LGALS7 exhibits tissue-specific expression, primarily in stratified epithelia:
Nuclear localization is observed in basal keratinocytes, suggesting roles in growth control and apoptosis .
LGALS7 regulates diverse cellular processes:
Induced by p53, LGALS7 triggers UVB-induced apoptosis in keratinocytes via JNK activation and cytochrome c release .
T Cell Dynamics: Reduces CD4+ T cells and increases CD8+ T cells in in vitro and in vivo models, enhancing SHP2 recruitment to PD-1 and dampening TCR signaling .
Myeloid Cell Recruitment: Binds CD11b+Ly6C<sup>hi</sup>Ly6G<sup>−</sup> myeloid cells, promoting immunosuppressive IL-10/TGF-β secretion .
Dual Roles:
Recent studies highlight LGALS7’s multifaceted roles:
Galectin-7 Deficiency: Reduces papilloma formation in Lgals7<sup>−/−</sup> mice, implicating LGALS7 in SCC progression .
Mechanism: Promotes immunosuppressive myeloid cell infiltration and T-cell inhibition .
SNPs rs567785577 (A) and rs138945880 (T) in the LGALS7 promoter are associated with intracerebral hemorrhage (ICH) risk (OR = 13.5) .
Mechanism: Altered cerebrovascular protein expression (e.g., serum amyloid A, myosin light chain) may impair blood-brain barrier integrity .
Knockdown Models: Silencing LGALS7 in mice decreases keratin-13 and myosin heavy chain expression, potentially affecting angiogenesis and cell migration .
Recombinant LGALS7 is used in research for functional assays and therapeutic studies:
Product Code | Tag | Activity Validated | Purity | Endotoxin |
---|---|---|---|---|
RPES4675 | None | Yes (RBC agglutination) | >95% | <1 EU/µg |
PKSH030935 | GST | Yes (ED50 <2 µg/mL) | >98% | <0.1 EU/µg |
PKSH032475 | None | No | >95% | <1 EU/µg |
CYT-016 | None | Yes | >95% | <1 EU/µg |
Recombinant Human Galectin-7, produced in E. coli, is a single, non-glycosylated polypeptide chain composed of 136 amino acids, resulting in a molecular weight of 15kDa. The purification of LGALS7 is achieved through proprietary chromatographic techniques.
LGALS7 was lyophilized from a concentrated (1mg/ml) solution in 20mM Tris, 150mM NaCl, 1mM EDTA and 5% Trehalose, pH 8.
To reconstitute the lyophilized Galectin-7, it is recommended to dissolve it in sterile distilled H2O at a concentration of at least 100µg/ml. This solution can then be further diluted in other aqueous solutions as needed.
The purity of the protein is determined to be greater than 95.0% as assessed by SDS-PAGE analysis.
MSNVPHKSSLPEGIRPGTVLRIRGLVPPNASRFHVNLLCGEEQGSDAALHFNP
RLDTSEVVFNSKEQGSWGREERGPGVPFQRGQPFEVLIIASDDGFKAVVGDAQ
YHHFRHRLPLARVRLVEVGGDVQLDSVRIF
The LGALS7 gene, located on chromosome 19q13.2, encodes the galectin-7 protein, which is comprised of 136 amino acids. Galectin-7 belongs to the prototypic group of the LGALS family of proteins—lectin molecules that bind to β-galactosides via carbohydrate recognition domains (CRDs). Unlike other galectins that are widely distributed throughout the human body, galectin-7 shows tissue- and cell-specific distribution patterns that include brain tissues. As a prototypic galectin, each galectin-7 molecule contains one CRD and can form homodimers upon binding to glycoconjugates .
Unlike other widely distributed galectins (such as Gal-1 and Gal-3), galectin-7 demonstrates a more restricted tissue- and cell-specific distribution pattern in the mammalian body, including brain tissues. This specific distribution pattern suggests galectin-7 may have specialized roles in certain tissues, potentially including the brain, where it could be involved in various physiological and pathological processes. This tissue specificity makes galectin-7 particularly interesting when studying organ-specific diseases such as cerebrovascular disorders .
Based on the research findings, LGALS7 appears to be involved in multiple biological processes, particularly those related to vascular function and inflammation. The gene product interacts with various proteins involved in signal transduction and molecular metabolic processes. In the cerebrovascular context, galectin-7 may influence blood vessel integrity, potentially through interactions with proteins like α-actin, myoglobin, and serum amyloid A, which could affect vascular contractility, nitric oxide regulation, and inflammatory processes. Methodologically, these functions have been identified through proteomic analyses comparing wild-type mice with those having altered LGALS7 expression .
Research has identified two significant single nucleotide polymorphisms (SNPs) in the LGALS7 promoter region that show association with intracerebral hemorrhage (ICH) risk: rs567785577 and rs138945880. Specifically, the A allele of rs567785577 and the T allele of rs138945880 were found to be associated with increased risk of ICH. These polymorphisms are located in the genomic region 19q13.2, immediately preceding the gene start code. The SNPs were identified through sequencing of the LGALS7 promoter region in patients with hemorrhagic stroke compared to healthy controls .
