LGALS7 Mouse refers to transgenic (TG) rodent models engineered to overexpress or silence the LGALS7 gene, encoding Galectin-7, a β-galactoside-binding lectin critical in epithelial homeostasis, apoptosis, and immune modulation . These models enable mechanistic studies of Galectin-7’s role in diseases like stroke, cancer, and immune disorders, leveraging genetic manipulation to alter LGALS7 expression .
TG mice are generated via microinjection of plasmid constructs into fertilized oocytes:
Overexpression: pcDNA3.1-LGALS7 vector inserts full-length LGALS7 cDNA into oocytes, resulting in systemic Galectin-7 overexpression .
Knockdown: pSilencer-LGALS7 vector introduces shRNA targeting LGALS7, suppressing its expression .
Protein | Expression Change | Proposed Function |
---|---|---|
Serum Amyloid A (SAA) | ↑ (Increased) | Inflammatory cell recruitment |
Actin 1 | ↑ (Increased) | Angiogenesis regulation |
Myosin Regulatory Light | ↓ (Decreased) | Contractility and cell migration |
Calsequestrin | ↑ (Increased) | Calcium storage in muscle |
Data derived from iTRAQ analysis of knockdown vs. WT mice .
Signal Transduction: Enriched GO terms include JNK activation and cytochrome c release, linking Galectin-7 to apoptosis .
Immune Modulation: Galectin-7 suppresses CD4+ T cells and enhances CD8+ T cells, reducing tumor growth in MC38 and LL/2 syngeneic models .
Intracerebral Hemorrhage (ICH): LGALS7 promoter SNPs (rs567785577, rs138945880) are associated with ICH risk (OR = 13.5, p = 0.002) . TG models enable study of Galectin-7’s role in vascular integrity.
Skin Carcinogenesis: Galectin-7 overexpression in Tg46 mice accelerates non-melanoma skin cancer (NMSC) by recruiting immunosuppressive myeloid cells .
Epithelial Tumors: Knockdown in gastric cancer models reduces proliferation and invasion .
The mouse LGALS7 gene is located on chromosome 19q13.2, analogous to its human counterpart. The full-length cDNA encodes a galectin-7 protein comprised of 136 amino acids . The protein structure contains a characteristic carbohydrate recognition domain (CRD) that is essential for its functionality in binding to glycosylated substrates. When investigating LGALS7 in mice, researchers should note that the protein sequence spans from Ser2 to Phe136 according to standard reference databases (Accession # AAK29385) . The promoter region, which is critical for expression regulation, contains several polymorphic sites that have been associated with pathological conditions in translational studies.
Multiple approaches exist for modulating LGALS7 expression in mice. Overexpression models can be generated through microinjection of pcDNA3.1-LGALS7 plasmid vectors into fertilized oocytes (one-cell-stage fertilized eggs) . For knockdown studies, researchers have successfully used pSilencer-LGALS7 plasmid vectors containing shRNA constructs targeting LGALS7 . Both transgenic lines can be maintained by backcrossing with BALB/c mice.
Genotyping of transgenic founders and offspring should be performed using PCR amplification of tail snip DNA with transgene-specific primers. Validation of successful genetic manipulation can be confirmed at both the transcript level (via PCR) and protein level (via Western blotting) . When designing these experiments, researchers should consider:
Genetic Manipulation | Vector System | Verification Method | Common Applications |
---|---|---|---|
Overexpression | pcDNA3.1-LGALS7 | PCR, Western blot | Tumor models, immune modulation studies |
Knockdown | pSilencer-LGALS7 (shRNA) | PCR, Western blot | Loss-of-function studies, phenotypic analysis |
Tissue-specific expression | Cre-loxP systems | Immunohistochemistry | Tissue-specific functional analysis |
For phenotypic validation, expression analysis in target tissues is essential, as shown in transgenic models where differential expression patterns were confirmed through PCR and protein detection methods .
When investigating LGALS7's effect on T cell populations, a multi-tiered experimental approach is recommended. Based on recent findings, galectin-7 reduces CD4+ T cell percentages while potentially increasing CD8+ T cells in both in vitro and in vivo systems .
For in vitro assessment, human PBMCs should be cultured with purified recombinant galectin-7 (recommended concentration range: 1-10 μg/ml) or BSA control for 7 days. Flow cytometry analysis should be performed to determine the relative percentages of CD4+ and CD8+ T cells among total lymphocytes, using appropriate fluorochrome-conjugated antibodies against CD3, CD4, and CD8 .
