LGALS2 spans ~3.5 kb on chromosome 22 (NCBI Gene ID: 3957), encoding a 14.5 kDa protein that forms homodimers. Key features include:
Binding specificity: Recognizes β-galactosides, particularly lactose and N-acetyllactosamine residues.
Post-translational modifications: No glycosylation reported; exists as a secreted protein or cytoplasmic homodimer.
Orthologs: Highly conserved across mammals (e.g., mouse Lgals2 shares 85% amino acid identity).
LGALS2 is predominantly expressed in the gastrointestinal tract and immune cells, with additional presence in:
Tissue/Organ | Expression Level | Cellular Localization |
---|---|---|
Colon | High | Epithelial cells, monocytes |
Small intestine | High | Epithelial layer |
Stomach | Moderate | Glandular epithelium |
Gallbladder | High | Exocrine cells |
Kidney | Moderate | Renal tubules |
Breast | Low (tumor tissue) | Stromal/immune cells |
Data sourced from Human Protein Atlas and NCBI Gene Expression .
Acute colitis: Lgals2-deficient mice show reduced severity in dextran sodium sulfate (DSS)-induced colitis, suggesting endogenous LGALS2 exacerbates epithelial damage under oxidative stress .
Mechanism: Promotes STAT3 phosphorylation in epithelial cells, enhancing apoptosis during oxidative insults .
Genome-wide CRISPR screens identified LGALS2 as a critical regulator of cell survival under H₂O₂ treatment. Cells lacking LGALS2 exhibit increased resistance to oxidative damage, indicating its role in stress-induced apoptosis .
LGALS2 exhibits context-dependent effects in malignancy:
Cancer Type | Expression | Clinical Outcome | Mechanism |
---|---|---|---|
Colorectal (CRC) | Downregulated | Larger tumors, worse prognosis | STAT3 hyperactivation → Proliferation |
Breast (BRCA) | Upregulated | Better survival (basal-like/TNBC subtypes) | Immune regulation, T-cell infiltration |
Tumor suppression: Lgals2-KO mice develop larger AOM/DSS-induced tumors due to enhanced STAT3 activation and cell proliferation .
TCGA data: Reduced LGALS2 expression correlates with tumor aggressiveness .
Prognostic marker: High LGALS2 expression associates with improved survival (HR = 0.74, P = 0.0018) and better response to immunotherapy .
Immune regulation: Enriched in T-cell clusters and correlates with M2 macrophage polarization, enhancing antitumor immunity .
Cancer Type | Diagnostic Value | AUC | Source |
---|---|---|---|
Breast (TNBC) | High specificity for basal-like | 0.787 | METABRIC |
Colorectal | Low expression indicates poor prognosis | N/A | TCGA |
Targeting LGALS2: Preclinical models suggest overexpression may inhibit CRC growth by suppressing STAT3 , while blockade could enhance immunotherapy in BRCA .
Drug Repurposing: Connectivity Map (CMap) identifies small molecules targeting LGALS2 pathways for breast cancer treatment .
Condition | LGALS2 Status | p-STAT3 Level | Outcome |
---|---|---|---|
CRC tumors | KO | ↑↑ | Proliferation, tumor growth |
HCT116 cells (CRC) | Overexpressed | ↓↓ | Reduced proliferation |
Data from in vivo and in vitro studies .
T-cell modulation: LGALS2 overexpression correlates with increased T-cell infiltration in BRCA, enhancing immune surveillance .
Macrophage polarization: Links to M2-like macrophages in breast cancer, promoting anti-tumor responses .
Biomarker Development: Validate LGALS2 as a predictive marker for immunotherapy response in TNBC.
Therapeutic Strategies: Explore LGALS2-based interventions (e.g., recombinant protein for CRC, antibodies for BRCA).
Molecular Mechanisms: Investigate epigenetic regulation (e.g., promoter methylation) and cross-talk with oncogenic pathways (e.g., WNT/β-catenin).
LGALS2 encodes Galectin-2 (Gal2), a member of the beta-galactoside binding protein family. Galectin-2 is predominantly expressed in the gastrointestinal tract, with particularly high expression throughout the human colon. Research methodologies for studying LGALS2 expression typically involve tissue-specific RNA sequencing, Western blotting, and immunohistochemistry to quantify both transcript and protein levels across different tissues. Studies have confirmed this expression pattern through comparative analysis of multiple tissues, establishing the GI tract as the primary site of LGALS2 expression .
For researchers investigating LGALS2, several complementary approaches can be employed:
Transcript detection: RT-qPCR remains the gold standard for quantifying LGALS2 mRNA levels, with primers targeting exons 2-3 junction showing optimal specificity.
Protein detection: Western blotting using validated anti-LGALS2 antibodies provides reliable detection, with expected band size at approximately 15 kDa.
