LGALS9 (Galectin 9) is a protein-coding gene in humans that produces galectin-9, a 36 kDa beta-galactoside-binding lectin involved in immune regulation, cancer biology, and metabolic processes . First isolated from mouse embryonic kidney in 1997, human galectin-9 is encoded by the LGALS9 gene located on chromosome 17 . It plays dual roles in tumorigenesis and immune modulation, making it a critical focus of biomedical research.
Galectin-9 contains two carbohydrate-recognition domains (CRDs) connected by a linker peptide, enabling interactions with glycosylated proteins . Alternative splicing generates multiple isoforms with distinct functional properties:
T-cell apoptosis: Galectin-9 binds TIM-3 on Th1 cells, inducing caspase-dependent apoptosis .
Checkpoint modulation: Interacts with PD-1 and VISTA, suppressing T-cell activation and granzyme B release .
Innate immunity: Regulates AMPK during lysosomal damage, linking metabolic stress to immune responses .
Pro-tumor effects: Promotes immune escape via TIM-3/Gal-9 interaction in Hodgkin’s lymphoma, AML, and lung cancer .
Anti-tumor effects: Inhibits metastasis by enhancing endothelial adhesion in hepatocellular carcinoma .
Obesity: Lgals9 deficiency in mice reduces diet-induced obesity and improves glucose tolerance .
Oxidative stress: Binds peroxiredoxin-2 (PRDX2), shifting its redox state to reduce oxidative damage .
Galectin-9-TIM-3 Axis in Cancer Immunotherapy
Metabolic Effects in Obesity
VISTA Interaction in Immune Suppression
Galectin-9 is ubiquitously expressed but enriched in immune and epithelial tissues :
Human Galectin-9 (LGALS9) is a 36-kDa tandem repeat galectin containing two carbohydrate recognition domains joined by a linker peptide. These domains exhibit high affinity for β-galactoside residues. The protein can be detected in various cellular compartments including the cytoplasm, nucleus, and cell surface depending on the cell type and physiological conditions .
Methodological approach for structural studies:
X-ray crystallography and NMR spectroscopy remain the gold standards for determining LGALS9 structure
Computational modeling using homology-based approaches can supplement experimental data
Recombinant expression systems (typically E. coli or mammalian cells) should be optimized for proper folding of both carbohydrate recognition domains
LGALS9 interacts with multiple receptors across immune and cancer cells, with T-cell immunoglobulin and mucin-domain containing-3 (TIM-3) being the most extensively characterized . This Gal-9/TIM-3 pathway is functional in numerous human cancer cell types and plays a critical role in immune regulation .
Receptor | Primary Cell Types | Binding Domain | Functional Outcome |
---|---|---|---|
TIM-3 | T cells, macrophages, dendritic cells | N-terminal CRD | Immunosuppression, apoptosis |
CD44 | Various cancer cells, immune cells | C-terminal CRD | Cell adhesion, metastasis |
Dectin-1 | Myeloid cells | Both CRDs | Innate immune modulation |
CD45 | Leukocytes | Variable | Phosphatase regulation |
Methodological approaches for studying LGALS9 regulation:
Quantitative PCR for transcript analysis
Western blotting and flow cytometry for protein expression analysis
Immunohistochemistry for tissue localization studies
Use of transcription factor binding site analysis and promoter studies to identify regulatory elements
Multiple complementary approaches should be used for comprehensive LGALS9 detection:
Immunohistochemistry (IHC): For LGALS9 detection in formalin-fixed paraffin-embedded tissues, optimize antibody concentration and antigen retrieval methods. In pancreatic tissues, LGALS9 staining may localize differently in normal versus cancerous tissue. In pancreatic intraepithelial neoplasia (PanIN), strong LGALS9 staining is observed at the apex and nucleus of cells, with diffuse cytoplasmic staining of lower intensity .
Flow Cytometry: For quantitative analysis of LGALS9 expression in single cell suspensions. This technique allows simultaneous analysis of LGALS9 expression in both immune (CD45+) and non-immune (CD45-) cell populations. Results should be reported as relative fluorescence intensity (RFI) in relation to isotype controls .
RT-qPCR: For transcript level analysis, which can detect changes in expression before protein changes become apparent.
Subcellular fractionation combined with Western blotting or imaging techniques provides the most accurate assessment of LGALS9 distribution:
Subcellular Fractionation Protocol:
Separate nuclear, cytoplasmic, membrane, and secreted fractions
Confirm fraction purity using compartment-specific markers
Quantify LGALS9 in each fraction by Western blot or ELISA
Confocal Microscopy Approach:
Use fluorescently labeled antibodies against LGALS9
Co-stain with organelle-specific markers
Perform z-stack imaging for complete cellular distribution
In experimental models such as pancreatic cancer, LGALS9 localizes differently depending on the cell type. In PanIN lesions, strong nuclear and apical staining is observed, while in immune cells, the distribution pattern may differ significantly .
