LGALS2 mice are primarily utilized to investigate Galectin-2’s biological functions. Key models include:
Knockout (KO) Mice: Generated by deleting exons 2 and 3 of Lgals2, resulting in disrupted Galectin-2 expression at both transcript and protein levels . These mice are viable and fertile, with no overt abnormalities under baseline conditions.
Antibody-Treated Models: Llama-derived anti-Galectin-2 antibodies (e.g., 2H8, 2C10) are administered to inhibit Galectin-2 binding to monocytes, enabling studies on arteriogenesis and immune modulation .
Tumor-Bearing Models: Overexpression of Lgals2 in cancer cell lines (e.g., 4T1, EMT6) or inhibition via antibodies to study tumor growth and immune evasion .
LGALS2 inhibition enhances collateral artery growth in ischemic conditions. In a murine hind limb model with femoral artery ligation, anti-Galectin-2 antibodies improved perfusion and arteriolar diameter while promoting M2 macrophage polarization .
Parameter | Vehicle (Control) | 2H8 Antibody | 2C10 Antibody |
---|---|---|---|
Perfusion Restoration (%) | 41.3 ± 2.7 | 53.1 ± 3.4 | 52.0 ± 3.8 |
Arteriolar Diameter (μm) | 17.25 ± 4.97 | 17.71 ± 5.01 | 17.84 ± 4.98 |
M2 Macrophage Fraction | 0.49 ± 0.24 | 0.73 ± 0.15 | 0.75 ± 0.18 |
LGALS2’s role in cancer is context-dependent:
Colorectal Cancer: Lgals2 KO mice showed reduced oxidative stress sensitivity and increased tumor growth, suggesting Galectin-2 acts as a tumor suppressor in this context .
Triple-Negative Breast Cancer (TNBC): Lgals2 overexpression in 4T1 and EMT6 cell lines promoted tumor growth in vivo, linked to immune suppression via M2 macrophage polarization and reduced cytotoxic T-cell activity .
Parameter | Control | LGALS2 Overexpression |
---|---|---|
Tumor Volume (mm³) | 100 | 150 |
CD8+ T Cells (%) | 30 | 15 |
M2 Macrophages (%) | 40 | 60 |
LGALS2 regulates immune cell infiltration and polarization:
M2 Macrophages: Overexpression of Lgals2 in TNBC models increased M2-like macrophages (CD206+, Arg1+), which correlate with immunosuppression .
Lymphoid Cells: Anti-LGALS2 antibody treatment reduced regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs) while boosting cytotoxic T cells and NK cells .
Parameter | Anti-LGALS2 Treatment | Control |
---|---|---|
Tregs (%) | ↓ | ↑ |
MDSCs (CD11b+Ly6g+) (%) | ↓ | ↑ |
CD8+ T Cells (Gzmb+/Prf1+) | ↑ | ↓ |
Subtype | LGALS2 Expression | Hazard Ratio (OS) | P-value |
---|---|---|---|
Basal-like | High | 0.74 | 0.0018 |
Luminal B | High | 0.87 | 0.23 |
HER2-overexpressing | High | 0.93 | 0.43 |
Anti-LGALS2 Antibodies: In TNBC models, these antibodies reduced tumor volume by 50% and restored antitumor immunity .
Mechanism: Blockade of LGALS2 disrupts the CSF1/CSF1R axis, reducing M2 macrophage proliferation and tumor-associated myeloid cells .
LGALS2 exhibits opposing roles in different cancers:
While LGALS2 has been specifically implicated in immune evasion in breast cancer models, its expression pattern differs from other galectin family members. According to transcriptome analyses of galectin expression, Lgals9 demonstrates maximal spatial distribution across the mouse brain with predominant roles in neurogenesis, whereas LGALS1 shows ubiquitous expression in human tissues . The heterogeneous expression patterns of galectins between mouse and human tissues highlight the importance of careful interpretation when extrapolating findings across species. Limbic regions associated with learning, memory, and emotions, as well as substantia nigra associated with motor movements, showed strikingly high expression of LGALS1 and LGALS8 in human versus mouse brain .