In situ detection: Immunohistochemistry and immunofluorescence with optimized antigen retrieval protocols (typically heat-mediated in citrate buffer) enable visualization of LGALS2 localization within tissues.
Secreted protein: ELISA assays can detect soluble LGALS2 in serum samples, which has been validated in clinical studies of cancer patients versus healthy controls .
When comparing LGALS2 expression across experimental conditions, researchers should control for tissue-specific expression patterns and consider the potential impact of oxidative stress, which has been shown to significantly affect LGALS2 levels .
LGALS2 has been demonstrated to influence several critical signaling pathways:
STAT3 signaling: LGALS2 appears to negatively regulate STAT3 phosphorylation, particularly under oxidative stress conditions. Experiments in colorectal cancer cell lines showed that LGALS2 overexpression significantly blunted H₂O₂-induced STAT3 phosphorylation, suggesting a direct or indirect inhibitory effect on this pathway .
CSF1/CSF1R axis: In triple-negative breast cancer models, tumor cell-intrinsic LGALS2 expression modulates the CSF1/CSF1R signaling axis, which plays a crucial role in macrophage recruitment, polarization, and proliferation within the tumor microenvironment .
Oxidative stress response: CRISPR knockout screening identified LGALS2 as a regulator of cellular response to oxidative stress, with LGALS2 deficiency conferring resistance to H₂O₂ treatment in multiple cell types .
Researchers studying these pathways should employ phospho-specific Western blotting, co-immunoprecipitation, and reporter assays to fully characterize the molecular interactions between LGALS2 and these signaling networks.
LGALS2 has significant immunomodulatory effects, particularly on macrophages within the tumor microenvironment:
Macrophage recruitment: Tumor cell-derived LGALS2 increases the number of tumor-associated macrophages (TAMs) in the tumor microenvironment.
M2-like polarization: LGALS2 promotes the polarization of macrophages toward an M2-like phenotype, which is generally considered pro-tumorigenic and immunosuppressive.
Macrophage proliferation: LGALS2 appears to enhance macrophage proliferation through the CSF1/CSF1R signaling axis.
Single-cell RNA sequencing of tumor-infiltrating immune cells has revealed that LGALS2 expression correlates strongly with M2-like macrophage scores (correlation coefficient = 0.591, P < 0.05), providing strong evidence for its role in shaping the immunosuppressive nature of the tumor microenvironment .
Experimental approaches to study these effects include co-culture systems, conditioned media experiments, flow cytometry with macrophage polarization markers, and in vivo models with selective macrophage depletion.
The relationship between LGALS2 and colorectal cancer appears complex and potentially context-dependent:
Expression pattern: Analysis of TCGA database has revealed that LGALS2 expression is significantly decreased in colon tumor tissues compared to normal tissues, suggesting a potential tumor-suppressive role .
Functional studies: Galectin-2 deficiency in experimental models promoted colorectal tumor growth despite ameliorating acute colitis. Specifically, in AOM/DSS-induced colorectal cancer mouse models, LGALS2 knockout mice developed significantly larger tumors than wild-type mice, although the total number of tumors was similar between groups .
Mechanistic basis: LGALS2 appears to suppress tumor cell proliferation under oxidative stress conditions. In vitro studies with colorectal cancer cell lines (HCT116 and Caco-2) demonstrated that LGALS2 overexpression significantly decreased proliferation and increased susceptibility to H₂O₂-induced cell death .
This apparent paradox—where LGALS2 deficiency reduces inflammation but promotes tumor growth—highlights the complex role of this protein in the tumor microenvironment and suggests stage-specific functions during cancer progression.
Recent research has identified LGALS2 as a potential immunotherapy target in triple-negative breast cancer (TNBC):
Immune escape mechanism: Functional screening and transcriptomic analysis identified LGALS2 as a candidate regulator of immune escape in TNBC. While LGALS2 overexpression did not affect cancer cell proliferation in vitro, tumors overexpressing LGALS2 grew significantly faster in vivo (P < 0.01), suggesting immunosuppression mediated by LGALS2 depends on interactions with the tumor microenvironment .
Serum biomarker potential: Patients with metastatic breast cancer had significantly increased concentrations of serum LGALS2 compared to healthy donors (P < 0.01), suggesting potential utility as a biomarker .
Therapeutic targeting: Blockade of LGALS2 using inhibitory antibodies successfully arrested tumor growth and reversed immune suppression in experimental models, highlighting its potential as a therapeutic target .
The differential effects of LGALS2 in various cancer types underscore the importance of context-specific research approaches when investigating this protein as a therapeutic target.
