LGALS10 regulates immune responses through carbohydrate binding and interactions with eosinophils, basophils, and Treg cells .
Carbohydrate Binding: Binds mannose and other glycoconjugates via its CRD, though with distinct specificity compared to other galectins .
Immune Suppression: Essential for the anergy and suppressive functions of CD25+ Treg cells .
Eosinophil Activity: Forms Charcot-Leyden crystals (CLCs) during eosinophil lysis, linked to inflammatory diseases .
LGALS10 has emerged as a potential biomarker in oncology, particularly in ovarian cancer.
Mechanistic Insight: LGALS10 may act as a tumor suppressor by modulating immune microenvironments, though its precise role requires further investigation .
Recombinant LGALS10 and specific antibodies enable functional studies.
Attribute | Detail |
---|---|
Sequence | Full-length human LGALS10 (1–162 aa with His-tag) . |
Applications | SDS-PAGE, mass spectrometry, binding assays . |
Storage | -20°C to -80°C (long-term); +2°C to +8°C (short-term) . |
Dimerization: Forms homo-dimers via face-to-face interactions, creating a binding site for monosaccharides .
Monosaccharide Binding: d-Glucose, d-allose, and D-N-acetylgalactosamine bind at dimer interfaces, influencing CLC stability .
Crystal Formation: Monosaccharides may inhibit CLC formation by disrupting dimer packing .
LGALS10 is predominantly expressed in eosinophils, basophils, and Treg cells.
Eosinophils: Stored in peripheral cytoplasm near the plasma membrane, not in granules .
Release: Released during eosinophil activation, contributing to CLC formation .
Mechanistic Studies: Elucidate how LGALS10 modulates Treg cell function and immune evasion in cancer.
Therapeutic Potential: Explore LGALS10 as a biomarker or target for eosinophilic disorders and ovarian cancer.
Species-Specificity: Investigate the absence of LGALS10 in rodents to identify alternative models for human studies .
Human LGALS10 (Galectin-10) belongs to the prototype galectin subfamily but exhibits a novel dimerization structure that distinguishes it from other prototype galectins such as Galectin-1, -2, and -7. Crystallographic studies at 1.55–2.00 Å resolution reveal that LGALS10 forms a unique dimer with a distinctive global shape . In the LGALS10 dimer, Glu33 from one subunit modifies the carbohydrate-binding site of the other subunit, which essentially inhibits disaccharide binding . This structural arrangement is not observed in other prototype galectins and contributes to LGALS10's unique functional properties.
Human LGALS10 is 142 amino acids in length with one Galectin domain (amino acids 6-138) containing two dimerization motifs (amino acids 6-10 and 131-135) . Two molecular weight isoforms of 15 and 14 kDa have been described, though these variants have not been fully characterized . Importantly, unlike other widely conserved galectins, there is no known structural rodent counterpart to human LGALS10, which creates challenges for preclinical research .
LGALS10 has a remarkably restricted expression pattern. It is predominantly and abundantly expressed in human eosinophils, where it is one of the most abundant cytoplasmic proteins . Besides eosinophils, LGALS10 is also expressed in basophils and CD25+ regulatory T cells (Tregs) . Some myeloid leukemia cells have also been found to express LGALS10, but otherwise, its expression is highly cell-type specific .
For detection, researchers can employ several methodologies:
Western blot analysis: Using specific antibodies such as Goat Anti-Human Galectin-10 Antigen Affinity-purified Polyclonal Antibody, LGALS10 can be detected at approximately 16 kDa under reducing conditions .
Flow cytometry: Intracellular staining of cells fixed and permeabilized with appropriate buffers allows detection of LGALS10 in specific cell populations. This is particularly useful for Treg analysis when combined with surface markers like CD25 .
PCR detection: Quantitative real-time PCR can be performed using specific primers:
Immunohistochemistry: LGALS10 can be detected in tissue sections using specific antibodies (typically at 1:100 dilution) with DAB staining and hematoxylin counterstaining .
LGALS10 forms Charcot-Leyden crystals (CLCs) in vivo, which are characteristic bipyramidal crystals observed in various eosinophilic diseases . These crystals have long been recognized as hallmarks of eosinophilic inflammation in tissues and body fluids. The formation of CLCs is linked to the release of LGALS10 through a specific cell death program called extracellular trap cell death (ETosis) .