LGALS9 plays a critical role in pancreatic cancer as both a biomarker and functional contributor to disease progression:
Expression Profile: Among multiple solid tumors, pancreatic ductal adenocarcinoma (PDAC) demonstrates the highest LGALS9 expression, with mRNA levels substantially exceeding those of PD-L1 .
Prognostic Value: High LGALS9 expression correlates with poor prognosis in PDAC and several other cancer types including renal cell carcinoma, acute myeloid leukemia, and gastric carcinoma .
Immunosuppressive Functions: LGALS9 contributes to the immunosuppressive tumor microenvironment through:
Experimental approaches for studying LGALS9 in pancreatic cancer include the use of genetically modified mouse models (GEMMs) such as the Pdx1-Cre; LstopL-KrasG12D (KC) model, which recapitulates the progression from early pancreatic intraepithelial neoplasia (PanIN) to adenocarcinoma .
LGALS9 mediates tumor immune evasion through multiple mechanisms affecting both innate and adaptive immunity:
Regulatory T Cell Modulation: Analysis of Treg levels in KC mouse models showed significant increases in both circulating and tumor-infiltrating Tregs compared to wild-type mice, regardless of age. This indicates that Treg circulation and infiltration represent early and sustained events in pancreatic cancer development .
T Cell Exhaustion: Through interaction with TIM-3 on effector T cells, LGALS9 can induce T cell exhaustion and apoptosis.
Innate Immune Suppression: LGALS9 influences myeloid cell differentiation and function, promoting immunosuppressive phenotypes.
Methodological approaches for studying these mechanisms include:
Flow cytometry analysis of immune cell populations in peripheral blood and tumor tissues
Functional assays measuring T cell activation and cytokine production
In vivo depletion or blocking studies to assess the contribution of specific immune cell populations
The selection of appropriate preclinical models is critical for LGALS9 research:
Genetically Engineered Mouse Models (GEMMs):
The Pdx1-Cre; LstopL-KrasG12D (KC) model is considered optimal for studying early pancreatic pathology, as it develops the full spectrum of tumor progression from acinar to ductal metaplasia (ADM) through preneoplastic lesions (PanIN) to adenocarcinoma .
This model is particularly valuable as the KrasG12D mutation is found in 75%-95% of human pancreatic cancers and in precancerous PanIN lesions .
The model recapitulates human disease in terms of activated signaling pathways and immune response establishment, including Treg accumulation .
Patient-Derived Xenografts (PDXs):
Maintain tumor heterogeneity more effectively than cell lines
Must be used in immunocompromised mice, limiting studies of immune interactions
Can be used to validate findings from GEMMs in human tumor tissues
Organoid Models:
Allow examination of LGALS9 expression and function in a 3D context
Can be derived from both normal and tumor tissues
Permit genetic manipulation via CRISPR/Cas9 to study LGALS9 regulatory mechanisms
Development of anti-LGALS9 immunotherapies requires systematic investigation of targeting strategies:
Antibody-Based Approaches:
Small Molecule Inhibitors:
Target the carbohydrate recognition domains to disrupt glycan binding
Develop isoform-specific inhibitors to minimize off-target effects
Assess pharmacokinetics and tissue distribution to ensure target engagement
Combination Strategies:
Test anti-LGALS9 therapies in combination with existing immunotherapies
Explore synergies with chemotherapy or targeted therapies
Measure effects on both cancer and immune cells to assess mechanism of action
Targeting Strategy | Advantages | Limitations | Development Stage |
---|---|---|---|
Monoclonal antibodies | High specificity, well-established production | Limited tissue penetration | Early clinical trials |
Small molecule inhibitors | Better tissue penetration, oral administration | Lower specificity | Preclinical |
Gene silencing (siRNA/shRNA) | Highly specific reduction of expression | Delivery challenges | Research tool |
Soluble receptor decoys | Block LGALS9-receptor interactions | Short half-life, manufacturing complexity | Preclinical |
Accurate quantification of LGALS9 in complex tissues presents several challenges:
Tissue Heterogeneity:
Different cell populations within the same tissue may express varying levels of LGALS9
In pancreatic cancer models, LGALS9 expression varies between PanIN lesions, normal pancreatic tissue, Langerhans islets, and infiltrating immune cells
Solution: Combine laser capture microdissection with qPCR or use single-cell RNA sequencing
Subcellular Localization Variations:
Expression Dynamics:
Understanding cell type-specific LGALS9 expression patterns is essential for targeted interventions:
Pancreatic Cells (CD45- Population):
Peripheral Immune Cells (Circulating CD45+ Cells):
Tumor-Infiltrating Immune Cells (Tissue CD45+ Cells):
Methodological approach: Flow cytometry analysis after mechanical and enzymatic digestion of tissues, with appropriate gating strategies to distinguish immune (CD45+) from non-immune (CD45-) cells. Expression should be reported as relative fluorescence intensity (RFI) compared to isotype controls .