The most informative experimental systems for studying LGALS2 include:
Experimental System | Application | Key Measurements |
---|---|---|
In vivo CRISPR screens | Functional genomics | Gene knockout phenotyping in tumor microenvironment |
Cell line overexpression models | Mechanistic studies | 4T1 and EMT6 murine TNBC cell lines |
In vitro co-culture systems | Cell-cell interaction | Macrophage polarization and proliferation |
Single-cell RNA sequencing | Transcriptional profiling | Immune cell population characterization |
Antibody blockade | Therapeutic potential | Tumor growth and immune cell changes |
These complementary approaches have been instrumental in uncovering LGALS2's role in immune regulation within the tumor microenvironment .
Single-cell RNA sequencing of 4T1 tumor-bearing BALB/c mice has revealed complex remodeling of the immune microenvironment by LGALS2. Specifically:
T cell compartment alterations:
Myeloid compartment remodeling:
Single-cell analysis identified six distinct macrophage clusters (Mφ1-Mφ6), with LGALS2 overexpression causing:
Decreased proportion of Mφ1
Increased proportions of Mφ2-Mφ6
Most dramatic difference in the Mφ5 cluster (9.62% of TIICs in LGALS2 overexpression versus 0.41% in vector control)
Substantial changes in the Mφ6 cluster, which acquired characteristics of the Mφ5 cluster
These findings demonstrate LGALS2's role as a master regulator of the immunosuppressive tumor microenvironment.
LGALS2 induces M2-like polarization and proliferation of macrophages through the CSF1/CSF1R axis . In vitro co-culture experiments confirmed that macrophages exposed to LGALS2-overexpressing tumor cells exhibit:
Upregulated expression of M2-like markers:
Enhanced proliferation compared to macrophages cocultured with control tumor cells
Transcriptome profiling identified CSF1 as a key mediator induced by LGALS2 in tumor cells. The CSF1/CSF1R axis is well-established in controlling macrophage development, differentiation, and M2-like polarization . This provides a molecular mechanism by which tumor cell-intrinsic LGALS2 influences the surrounding myeloid compartment to establish an immunosuppressive microenvironment.
Single-cell RNA sequencing has provided unprecedented resolution of LGALS2's effects on immune cell populations. The methodology applied in recent studies included:
Collection and analysis of cells from LGALS2-overexpressing (5,548 cells) and vector control (4,802 cells) tumors
Cell type identification using Cell Ranger and Seurat algorithms
Marker-based classification of immune cells (CD45+) and tumor cells (CD45−KRT18+)
t-SNE visualization of 13 distinct immune cell clusters
This approach revealed that LGALS2 overexpression caused:
Complex remodeling of immune cell populations
Heterogeneity within macrophage populations that would be missed by bulk RNA-seq or flow cytometry
Specific transcriptional signatures associated with immunosuppression
The single-cell approach was crucial for identifying the dramatic expansion of specific macrophage subsets (particularly Mφ5) and detecting subtle changes in gene expression within individual cell types .
Based on published methodologies, the following approach has proven successful:
Generation of stable overexpression models:
Validation should include:
Controls must include:
LGALS2 blockade using a single-domain llama-derived therapeutic antibody has demonstrated significant anti-tumor effects. For rigorous evaluation of LGALS2 blockade efficacy, researchers should:
Measure tumor parameters:
Analyze immune cell populations by flow cytometry:
Statistical analysis:
Based on published research, effective flow cytometry panels should include:
Cell Type | Surface Markers | Functional Markers |
---|---|---|
Cytotoxic T cells | CD45+CD3+CD8+ | Granzyme B, IFN-γ, PD-1 |
NK cells | CD45+CD3-NK1.1+ | Granzyme B, IFN-γ |
Regulatory T cells | CD45+CD3+CD4+CD25+FOXP3+ | - |
M2-like macrophages | CD45+CD11b+F4/80highCD206+ | - |
MDSCs | CD45+CD11b+Ly6g+ | - |
Proliferating cells | - | Ki67 |
For functional assessment, ex vivo stimulation of T cells followed by intracellular cytokine staining is recommended to evaluate production of effector molecules like Granzyme B and IFN-γ .
The distinct effects of LGALS2 in vitro versus in vivo highlight the importance of the tumor microenvironment in mediating LGALS2's functions. When interpreting these discrepancies:
Recognize that LGALS2's primary function appears to be immune modulatory rather than directly affecting cancer cell proliferation:
Consider experimental design limitations:
In vitro systems lack the complex immune microenvironment
Different mouse strains may have variable immune compositions
Timing of measurements can affect outcomes
Recommendations for reconciling discrepancies:
Use co-culture systems to bridge the gap between in vitro and in vivo findings
Implement humanized mouse models for better clinical translation
Employ multi-parameter analysis including spatial considerations (e.g., multiplexed immunohistochemistry)
Based on published methodologies, the following statistical approaches are recommended:
For comparing continuous variables:
For survival analysis:
For correlation analysis:
For multiple testing correction:
Results should be presented as means ± SEM unless otherwise indicated, with two-sided P values < 0.05 considered statistically significant .