For in vivo validation, two complementary approaches can be implemented:
Humanized mouse models: Inject human PBMCs into NSG mice and treat with galectin-7 (e.g., every 4 days starting from day 14 after PBMC injection). Compare with PBS-treated controls.
Syngeneic tumor models: Use mouse cancer cell lines (e.g., MC38 colon cancer or LL/2 lung cancer) with either:
Critically, researchers should include both wild-type and specialized knockout models (e.g., PD-1 KO mice) to address mechanistic questions. Endpoints should include analysis of T cell subpopulations in peripheral blood, spleen, and tumor tissues by flow cytometry, with particular attention to CD4+ T cells, CD4+CD25+ regulatory T cells, and CD8+ T cells .
Investigation of LGALS7 promoter polymorphisms requires careful genetic analysis methodology. Based on recent studies exploring ICH susceptibility, researchers should:
Define clear case and control populations with detailed inclusion/exclusion criteria
Extract genomic DNA using standardized protocols
Sequence the entire LGALS7 promoter region, particularly focusing on known SNP locations (e.g., rs567785577 and rs138945880 on 19q13.2)
Employ multiple genetic models for analysis:
Statistical analysis should include calculation of odds ratios with 95% confidence intervals and appropriate correction for multiple testing. Continuous variables should be presented as mean ± SE, with p-values < 0.05 considered statistically significant . For comprehensive pathway analysis, researchers should consider conducting differential protein expression studies, as demonstrated in the transgenic mouse models where iTRAQ peptide analysis identified 1,009 differentially expressed proteins with 28 known proteins showing significant correlation with LGALS7 expression levels .
The relationship between galectin-7 and tumor growth is context-dependent and requires careful experimental design to elucidate. Based on syngeneic mouse models, galectin-7 demonstrates distinct effects depending on the immune competence of the host and molecular context.
In wild-type immunocompetent mice, galectin-7 administration (1.5 mg/kg every 4 days) suppressed MC38 tumor growth, correlating with a reduction in CD4+ T cells and CD4+CD25+ regulatory T cells . This effect appears to be dependent on PD-1 signaling, as galectin-7 treatment did not reduce CD4+ T cell percentages or suppress tumor growth in PD-1 knockout mice .
The proposed mechanism involves:
Selective reduction of CD4+ T cells (including regulatory T cells)
Relative sparing of CD8+ T cells
De-suppression of anti-tumor immune responses
Host Immune Status | Effect of Galectin-7 on Tumor | Proposed Mechanism |
---|---|---|
Immunocompetent | Tumor suppression | Reduction of regulatory T cells, de-suppression of anti-tumor immunity |
PD-1 deficient | No effect | Unable to modulate T cell populations due to disrupted PD-1 signaling |
Immunodeficient | Tumor promotion | Direct pro-tumorigenic effects on cancer cells without immune regulation |
These differential effects highlight the importance of considering both the tumor microenvironment and systemic immune context when investigating galectin-7's role in cancer progression .
Proteomic analysis of LGALS7 transgenic and knockdown mice has revealed complex molecular networks influenced by this protein. Using iTRAQ peptide identification and mass spectrometry, researchers identified 1,009 differentially expressed proteins associated with galectin-7 expression levels .
Key protein interaction changes observed in LGALS7 knockdown mice include:
Relatively increased expression:
Relatively decreased expression:
Conversely, LGALS7 overexpression progressively increased:
Calsequestrin
Myosin light chain 1/3
Myoglobin
Myosin regulatory light chain 2
Myosin-1
Pathway analyses revealed that these protein changes affect:
Signal transduction cascades
Molecular metabolic processes
Cerebrovascular functions
Blood-brain barrier permeability
Inflammatory responses, particularly through SAA-mediated chemotactic recruitment of inflammatory cells
These interactions suggest that LGALS7 influences multiple pathways including actin-driven angiogenesis, endothelial cell activities, and vascular basement membrane integrity. The downstream effects include potential impacts on cerebral amyloid angiopathy (CAA) and inflammatory processes around blood vessels .
Detection of galectin-7 in mouse tissues requires appropriate methodological approaches depending on the research question. Immunohistochemistry (IHC) has been successfully employed for tissue localization studies, particularly in skin sections where galectin-7 expression is prominent .