Researchers investigating LGALS2 function in vivo should consider these methodological approaches:
Genetic models: LGALS2 knockout mice, with deletion of exons 2 and 3, provide a valuable tool for studying the physiological consequences of LGALS2 deficiency. These models have been validated and show normal viability and fertility with no obvious gross abnormalities .
Inflammation models: The dextran sodium sulfate (DSS)-induced colitis model has been effectively employed to study LGALS2's role in intestinal inflammation. Researchers typically administer 3-5% DSS in drinking water for 5-7 days, assessing clinical parameters, histological changes, and inflammatory markers .
Cancer models: The azoxymethane (AOM)/DSS model of colitis-associated colorectal cancer represents a validated approach. This typically involves intraperitoneal injection of AOM (10 mg/kg) followed by repeated cycles of DSS treatment, enabling analysis of both tumor initiation and progression .
Immune microenvironment models: For TNBC research, orthotopic implantation of genetically modified cancer cells (LGALS2-overexpressing or knockout) into immunocompetent mice allows investigation of immunomodulatory effects. Flow cytometry, immunohistochemistry, and single-cell RNA sequencing can then characterize changes in the tumor immune microenvironment .
Therapeutic intervention: Anti-LGALS2 antibody administration in tumor-bearing mice provides a translational approach to assess therapeutic potential, with tumor growth measurement and immune profiling as key outcome measures .
These complementary approaches allow for comprehensive evaluation of LGALS2 function across different physiological and pathological contexts.
When designing experiments involving LGALS2 manipulation, researchers should implement these critical controls and validation steps:
Expression validation:
For knockout models: Confirm deletion at both mRNA level (RT-qPCR) and protein level (Western blot)
For overexpression models: Quantify expression levels relative to physiological expression in relevant tissues
Functional validation:
For cell line models: Test resistance to H₂O₂ challenge, as LGALS2 knockout should confer increased survival while overexpression should increase susceptibility
For in vivo models: Measure serum LGALS2 levels to confirm systemic effects of genetic manipulation
Specificity controls:
Include multiple LGALS2-targeting guide RNAs or siRNAs to rule out off-target effects
Consider rescue experiments by re-expressing LGALS2 in knockout models
Assess expression of other galectin family members to identify potential compensatory effects
Context-dependent controls:
Include both in vitro and in vivo experiments, as LGALS2 functions often differ between these contexts
Compare effects in multiple cell types, particularly those with high endogenous LGALS2 expression (GI tract-derived) versus low expression
Technical validation:
Employ multiple sgRNAs in CRISPR experiments with verification of editing efficiency
Use matched isogenic cell lines to minimize background genetic variation
These rigorous controls ensure that observed phenotypes can be confidently attributed to LGALS2 modulation rather than experimental artifacts or compensatory mechanisms .
The paradoxical effects of LGALS2—ameliorating inflammation while suppressing tumor growth—represent a significant research question. Several hypotheses may explain this apparent contradiction:
Cellular context specificity: LGALS2 may have distinct effects on different cell types. In acute colitis, LGALS2 primarily affects epithelial cells and immune cells, promoting epithelial cell death under oxidative stress. In contrast, in the tumor context, LGALS2 may predominantly suppress cancer cell proliferation .
Dose-dependent effects: The DSS dose used in colitis models (typically 3-5%) is higher than that used in tumor models (~1-2%), which may influence LGALS2's impact. Lower DSS doses may reduce the prominence of LGALS2's effect on epithelial damage .
Temporal dynamics: The acute phase of inflammation versus the chronic process of tumorigenesis may engage different LGALS2-dependent pathways. This phenomenon has been observed with other immune mediators like IL-17F, which shows similar paradoxical effects .
Microenvironmental factors: The inflammatory microenvironment differs substantially from the tumor microenvironment, potentially altering LGALS2's interaction partners and downstream effects.
Cell-autonomous versus non-cell-autonomous effects: LGALS2's effect on tumor growth may depend on interactions with immune cells, while its impact on colitis may be more epithelial cell-autonomous.
To address these hypotheses, researchers should consider cell type-specific knockout models, dose-response studies, and temporal analysis of LGALS2 function across disease progression stages.