When evaluating translatability of LGALS2 findings, researchers should consider:
Expression pattern differences:
Immune system differences:
Mouse and human immune systems have distinct compositions and functions
Different galectin family members may have evolved specialized functions across species
Recommendations for enhancing translatability:
Validate key findings in human cancer cell lines and patient-derived xenografts
Analyze LGALS2 expression in human tumor datasets (e.g., TCGA)
Perform comparative studies of LGALS2 function across species
Consider humanized mouse models for immunotherapy studies
Given LGALS2's role in immune escape, several combinatorial approaches warrant investigation:
Combination with immune checkpoint inhibitors:
Combination with CSF1R inhibitors:
Combination with conventional therapies:
Chemotherapy (may enhance immunogenic cell death)
Radiation therapy (may improve tumor antigen presentation)
The anti-LGALS2 antibody has already demonstrated significant anti-tumor effects as a monotherapy, suggesting potential for enhanced efficacy in combination regimens .
To expand our understanding of LGALS2 biology, researchers should consider:
Genetically engineered mouse models:
LGALS2 knockout mice for studying physiological functions
Conditional LGALS2 expression models to study temporal effects
Tissue-specific LGALS2 overexpression to dissect cell-intrinsic versus microenvironmental effects
Humanized mouse models:
Engraft human immune cells into immunodeficient mice
Useful for testing human-specific LGALS2 antibodies
Better recapitulate human immune responses
Patient-derived xenograft (PDX) models:
Maintain tumor heterogeneity and architecture
Allow testing of LGALS2 blockade in diverse tumor types
Evaluate biomarkers of response to LGALS2-targeted therapies
Emerging technologies that could advance LGALS2 research include:
Spatial transcriptomics:
Map LGALS2 expression and effects with spatial resolution
Understand localized immune interactions in the tumor microenvironment
Identify niches of LGALS2-responsive cells
Multi-omics approaches:
Integrate transcriptomics, proteomics, and metabolomics data
Provide comprehensive view of LGALS2's effects on multiple cellular processes
Identify novel mechanisms and pathway interactions
Advanced in vivo imaging:
Intravital microscopy to visualize immune cell dynamics in real-time
PET imaging with labeled anti-LGALS2 antibodies to track biodistribution
Bioluminescence imaging to monitor tumor response longitudinally
CRISPR-based screens:
Expand on existing in vivo CRISPR screens with higher sensitivity
Identify synthetic lethal interactions with LGALS2
Discover resistance mechanisms to LGALS2 blockade
Galectin-2 is an approximately 14 kDa homodimeric protein belonging to the S-type lectin family . It consists of a single carbohydrate-recognition domain (CRD), which is responsible for its binding to specific carbohydrate structures . The mouse Galectin-2 shares 86% and 66% amino acid sequence identity with rat and human Galectin-2, respectively .
Galectin-2 plays a significant role in modulating immune responses. It is known to induce a proinflammatory, anti-arteriogenic phenotype in monocytes and macrophages . This means that Galectin-2 can promote inflammation and inhibit the formation of new blood vessels, which is particularly relevant in the context of cardiovascular diseases . The protein exerts its effects through the CD14/toll-like receptor (TLR)-4 pathway, influencing the behavior of monocytes and macrophages .
Recombinant mouse Galectin-2 is widely used in research to study its biological functions and potential therapeutic applications. It is produced using human embryonic kidney cell (HEK293)-derived systems, ensuring high purity and activity . The recombinant protein is often used in experiments to understand its role in immune modulation, cell adhesion, and its potential as a therapeutic target in inflammatory and cardiovascular diseases .
Recombinant mouse Galectin-2 is typically lyophilized and should be reconstituted in water before use . It is recommended to store the protein at -20 to -70°C to maintain its stability and avoid repeated freeze-thaw cycles . Once reconstituted, it can be stored under sterile conditions at 2 to 8°C for up to one month or at -20 to -70°C for up to three months .