optimal signal-to-noise ratio
Consideration: For expanded T cells, higher concentrations (25 μg/ml) have been used successfully
Solution: Improve blocking and washing steps
Approach: Use 5-10% serum from the same species as the secondary antibody; increase washing time/volume; use fluorescence minus one (FMO) controls
Consideration: For LAYN specifically, adjust compensation carefully as expression can overlap with other T cell markers
Solution: Include positive control samples with known LAYN expression
Approach: Use cell lines with confirmed LAYN expression such as HepG2 or A549 cells
Consideration: LAYN expression changes upon T cell activation, so standardize activation conditions
Solution: Optimize fixation and permeabilization protocols
Approach: Compare different fixatives (e.g., 2-4% PFA vs. methanol) and permeabilization reagents (e.g., 0.1-0.5% Triton X-100, 0.1-0.5% saponin)
Consideration: LAYN's association with integrins may mask epitopes; gentle permeabilization might be required
Troubleshooting tip: For studying LAYN in exhausted T cells, combine LAYN staining with other markers (CD8, PD-1, TCR clonality markers) to properly identify the relevant population, as LAYN is selectively expressed on clonally expanded T cells in tumors .
Based on recent findings about LAYN's role in integrin activation and T cell adhesion , a comprehensive experimental design should include:
Cell adhesion assays:
Static adhesion to ICAM-1: Coat plates with ICAM-1 (natural ligand for LFA-1) at 2-10 μg/ml
Comparison groups: Control vs. LAYN knockout/knockdown T cells (using CRISPR-Cas9 or siRNA)
Validation approach: Include LFA-1 blocking antibody condition to confirm specificity
Readout: Quantify adherent cells after centrifugal washing
Expected result: LAYN deletion should result in reduced LFA-1-dependent cellular adhesion
Dynamic adhesion under flow:
Setup: ICAM-1-coated flow chambers with controlled shear stress
Analysis: Compare rolling, firm adhesion, and crawling behaviors
Controls: Include PMA-activated cells as positive control for integrin activation
T cell migration assays:
Transwell migration: Assess migration toward chemokines (CXCL10, CXCL12)
3D collagen matrices: Evaluate velocity and directionality using time-lapse microscopy
In vivo migration: Adoptive transfer of differentially labeled control vs. LAYN-deficient T cells
Molecular mechanism studies:
Integrin activation: Measure activated LFA-1 using conformation-specific antibodies (e.g., KIM127, mAb24)
Proximity analysis: Use proximity ligation assays to quantify LAYN-LFA-1 colocalization
Signaling events: Analyze phosphorylation of adhesion-related proteins (FAK, Pyk2)
Functional outcome assessment:
This experimental approach has demonstrated that layilin colocalizes with LFA-1 and enhances T cell adhesion, which is critical for anti-tumor immunity despite the exhausted phenotype of LAYN-expressing T cells .
Selecting the appropriate LAYN antibody requires evaluation of several critical factors:
1. Application compatibility and validation:
Review the validated applications for each antibody (WB, IHC, IF, Flow, etc.)
Examine published validation data specific to your application of interest
Example: For Western blot, antibody 20535-1-AP has been validated at 1:1000-1:8000 dilution in A549 cells and mouse brain tissue
2. Epitope and domain recognition:
Consider which domain of LAYN your research focuses on
Some antibodies target the C-type lectin domain, while others target other regions
Example: Anti-human LAYN mAb clone PAT20G8AT is derived from immunization with recombinant human LAYN protein spanning amino acids 22-235
3. Species reactivity and cross-reactivity:
Ensure the antibody recognizes LAYN in your species of interest
Check for validated reactivity in the specific species (human, mouse, rat, etc.)