During ETosis, eosinophils rapidly disintegrate their plasma membranes, releasing the majority of cytoplasmic LGALS10, which can subsequently form CLCs in the extracellular environment . Therefore, the presence of CLCs in tissue samples or body fluids indicates not only eosinophilic inflammation but specifically suggests active ETosis of eosinophils at the site of inflammation.
Current research indicates that LGALS10/CLCs are not merely passive markers of eosinophilic inflammation but may play functional roles in immunity and disease pathogenesis . This represents an important shift in understanding, as these structures were previously viewed as simply diagnostic indicators.
The underlying mechanisms for LGALS10's favorable prognostic impact may relate to:
Differential expression patterns: Studies have shown that LGALS10 mRNA and protein expression are decreased in ovarian cancer cells compared to normal ovarian cells (P<0.05) . This suggests that loss of LGALS10 may contribute to cancer development or progression.
Subtype specificity: LGALS10 predicts a longer OS specifically in females with serous ovarian cancer across all stages, suggesting that its function may interact with the unique biology of this histological subtype .
Immune modulation: Given LGALS10's expression in regulatory T cells and its role in eosinophil function, it may influence tumor immunity, potentially enhancing anti-tumor immune responses when present at higher levels.
Researchers investigating LGALS10 in ovarian cancer should consider multivariate analysis to determine if its prognostic value is independent of established clinical factors. Additionally, functional studies examining how LGALS10 affects ovarian cancer cell proliferation, migration, and immune evasion would provide mechanistic insights.
LGALS10 exhibits atypical saccharide binding properties compared to other galectins. Structural and biochemical studies have revealed several unique features:
Inhibition of disaccharide binding: In the LGALS10 dimer, Glu33 from one subunit modifies the carbohydrate-binding site of another, inhibiting disaccharide binding .
Interaction with small hydroxylated molecules: Despite limited disaccharide binding, LGALS10 can interact with glycerol and potentially other small hydroxylated molecules. His53 appears to be crucial for this binding .
Negative regulation by Trp72: Surprisingly, alanine substitution of the conserved Trp72 (which is crucial for saccharide binding in other galectins) actually enhances erythrocyte agglutination, suggesting that Trp72 negatively regulates LGALS10 ligand binding .
To experimentally test hypotheses regarding LGALS10's binding properties, researchers could:
Site-directed mutagenesis: Create mutants targeting key residues (His53, Trp72, Glu33) to assess their impact on binding properties.
Glycan array screening: Utilize glycan arrays to comprehensively profile LGALS10's binding specificity to various glycans and identify preferred ligands.
Surface plasmon resonance (SPR): Determine binding kinetics and affinities for potential ligands, comparing wild-type LGALS10 with mutant variants.
X-ray crystallography: Solve co-crystal structures with various ligands to visualize binding modes at atomic resolution.
Cellular assays: Assess how mutations affect LGALS10's functional properties in cell-based systems, such as agglutination assays or cell adhesion studies.
These approaches would provide insights into the structural basis of LGALS10's unique binding properties and potentially identify physiologically relevant ligands.
The release of LGALS10 from eosinophils primarily occurs through extracellular trap cell death (ETosis) rather than conventional secretory processes like piecemeal degranulation or exocytosis . During ETosis, eosinophils rapidly disintegrate their plasma membranes, releasing the majority of cytoplasmic LGALS10 into the extracellular environment .
To study and detect ETosis-mediated LGALS10 release, researchers can employ several methodologies:
Immunofluorescence microscopy: Visualization of extracellular DNA fibers (using DNA stains) co-localized with LGALS10 immunostaining provides direct evidence of ETosis. The characteristic web-like structures of extracellular traps containing LGALS10 can be quantified.
Flow cytometry: Detection of cells undergoing ETosis through the combination of vital dyes (indicating membrane integrity loss), DNA stains, and LGALS10 antibodies.
ELISA for soluble LGALS10: Quantification of LGALS10 levels in cell supernatants or biological fluids (serum, bronchoalveolar lavage fluid) as a surrogate marker for ETosis.