LGALS9 expression correlates with specific changes in T cell populations:
Total CD4+ T Cells:
No significant differences in peripheral CD4+ T cell levels between KC and wild-type mice
Significant increase in pancreatic infiltrating CD4+ T cells in KC mice compared to wild-type mice
More than 50% of wild-type mice had no lymphocyte infiltration, whereas all KC mice had some degree of infiltration
Regulatory T Cells (Tregs):
Correlation Analysis:
Methodological approach: Multi-parameter flow cytometry with appropriate markers for Tregs (typically CD4+CD25+FOXP3+) and analysis of both peripheral blood and tumor tissues to assess systemic versus local immune changes .
Ensuring specificity of anti-LGALS9 therapeutics requires comprehensive validation:
In Vitro Validation:
Binding affinity assays using recombinant LGALS9 proteins
Competitive binding assays with known LGALS9 ligands
Cross-reactivity testing against other galectin family members
Functional assays measuring inhibition of LGALS9-dependent activities
Cross-Species Reactivity:
Tissue Distribution Studies:
Assess binding to LGALS9 in different tissue types using immunohistochemistry
Validate specificity using LGALS9 knockout or knockdown controls
Evaluate potential off-target binding to similar proteins or glycan structures
Recent research suggests LGALS9 may play important roles in viral infections:
SARS-CoV-2 Interactions:
Methodological Approaches:
Viral replication assays in the presence/absence of LGALS9
Analysis of LGALS9 expression in COVID-19 patient samples
In vitro binding studies between LGALS9 and viral components
Therapeutic targeting of LGALS9 in viral infection models
Researchers investigating this area should consider both direct virus-LGALS9 interactions and indirect effects through immune modulation .
Systematic approaches to data mining can yield valuable insights:
Public Database Resources:
Data Extraction Methods:
Analysis Approaches:
Comparative analysis across different tissues and disease states
Correlation of LGALS9 expression with clinical outcomes
Network analysis to identify LGALS9-associated pathways
Meta-analysis combining multiple datasets for increased statistical power
Database | Data Type | Application in LGALS9 Research | Access Method |
---|---|---|---|
GEO | Transcriptomics | Expression analysis across conditions | R/GEOquery, Web interface |
TCGA | Multi-omics | Cancer-specific expression patterns | cBioPortal, Firehose |
GTEx | Tissue-specific expression | Normal tissue baseline expression | Web portal, R packages |
PRIDE | Proteomics | Protein expression and modifications | Web interface, APIs |
STRING | Protein interactions | LGALS9 interaction network | Web interface, R packages |
Data Distribution Assessment:
Significance Testing:
Visualization Approaches:
Correlation Analysis:
Assess relationships between LGALS9 expression and clinical parameters
Use Spearman correlation for non-parametric data
Consider multivariate analysis to account for confounding factors
When analyzing flow cytometry data, report relative fluorescence intensity (RFI) in relation to isotype controls to standardize results across experiments .
Galectin-9 was first isolated from mouse embryonic kidney in 1997 as a 36 kDa beta-galactoside lectin protein . It contains two carbohydrate recognition domains (CRDs) connected by a linker peptide, making it a tandem-repeat type galectin . The human recombinant form of Galectin-9 is typically produced in E. coli and has a molecular weight of approximately 34-37 kDa .
Galectin-9 plays a significant role in various biological processes:
Recombinant human Galectin-9 is widely used in research to study its effects on cell proliferation, apoptosis, and immune responses. It has been shown to support the adhesion of Jurkat cells, a type of human T cell leukemia . Additionally, it is used to investigate its potential therapeutic applications in cancer and autoimmune diseases.
Recombinant human Galectin-9 is typically produced in E. coli and purified to a high degree of purity (>95%) using SDS-PAGE under reducing conditions . The protein is lyophilized and can be stored at -20°C to -80°C for up to 12 months. Once reconstituted, it should be stored at 4-8°C for short-term use or at -20°C for longer-term storage .