Example: Product 112159 reports reactivity with human and mouse samples
4. Clone type and reproducibility needs:
Polyclonal: Better for detection of low-abundance targets but may have batch variation
Monoclonal: Superior reproducibility but may be less sensitive
Recombinant antibodies: High consistency across lots
Example: ab192610 is a rabbit recombinant monoclonal, offering high reproducibility
5. Technical specifications and quality control:
Examine the antibody's production method and purification technique
Check for lot-specific validation data and consistent performance
Example: LAYN antibody from ProSpec is purified from mouse ascitic fluids by protein-A affinity chromatography
6. Antibody format and conjugation:
Consider whether you need unconjugated or directly conjugated antibodies
For multicolor flow cytometry, evaluate available fluorophore conjugates
Example: Most commercial LAYN antibodies are available unconjugated, requiring secondary detection
7. Independent validation:
Check for antibodies validated by independent initiatives like YCharOS
Review literature citations where the antibody has been successfully used
Example: Recent publications reporting substantial antibody characterization issues highlight the importance of independent validation
This systematic evaluation approach will help researchers select the most appropriate LAYN antibody for their specific experimental conditions, reducing reproducibility issues and improving data quality.
Interpreting LAYN expression patterns requires consideration of biological context and technical factors:
Biological interpretation framework:
Cancer type-specific patterns:
Immune cell population analysis:
CD8+ T cells: LAYN expression correlates with clonal expansion and exhaustion phenotype
Correlation with other markers: Analyze LAYN in relation to exhaustion markers (PD-1, TIM-3, LAG-3)
Functional state assessment: High LAYN expression is associated with diminished antitumor effects despite enhanced adhesion
Clinical correlation approach:
Analyze LAYN expression in relation to tumor infiltrating lymphocyte density
Correlate with patient survival data and response to immunotherapy
Stratify patients based on combined LAYN expression and immune signature analysis
Technical considerations for accurate interpretation:
Quantification methods:
For flow cytometry: Report percentage of positive cells and mean fluorescence intensity
For IHC/IF: Use digital image analysis with consistent thresholding
For transcriptomics: Apply appropriate normalization methods and consider isoform-specific analysis
Reference populations:
Compare LAYN expression to matched normal tissues
Use sorted T cell populations from peripheral blood as baseline
Include paired tumor-adjacent normal tissues when available
Integrated multi-omics approach:
Combine protein-level data (IHC, flow cytometry) with transcriptomic data
Use single-cell RNA-seq to dissect heterogeneity within LAYN+ populations
Correlate with spatial information using technologies like imaging mass cytometry
Recent studies using these approaches have revealed that LAYN serves as both a prognostic biomarker and a functional regulator of CD8+ T cell adhesion and tumor infiltration, with complex implications for antitumor immunity .
Distinguishing T cell exhaustion from activation using LAYN requires careful methodological considerations:
1. Temporal analysis and kinetics:
Approach: Perform time-course experiments following T cell activation
Method: Stimulate T cells with anti-CD3/CD28 beads and analyze LAYN expression at multiple timepoints (baseline, 24h, 48h, 72h, 7 days)
Interpretation: Transient LAYN upregulation may indicate activation, while persistent high expression correlates with exhaustion
2. Multi-parameter phenotyping:
Panel design: Include markers distinguishing activation (CD25, CD69, HLA-DR) from exhaustion (PD-1, TIM-3, TIGIT)
Analysis approach: Use high-dimensional analysis methods (tSNE, UMAP) to identify cell clusters
Key consideration: LAYN+ exhausted T cells maintain high CD69 expression, requiring additional markers for proper classification
3. Functional assessment:
Cytokine profiling: Measure polyfunctionality (IFN-γ, TNF-α, IL-2 production)
Proliferation capacity: Assess using Ki-67 staining or CFSE dilution
Killing capacity: Evaluate using tumor cell killing assays (e.g., CFSE/PI staining)
Interpretation: LAYN+ exhausted cells typically show reduced cytokine polyfunctionality despite maintained killing capacity
4. Molecular signaling analysis:
Approach: Examine TCR signaling components and their phosphorylation status
Key pathways: Assess NFAT vs. AP-1 balance (exhaustion vs. activation)
Integration: Correlate LAYN expression with signaling pathway activation
5. Contextual assessment:
Tissue source: LAYN expression patterns differ between peripheral blood, lymphoid tissues, and tumor sites
Antigen exposure: Consider chronic vs. acute antigen stimulation history
Environmental factors: Evaluate impact of cytokines (IL-2, IL-15, TGF-β) on LAYN expression
6. Clonality analysis:
Method: Combine LAYN staining with TCR sequencing or clonotype analysis
Significance: LAYN is preferentially expressed on clonally expanded T cells in tumors
Interpretation: High clonality with high LAYN suggests tumor-reactive but potentially exhausted T cells
This comprehensive approach has revealed that LAYN is selectively expressed on highly activated, clonally expanded, but phenotypically exhausted CD8+ T cells in human cancers, indicating its complex role in T cell biology .