Live cell imaging: Real-time visualization of the ETosis process using fluorescently labeled LGALS10 or antibodies against LGALS10.
Electron microscopy: Ultrastructural analysis of eosinophils undergoing ETosis and LGALS10 release.
Understanding the regulatory mechanisms of ETosis-mediated LGALS10 release is crucial, as elevated LGALS10 levels in serum and tissue suggest a high degree of eosinophil ETosis, which may serve as a biomarker for disease activity and treatment effectiveness in various eosinophilic conditions .
Detecting LGALS10 requires different approaches depending on the sample type. Here are optimized protocols for various applications:
Sample preparation:
For cell lines (e.g., HL-60): Lyse cells in RIPA buffer with protease inhibitors
For tissue samples: Homogenize in tissue lysis buffer with protease inhibitors
Electrophoresis conditions:
Use 12-15% SDS-PAGE gels
Load 20-30 μg protein per lane
Run under reducing conditions
Transfer and detection:
Transfer to PVDF membrane
Block with 5% non-fat milk in TBST
Probe with anti-LGALS10 antibody (1 μg/mL)
Use appropriate HRP-conjugated secondary antibody
Develop using ECL detection system
Cell preparation:
Isolate cells (PBMCs, eosinophils, or cultured cells)
Surface stain for relevant markers (e.g., CD25 for Tregs)
Fixation and permeabilization:
Fix cells with 4% paraformaldehyde for 15 minutes
Permeabilize with a suitable buffer (e.g., FlowX FoxP3/Transcription Factor Fixation & Perm Buffer)
Staining:
Incubate with anti-LGALS10 antibody (optimal concentration determined empirically)
Add fluorophore-conjugated secondary antibody
Wash and analyze by flow cytometry
Controls: Include isotype control antibody to assess non-specific binding
Tissue processing:
Fix tissue in formalin and embed in paraffin
Section at 4-5 μm thickness
Antigen retrieval:
Deparaffinize and rehydrate sections
Perform heat-induced epitope retrieval (citrate buffer pH 6.0)
Staining procedure:
Block endogenous peroxidase with 3% H₂O₂
Block non-specific binding with serum
Incubate with anti-LGALS10 antibody (1:100 dilution) at 4°C overnight
Apply secondary antibody
Develop with DAB and counterstain with hematoxylin
Scoring system: Semi-quantitative assessment based on staining intensity and percentage of positive cells
Accurate quantification of LGALS10 expression is essential for comparative studies. Here are methodological approaches for both mRNA and protein quantification:
Real-time quantitative PCR (RT-qPCR):
Extract total RNA using standard methods (TRIzol or column-based)
Synthesize cDNA using reverse transcriptase
Perform qPCR with LGALS10-specific primers:
Use GAPDH as reference gene:
Calculate relative expression using the 2^(-ΔΔCt) method
Digital droplet PCR (ddPCR):
For absolute quantification without standard curves
Particularly useful for low-abundance transcript detection
Use the same primers as for RT-qPCR
RNA-Seq:
For transcriptome-wide analysis with LGALS10 expression as part of the dataset
Allows identification of correlations with other genes and potential regulatory networks
Western blot with densitometry:
Perform western blot as described in section 3.1.1
Use β-actin or GAPDH as loading control
Quantify band intensity using image analysis software
Express results as relative density normalized to loading control
ELISA:
For quantification in serum, plasma, or cell culture supernatants
Either commercial kits or laboratory-developed assays
Standard curve using recombinant LGALS10 for absolute quantification
Mass spectrometry-based proteomics:
For global proteome analysis with LGALS10 quantification
Label-free or isotope-labeled approaches
Provides absolute quantification and identification of post-translational modifications
Standardization: Use the same methodology across all samples being compared
Technical replicates: Minimum of three replicates to assess variability
Biological replicates: Analyze multiple independent samples
Statistical analysis: Appropriate tests (t-test, ANOVA) with multiple testing correction
Validation: Confirm key findings using an alternative method
Human eosinophilic cell lines:
HL-60 cells differentiated toward eosinophilic lineage