Recommended IHC Protocol for Galectin-7 Detection:
Fix tissue samples (e.g., mouse skin) using immersion fixation in formaldehyde
Process and embed in paraffin following standard protocols
Prepare sections (typically 4-6 μm thickness)
Perform heat-induced epitope retrieval using basic retrieval reagents (pH 9.0)
Block endogenous peroxidase activity
Incubate with primary antibody against mouse galectin-7 (optimal concentration: 1 μg/ml) for 1 hour at room temperature
Use HRP-polymer antibody detection systems for visualization
Develop with DAB (brown) and counterstain with hematoxylin (blue)
Evaluate staining patterns, noting that galectin-7 localizes to both cytoplasm and nucleus
For quantitative protein analysis, Western blotting provides reliable results using antibodies with confirmed specificity against mouse galectin-7 . Flow cytometry can also be employed for detection of galectin-7 in cell suspensions, particularly when analyzing interactions with immune cell populations .
Importantly, researchers should consider that galectin-7 expression is tissue-specific, with high expression in epithelial tissues, particularly the skin. When analyzing other tissues, appropriate positive and negative controls should be included to validate detection methods .
To analyze the functional relationship between LGALS7 and immune cell populations, researchers should implement a multi-faceted approach:
In vitro T cell assays:
In vivo immune composition analysis:
Use syngeneic mouse tumor models with galectin-7 treatment (1.5 mg/kg every 4 days)
At defined endpoints, harvest peripheral blood, spleen, and tumor tissues
Perform comprehensive flow cytometry analysis of immune cell populations
Focus on CD4+ T cells, CD8+ T cells, and CD4+CD25+ regulatory T cells
Genetic manipulation approaches:
Immune modulation experiments:
These approaches should be complemented with appropriate controls, including PBS vehicle controls and isotype control antibodies. Statistical analysis should account for biological variability, with experiments performed in triplicate at minimum.
The contradictory findings regarding LGALS7's role in cancer progression reflect its context-dependent functions and require careful interpretation. Researchers should consider several key factors when reconciling seemingly contradictory data:
Immune context dependency:
Signaling pathway dependencies:
Cancer type specificity:
Local glycome composition:
To address these contradictions methodologically, researchers should:
Design experiments with multiple cancer models
Include both immunocompetent and immunodeficient backgrounds
Analyze detailed immune cell profiles in tumor microenvironments
Consider glycomic analyses alongside proteomic approaches
Implement genetic manipulation studies (knockdown, overexpression, and domain mutants) to dissect mechanism
Sample preparation standardization:
Data filtering and validation:
Biological pathway analysis:
Result interpretation framework:
Data Pattern | Potential Interpretation | Validation Approach |
---|---|---|
Proteins increased in LGALS7 knockdown | Negatively regulated by galectin-7 | Rescue experiments with recombinant galectin-7 |
Proteins decreased in LGALS7 knockdown | Positively regulated by galectin-7 | siRNA knockdown of these proteins in WT background |
Progressive increase with LGALS7 overexpression | Dose-dependent regulation | Titration experiments with varying galectin-7 levels |
Researchers should be cautious about several factors that could influence interpretation, including potential off-target effects of genetic manipulations, compensatory mechanisms in transgenic models, and the influence of developmental timing on observed phenotypes .
Galectin-7 is a member of the galectin family, which consists of proteins that bind to β-galactoside sugars. These proteins play crucial roles in various cellular processes, including cell-cell adhesion, cell-matrix interactions, and apoptosis. Galectin-7, in particular, is known for its involvement in the regulation of apoptosis and its expression in epithelial tissues.
Galectin-7 is a 14 kDa protein encoded by the LGALS7 gene. The recombinant form of mouse Galectin-7 is typically expressed in Escherichia coli and purified to a high degree of purity, often exceeding 95% as determined by SDS-PAGE . The protein consists of 136 amino acids and includes a His tag at the N-terminus to facilitate purification .
Galectin-7 is primarily known for its pro-apoptotic functions. It acts upstream of the JNK activation and mitochondrial cytochrome c release, which are critical steps in the apoptosis pathway . This protein is particularly important in the context of UV-induced apoptosis of keratinocytes, playing a vital role in maintaining epidermal homeostasis .
Recombinant mouse Galectin-7 is widely used in research to study its role in apoptosis and other cellular processes. It is suitable for various applications, including SDS-PAGE and mass spectrometry (MS) . Researchers often use this protein to investigate the mechanisms underlying cell death and survival, as well as its potential implications in diseases such as cancer.