Recent research has revealed crucial insights into LAYN's molecular function:
Key molecular mechanism discoveries:
LAYN-integrin colocalization:
Enhancement of integrin activation:
Functional impact on T cell-tumor interactions:
Technical approaches enabling these discoveries:
Static adhesion assays:
Control and LAYN-edited CD8+ T cells were compared in adhesion to ICAM-1-coated plates
Both in the presence and absence of T cell activation with PMA, LAYN-deleted cells displayed significantly reduced adhesion
LFA-1 blocking antibody abrogated all ICAM-1 binding, confirming LAYN's role in LFA-1-dependent adhesion
In vivo tumor models:
Therapeutic implications:
Potential for immunotherapy enhancement:
These findings represent a paradigm shift in understanding how "exhausted" or "dysfunctional" CD8+ T cells can maintain cytotoxic potential through enhanced cellular adhesiveness mediated by LAYN-integrin interactions .
LAYN antibodies show significant potential in several emerging therapeutic strategies:
1. Immune checkpoint modulation approaches:
Mechanism: Unlike traditional checkpoint blockade, LAYN targeting could enhance T cell adhesion while reversing exhaustion
Strategy: Develop antagonistic antibodies that block LAYN's inhibitory effects while preserving adhesion function
Preclinical evidence: LAYN antagonist treatment partially restored exhausted CD8+ T cell function
Combination therapy: LAYN targeting could complement PD-1/PD-L1 blockade by addressing different aspects of T cell dysfunction
2. Antibody-drug conjugates (ADCs):
Targeting strategy: LAYN is selectively expressed in tumors and exhausted TILs, offering specificity
Design considerations: Use non-depleting anti-LAYN antibodies conjugated to immunomodulatory payloads
Potential applications: Selective delivery of TLR agonists, STING activators, or other immunomodulators to the tumor microenvironment
Technical approach: Optimize antibody-payload ratios and linker chemistry for tumor-specific release
3. Bispecific antibody development:
Design concept: Create bispecific antibodies targeting both LAYN and tumor antigens
Functional mechanism: Enhance T cell-tumor cell adhesion through forced proximity
Technical approach: Use computational design methods similar to those for antibody nanocages
Advantage: Could redirect exhausted but tumor-specific T cells back to tumor cells
4. Engineered T cell therapies:
CAR-T enhancement: LAYN manipulation in CAR-T cells could improve tumor infiltration
Modification strategy: CRISPR/Cas9 editing to optimize LAYN expression levels
Rationale: Enhanced adhesion through LAYN could improve CAR-T persistence in solid tumors
Testing approach: Compare tumor infiltration and killing by LAYN-modified vs. standard CAR-T cells
5. Antibody nanocage architectures:
Design approach: Use modular antibody nanocage (AbC) technology described in recent research
Application: Create multivalent LAYN-targeting structures with precise geometry
Potential benefit: Controlled crosslinking of LAYN could optimize integrin activation
Technical consideration: Design proteins that assemble anti-LAYN antibodies into specific nanocage architectures
6. Diagnostic and patient stratification applications:
Approach: Develop standardized LAYN antibody-based assays for tissue and liquid biopsies
Clinical application: Identify patients with high LAYN expression as candidates for specific immunotherapies
Implementation: IHC, flow cytometry, or mass cytometry panels including LAYN
Integration: Combine with other immune profiling markers for comprehensive assessment
These approaches leverage recent discoveries about LAYN's role in T cell function and tumor biology, offering promising new directions for cancer immunotherapy research and development .