AML14.3D10 cells (eosinophilic cell line)
Primary human eosinophils isolated from peripheral blood
LGALS10 overexpression/knockdown systems:
Lentiviral transduction for stable expression in various cell types
siRNA or CRISPR-Cas9 for gene silencing in cells that naturally express LGALS10
Inducible expression systems for temporal control
3D culture systems:
Organoids from tissues affected by eosinophilic diseases
Co-culture systems with eosinophils and target tissue cells
Human tissue explants:
Short-term culture of tissue biopsies from patients with eosinophilic diseases
Treatment with recombinant LGALS10 or neutralizing antibodies
Precision-cut tissue slices:
Maintain 3D architecture and cellular diversity
Useful for studying LGALS10 in tissue context
Humanized mouse models:
Mice engrafted with human immune cells including eosinophils
Allows study of human LGALS10 in a living organism
Transgenic expression models:
Generate mice expressing human LGALS10 under tissue-specific promoters
Study effects of human LGALS10 in specific compartments
Non-human primates:
Potential for studying LGALS10 in species with closer homology to humans
Limited by ethical and practical considerations
Structure-based functional prediction:
Use solved crystal structures to predict functional interactions
Molecular dynamics simulations to study protein behavior
Systems biology:
Integrate -omics data to predict LGALS10 functions in different contexts
Network analysis to identify functional associations
Each model system has advantages and limitations, and researchers should select the appropriate model based on their specific research questions and available resources.
LGALS10 has emerged as a promising biomarker for various eosinophilic diseases, including asthma, eosinophilic esophagitis, rhinitis, sinusitis, atopic dermatitis, and eosinophilic granulomatosis with polyangiitis . Its utility spans disease activity assessment, diagnosis, and monitoring treatment effectiveness.
Disease | LGALS10 Biomarker Applications | Key Advantages |
---|---|---|
Asthma | Disease activity monitoring, Treatment response prediction | More specific than general eosinophil counts |
Eosinophilic Esophagitis | Diagnosis, Treatment monitoring | Less invasive than repeated biopsies when measured in blood |
Rhinitis/Sinusitis | Disease severity assessment | Correlates with symptom scores |
Atopic Dermatitis | Disease activity monitoring | Reflects eosinophil ETosis in tissues |
Eosinophilic Granulomatosis with Polyangiitis | Diagnosis, Relapse prediction | Complements ANCA testing |
Serum/Plasma LGALS10:
ELISA-based quantification
Reference ranges need to be established for different patient populations
Sample handling critical: immediate processing or storage at -80°C
Tissue LGALS10 Detection:
Immunohistochemistry for formalin-fixed paraffin-embedded (FFPE) tissue
Dual staining with eosinophil markers (e.g., major basic protein)
Digital pathology for quantitative assessment
Charcot-Leyden Crystal Detection:
Microscopic evaluation of respiratory samples (sputum, nasal lavage)
Wright-Giemsa or H&E staining
Confirmation with LGALS10 immunostaining
Non-invasive Sampling:
Induced sputum for respiratory diseases
Nasal lavage fluid for upper airway disorders
Tears for ocular allergic disorders
Pre-analytical factors:
Standardize collection procedures
Define acceptable time intervals between collection and processing
Establish stability data for different storage conditions
Analytical validation:
Precision (intra- and inter-assay variability <10%)
Accuracy (recovery 90-110%)
Linearity within the clinical measurement range
Detection limits appropriate for disease states
Clinical validation:
Establish reference intervals in healthy individuals
Determine decision thresholds for specific clinical applications
Assess confounding factors (medications, comorbidities)
Research has begun to elucidate the prognostic significance of LGALS10 in various cancers, with the most substantive evidence in ovarian cancer. Understanding its performance relative to established biomarkers is essential for potential clinical implementation.