The translational potential of LGALS2 in cancer research spans both therapeutic and diagnostic applications:
Therapeutic targeting:
Anti-LGALS2 antibodies have shown promise in preclinical models, arresting tumor growth and reversing immune suppression
The increased levels of serum LGALS2 in metastatic breast cancer patients suggest potential accessibility for therapeutic targeting
The observation that LGALS2 downregulation appears common in multiple cancer types (testicular germ cell tumors, kidney cancers, ovarian cancer) suggests broader applicability beyond breast cancer
Biomarker development:
Serum LGALS2 levels could serve as a prognostic or predictive biomarker
The correlation between LGALS2 and M2-like macrophage infiltration suggests potential utility in selecting patients for immunotherapy
Tissue LGALS2 expression patterns might help stratify patients for targeted therapies
Combination therapy strategies:
Given LGALS2's role in modulating macrophage polarization, combination with CSF1R inhibitors might enhance efficacy
The relationship between LGALS2 and STAT3 signaling suggests potential synergy with STAT3 pathway inhibitors
Anti-LGALS2 approaches might sensitize tumors to existing immunotherapies by reversing immune suppression
Clinical development considerations:
Target validation through correlative studies in human tumors
Development of companion diagnostics to identify patients likely to benefit
Monitoring of potential adverse effects, particularly in the GI tract where LGALS2 is highly expressed
The apparent tissue specificity of LGALS2 expression may provide a therapeutic window, potentially reducing systemic toxicity compared to broadly expressed targets .
Researchers investigating LGALS2's impact on immune cells should consider these methodological approaches:
Flow cytometry panels:
Basic panel: CD45, CD3, CD4, CD8, CD19, CD11b, F4/80, CD206 (to distinguish major immune populations)
Advanced panel: Add markers for M1/M2 polarization (iNOS, Arg1), T cell exhaustion (PD-1, Tim3), and activation status (CD69)
Single-cell RNA sequencing:
Spatial transcriptomics/proteomics:
Maps the spatial relationship between LGALS2-expressing cells and immune infiltrates
Provides context for cell-cell interactions that may mediate LGALS2 effects
In vitro co-culture systems:
Macrophage polarization assays with conditioned media from LGALS2-modified cancer cells
T cell functional assays (proliferation, cytokine production) in the presence of recombinant LGALS2
In vivo immune depletion:
Selective depletion of macrophages (using clodronate liposomes) or T cells (using depleting antibodies) to determine which immune populations mediate LGALS2 effects
When analyzing results, researchers should consider both quantitative changes (cell numbers) and qualitative alterations (activation status, polarization, functional capacity) across different immune compartments .
CRISPR screens have successfully identified LGALS2 as a regulator of oxidative stress response and immune escape. When designing similar screens, researchers should consider:
Library design:
Genome-wide versus focused libraries (the latter may provide higher coverage and statistical power)
Multiple sgRNAs per gene (at least 4-6) to ensure robust hit identification
Inclusion of non-targeting and essential gene controls
Selection strategy:
For oxidative stress responses: Sublethal H₂O₂ challenge (0.5 mM for 6 days has been validated)
For immune escape: In vivo screens comparing immunocompetent versus immunodeficient hosts
Readout methods:
Next-generation sequencing of sgRNA abundance
Analysis tools: casTLE or similar algorithms for statistical analysis of enrichment/depletion
Validation approaches:
Individual knockout of top hits using multiple sgRNAs
Functional assays (e.g., Cell Counting Kit/CCK8 for viability under stress)
Complementary gain-of-function experiments
Analysis across multiple cell types to address context specificity
Downstream analysis:
Gene Ontology analysis to identify enriched pathways
Integration with transcriptomic data to understand mechanism
Cross-referencing with patient databases to establish clinical relevance
These considerations will help ensure robust identification of LGALS2-related pathways and minimize false discoveries .
Galectin-2 is a member of the galectin family, which consists of carbohydrate-binding proteins with a high affinity for β-galactosides. These proteins play crucial roles in various biological processes, including cell-cell adhesion, cell-matrix interactions, and immune responses. Galectin-2, in particular, has garnered interest due to its unique properties and potential therapeutic applications.
Galectin-2 is a homodimeric protein with a molecular weight of approximately 14 kDa . It shares structural similarities with galectin-1 but has a distinct expression profile. Unlike galectin-1, which is widely expressed in various tissues, galectin-2 is primarily confined to the gastrointestinal tract . This restricted expression pattern suggests specialized functions in this region.
The gene encoding galectin-2 is located on chromosome 22 in humans. The protein consists of a single carbohydrate recognition domain (CRD), which is responsible for its binding to β-galactosides . The amino acid sequence of human galectin-2 shares significant identity with its mouse and rat counterparts, indicating a conserved function across species .
Galectin-2 has been shown to play a role in various biological processes:
Recombinant human galectin-2 is typically produced using human embryonic kidney (HEK) 293 cells. The recombinant protein is purified to high levels of purity, often exceeding 95%, and is free from endotoxins . This high-quality recombinant protein is used in various research applications, including studies on cell adhesion, apoptosis, and immune regulation.
The unique properties of galectin-2, particularly its ability to induce apoptosis in activated T cells, make it a promising candidate for therapeutic applications. It could be explored as a treatment for autoimmune diseases, where the selective elimination of activated T cells is desirable. Additionally, its role in modulating immune responses opens avenues for its use in cancer immunotherapy and inflammatory diseases .