In light of the "antibody characterization crisis" affecting biomedical research , implementing rigorous quality control for LAYN antibodies is critical:
1. Antibody sourcing and documentation:
RRID verification: Ensure the antibody has a unique Research Resource Identifier (RRID) for tracking and reproducibility
Vendor evaluation: Select vendors that provide comprehensive characterization data and lot-specific quality control
Documentation requirements: Record complete antibody information (catalog number, lot number, clone, host species, isotype)
Best practice: Create a standardized antibody metadata collection form for your laboratory
2. Application-specific validation:
3. Cross-platform verification:
Approach: Verify LAYN expression or function using orthogonal methods
Example: Combine antibody-based detection with mRNA quantification
Implementation: Compare LAYN protein levels (WB/IHC) with mRNA expression (RT-qPCR)
4. Benchmarking against literature standards:
Literature comparison: Test antibody performance against published results
Reference samples: Use established cell lines with known LAYN expression (A549, HepG2, NCI-H1299)
Control tissues: Human liver cancer tissue and mouse brain tissue show positive LAYN expression
5. Titration and optimization protocols:
Dilution series: Test multiple concentrations to determine optimal signal-to-noise ratio
Recommended starting ranges:
Documentation: Create detailed protocol records including all optimization steps
6. Reproducibility assessment:
Batch testing: Test multiple antibody lots before large-scale studies
Inter-operator validation: Have multiple researchers perform the same protocol
Long-term monitoring: Implement antibody performance tracking over time
7. Specialized validation for T cell research:
T cell activation controls: Compare resting vs. activated T cells (expect higher LAYN in activated state)
Exhaustion model validation: Test antibody in well-characterized T cell exhaustion models
Functional correlation: Validate that LAYN detection correlates with expected functional outcomes
By implementing these quality control measures, researchers can significantly improve the reproducibility and reliability of LAYN antibody-based experiments, addressing the broader concerns about antibody quality in biomedical research .
Several cutting-edge technologies show promise for advancing LAYN research:
1. Advanced single-cell and spatial biology approaches:
Single-cell proteomics: Mass cytometry (CyTOF) and spectral flow cytometry for high-parameter analysis of LAYN alongside >40 other markers
Spatial proteomics: CODEX, Imaging Mass Cytometry, or GeoMx DSP to analyze LAYN expression within the spatial context of the tumor microenvironment
Spatial transcriptomics: Visium or MERFISH to correlate LAYN mRNA with spatial distribution of different cell types
Research application: These technologies would reveal how LAYN+ cells position themselves relative to tumor cells and other immune populations
2. Protein-protein interaction mapping technologies:
Proximity labeling: BioID or TurboID fusion proteins to identify novel LAYN interaction partners
Live-cell protein interaction imaging: FRET, BiFC, or FLIM to visualize LAYN-integrin interactions in real time
Interactome analysis: AP-MS combined with cross-linking to capture transient LAYN interactions
Research application: These approaches could reveal the complete molecular complex formed by LAYN and integrins during T cell adhesion
3. Advanced genetic manipulation techniques:
Base editing and prime editing: More precise genetic manipulation of LAYN with fewer off-target effects
Inducible expression systems: Temporal control of LAYN expression using optogenetics or chemical-induction
Domain-specific CRISPR interference: Targeted disruption of specific LAYN functional domains
Research application: These tools would allow precise dissection of LAYN domain functions in T cell biology
4. Intravital imaging technologies:
Multiphoton intravital microscopy: Real-time visualization of LAYN+ T cells in tumor microenvironments
Optogenetic manipulation: Light-controlled activation/inhibition of LAYN during imaging
Reporter mouse models: Development of LAYN-reporter mice for tracking expression dynamics
Research application: These approaches would reveal how LAYN affects T cell migration, interaction with tumor cells, and retention within tumors
5. High-throughput antibody engineering platforms:
Active learning for antibody optimization: Using machine learning approaches as described in recent research
Library-on-library screening: Testing many antibody variants against many LAYN epitopes
Absolut! simulation framework: Computational prediction of antibody-antigen binding
Research application: These methods could generate more specific and versatile anti-LAYN antibodies for research and therapeutic applications
6. Organoid and advanced 3D culture systems:
Tumor-immune co-culture organoids: Study LAYN+ T cells in physiologically relevant 3D environments
Microfluidic organ-on-chip: Model LAYN's role in T cell trafficking through tissues
Patient-derived organoids: Test LAYN-targeting approaches in personalized models
Research application: These systems would bridge the gap between in vitro and in vivo studies, providing more translatable insights