Ovarian Cancer Subgroup | Hazard Ratio (95% CI) | P-value | Prognostic Impact |
---|---|---|---|
All patients | 0.82 (0.71-0.95) | 0.0065 | Favorable |
Serous subtype | Significant* | <0.05 | Favorable |
All stages | Significant* | <0.05 | Favorable |
Grade III | Significant* | <0.05 | Favorable |
*Exact hazard ratios not provided in the source material
Retrospective cohort analysis:
Tissue microarray analysis of archival samples
Correlation with long-term clinical outcomes
Multivariate regression models adjusting for clinicopathological factors
Prospective validation:
Include LGALS10 measurement in clinical trials
Pre-specified analysis of prognostic value
Time-dependent receiver operating characteristic (ROC) analysis
Integration with molecular profiling:
Combine with genomic signatures
Pathway analysis to understand biological context
Machine learning approaches for integrated biomarker models
The translation of LGALS10 detection from research to clinical applications faces several technical challenges that must be addressed to ensure reliability and reproducibility:
Sample collection and processing:
Blood collection tubes (EDTA vs. heparin vs. serum)
Processing delays affect LGALS10 stability
Cell lysis during processing may release intracellular LGALS10
Tissue handling:
Fixation protocols affect epitope availability
Cold ischemia time impacts protein integrity
Decalcification procedures for bone marrow samples
Antibody variability:
Different commercial antibodies show variable specificity and sensitivity
Lot-to-lot variability affects quantitative measurements
Need for reference standards and calibrators
Assay platforms:
ELISA vs. multiplex vs. automated immunoassay platforms
Need for platform-specific reference ranges
Inter-laboratory harmonization
Cut-off determination:
Disease-specific thresholds required
Population-specific reference ranges
Statistical approaches for optimal cut-point selection
Result interpretation:
Context-specific interpretation (disease type, treatment status)
Integration with other laboratory and clinical findings
Longitudinal monitoring protocols
Quality assurance:
Internal quality control materials
External quality assessment programs
Proficiency testing
Reference materials development:
Purified recombinant LGALS10 as primary calibrator
Stabilized cell lysates as secondary standards
Certified reference materials through international collaboration
Assay standardization initiatives:
International working groups for method harmonization
Standard operating procedures for pre-analytical variables
Clinical Laboratory Standards Institute (CLSI) guidelines development
Digital pathology approaches:
Automated image analysis algorithms for tissue LGALS10 quantification
Machine learning for standardized interpretation
Virtual microscopy for centralized expert review
Given the emerging understanding of LGALS10's functions, several potential therapeutic applications warrant exploration:
Anti-LGALS10 antibodies:
Neutralization of extracellular LGALS10
Prevention of Charcot-Leyden crystal formation
Disruption of eosinophil-epithelial interactions
Small molecule inhibitors:
Target the unique LGALS10 carbohydrate-binding site
Exploit the distinct Glu33-mediated binding inhibition
Design based on His53 interaction with small hydroxylated molecules
ETosis modulation:
Inhibit pathways leading to LGALS10 release via ETosis
Targeted degradation of extracellular LGALS10
Enhancement of Charcot-Leyden crystal resolution
LGALS10 induction therapy:
Based on favorable prognostic impact in ovarian cancer
Epigenetic modifiers to restore LGALS10 expression
Differentiation therapy to enhance LGALS10 in cancer cells
Immune modulation:
Targeting LGALS10 in regulatory T cells
Enhancing anti-tumor immunity
Combination with immune checkpoint inhibitors
Biomarker-guided therapy:
LGALS10 expression as predictor of response
Treatment stratification based on LGALS10 levels
Monitoring therapy effectiveness through LGALS10 dynamics
Targeted nanoparticles:
Eosinophil-targeted delivery of LGALS10 modulators
Tumor-specific delivery systems
Inhalational formulations for respiratory applications
Gene therapy:
CRISPR-based modulation of LGALS10 expression
AAV-mediated delivery to specific tissues
mRNA therapeutics for transient LGALS10 modulation
Advanced -omics technologies offer powerful approaches to deepen our understanding of LGALS10:
Single-cell RNA sequencing:
Cell-specific expression patterns in health and disease
Identification of previously unknown LGALS10-expressing cell populations
Trajectory analysis for developmental regulation
Epigenetic profiling:
DNA methylation analysis of the LGALS10 promoter
Histone modification patterns affecting expression
Chromatin accessibility studies (ATAC-seq)
Genome-wide association studies (GWAS):
Identification of genetic variants influencing LGALS10 expression
Population-specific differences in regulation
Disease associations with LGALS10 polymorphisms
Interactome analysis:
Proximity labeling techniques (BioID, APEX) to identify LGALS10 protein partners
Affinity purification-mass spectrometry
Protein-protein interaction networks in different cellular contexts
Post-translational modifications:
Comprehensive PTM mapping (phosphorylation, glycosylation, etc.)
Functional consequences of modifications
PTM dynamics during cellular activation and disease states
Spatial proteomics:
Subcellular localization in different cell types
Translocation during activation and ETosis
Imaging mass cytometry for tissue context
Data integration approaches:
Combined genomic, transcriptomic, and proteomic analysis
Network biology to identify regulatory hubs
Causal inference methods for mechanistic insights
Temporal dynamics:
Time-course experiments during cell activation
Disease progression analysis
Treatment response monitoring
Computational modeling:
Machine learning for pattern recognition
Pathway enrichment analysis
Systems biology approaches to predict functional impacts
Despite important insights from the crystal structure of LGALS10, several structural biology techniques could provide additional critical information:
Cryo-electron microscopy (Cryo-EM):
Visualization of LGALS10 in different conformational states
LGALS10 interactions with binding partners
Structure of Charcot-Leyden crystals at near-atomic resolution
Nuclear magnetic resonance (NMR) spectroscopy:
Solution structure dynamics
Ligand binding studies
Protein-protein interaction interfaces
Hydrogen-deuterium exchange mass spectrometry (HDX-MS):
Conformational dynamics in solution
Effects of ligand binding on protein flexibility
Allosteric regulation mechanisms
Molecular dynamics simulations:
Conformational sampling of LGALS10
Binding pocket dynamics
Effects of mutations on structure and function
Virtual screening and docking:
Identification of novel LGALS10 binders
Structure-based drug design
Exploitation of unique binding properties
Quantum mechanics/molecular mechanics (QM/MM):
Detailed analysis of binding site interactions
Reaction mechanisms
Electronic properties relevant to function
Co-crystallization studies:
LGALS10 with physiological ligands
Complexes with binding partners
Drug candidates targeting LGALS10
Small-angle X-ray scattering (SAXS):
Low-resolution shape of LGALS10 complexes in solution
Oligomerization states
Conformational ensembles
Cross-linking mass spectrometry:
Mapping interaction surfaces
Validation of computational models
Analysis of multi-protein complexes
The Charcot-Leyden Crystal Protein (CLC-P), also known as Galectin-10, is a protein predominantly found in human eosinophils and basophils . It is named after the distinctive Charcot-Leyden crystals, which are hexagonal bipyramidal structures formed by the crystallization of this protein . These crystals are often observed in tissues and secretions associated with eosinophilic inflammation, such as in conditions like asthma, allergic reactions, and parasitic infections .
The discovery of Charcot-Leyden crystals dates back to the mid-19th century. Friedrich Albert von Zenker first noticed these crystals in 1851, and they were later described by Jean-Martin Charcot and Charles-Philippe Robin in 1853 . Ernst Viktor von Leyden further studied these crystals in 1872, contributing to the understanding of their association with eosinophilic diseases .
CLC-P is a lysophospholipase, an enzyme that hydrolyzes lysophospholipids to release free fatty acids . It has been shown to possess lysophospholipase activity, releasing palmitate from lysopalmitoylphosphatidylcholine . Additionally, CLC-P functions as an acyl-protein thioesterase (APT), which catalyzes the removal of thioester-linked fatty acids from cysteine residues of proteins . This activity is crucial for the regulation of protein palmitoylation, a post-translational modification that affects membrane localization, vesicular transport, and secretion .
Charcot-Leyden crystals are indicative of eosinophilic inflammation and are commonly found in conditions such as asthma, bronchitis, allergic rhinitis, and parasitic infections . The presence of these crystals in tissues and secretions serves as a marker for eosinophil-rich inflammatory conditions . Recent studies have shown that CLC-P can activate the NLRP3 inflammasome, leading to IL-1β-driven inflammation . This highlights the potential role of CLC-P in promoting pro-inflammatory activities in eosinophilic disorders.
Human recombinant CLC-P is produced using recombinant DNA technology, which involves inserting the gene encoding CLC-P into a suitable expression system, such as bacteria or yeast, to produce the protein in large quantities. This recombinant protein is used in research to study the biochemical properties and functions of CLC-P, as well as its role in eosinophilic inflammation and related diseases .