For effective detection of LGALS7 genetic variants, researchers should implement a multi-stage approach similar to that described in the referenced study. This would include: (1) An exploratory phase using animal models with altered LGALS7 expression to identify potential phenotypic effects; (2) A discovery phase involving genomic DNA extraction from peripheral blood samples (using methods such as the AxyPrep Blood Genomic DNA Miniprep Kit); (3) PCR amplification of target regions at optimized annealing temperatures; (4) Sanger sequencing for genotyping; and (5) Statistical analysis applying different genetic models (dominant, recessive, overdominant, codominant, additive, and multiplicative) to identify significant associations. The study design should include appropriate case and control cohorts with clearly defined inclusion/exclusion criteria .
For analyzing haplotype structures of LGALS7 SNPs, researchers should first ensure they have high-quality genotyping data from a sufficiently large sample. Based on the provided research, the methodological approach should include: (1) Verification that allele frequencies are in Hardy-Weinberg Equilibrium (HWE) with p > 0.05; (2) Construction of haplotype blocks covering the entire study population; (3) Calculation of allele frequencies within these blocks; (4) Determination of odds ratios (OR) with 95% confidence intervals to assess disease association; and (5) Statistical analysis using appropriate models to adjust for potential confounding factors such as age, hypertension, diabetes, and lipid disorders. This approach allowed researchers to identify significant associations between specific LGALS7 alleles and ICH risk (e.g., Allele T: OR = 13.5, 95% CI = 2.249–146.5, p = 0.002) .
To generate effective transgenic mouse models for studying LGALS7 function, researchers should follow a systematic approach that includes: (1) Creating both overexpression models (TG LGALS mice) and knockdown models (TG LGALS-DOWN mice) using appropriate vectors; (2) Confirming successful transgene integration through PCR amplification at different annealing temperatures (as shown in Figure 2 of the research); (3) Verifying altered expression at both gene and protein levels using PCR and Western blot analysis, respectively; (4) Establishing breeding programs to create different generations (F0, F1, F2) for studying inheritance patterns; and (5) Using multiple primer sets for verification (producing fragments of approximately 323 bp and 372 bp). The validation of these models should include demonstration of differential expression before proceeding with phenotypic analyses .
Based on the research findings, the most suitable proteomics approach for identifying galectin-7-associated proteins involves: (1) Using iTRAQ (isobaric tags for relative and absolute quantitation) labeling of peptides; (2) Implementing HPLC separation with gradient elution using a sandwich-based method; (3) Conducting MS analysis in triplicate to ensure reliability, with a requirement that peptides be identified at least twice before inclusion in analysis; (4) Employing comparative analysis between galectin-7 expressing and knockdown models to identify differentially expressed proteins; (5) Utilizing bioinformatics tools like the KEGG database, Reactome pathway database, and STRING database to detect functional interactions and classify proteins into biochemical pathways; and (6) Applying Gene Ontology (GO) analysis to categorize proteins by biological process, cellular component, and molecular function. This comprehensive approach led to the identification of 1,009 differentially expressed proteins, of which 28 were known proteins with altered expression in relation to galectin-7 levels .
The technical challenges in analyzing LGALS7 promoter sequences include: (1) Ensuring high-quality DNA extraction from peripheral blood samples; (2) Optimizing PCR conditions for the specific GC content of the promoter region; (3) Achieving consistent and reliable sequencing results, particularly in regions with potential polymorphisms; (4) Correctly identifying and validating novel SNPs; and (5) Appropriately analyzing the statistical significance of identified variants in relation to disease risk.
To overcome these challenges, researchers should: (1) Use standardized DNA extraction kits optimized for blood samples; (2) Test multiple annealing temperatures for PCR reactions (as demonstrated in Figure 2); (3) Implement technical replicates and bidirectional sequencing; (4) Verify sequencing results with alternative methods; (5) Use vector insertion of the promoter region (750 bp) to confirm sequence integrity; and (6) Apply appropriate statistical models with correction for multiple testing when analyzing associations. These approaches helped researchers successfully identify and validate the significant SNPs rs567785577 and rs138945880 .
When designing case-control studies to investigate LGALS7 associations with ICH, researchers should: (1) Clearly define the patient population (e.g., spontaneous non-traumatic ICH patients) and collect comprehensive clinical data including traditional vascular risk factors (hypertension, diabetes, lipid disorders); (2) Select demographically matched controls from the same ethnic population to minimize population stratification effects; (3) Implement a structured two-stage approach with an exploratory phase utilizing animal models followed by a discovery phase with human subjects; (4) Collect standardized biological samples (peripheral whole blood in appropriate anticoagulants) and process them according to validated protocols; (5) Apply consistent genotyping methods; and (6) Use appropriate statistical analyses that consider different genetic models and adjust for potential confounding factors. In the referenced study, this approach revealed that traditional vascular disease risk factors were significantly more common in the stroke group than in the control group (p < 0.001), providing context for genetic findings .
The relationship between LGALS7 variants and traditional risk factors for stroke appears complex and multifactorial. Research indicates that while the identified LGALS7 SNPs (rs567785577 and rs138945880) show significant associations with ICH risk, traditional vascular risk factors remain highly important. In the study population, rates of hypertension, diabetes, and lipid disorders were significantly higher in ICH cases compared to controls. This suggests that LGALS7 variants may interact with or add to the risk conferred by traditional factors.
From a methodological perspective, researchers investigating this relationship should: (1) Collect comprehensive data on traditional risk factors; (2) Apply multivariate analyses to determine independent contributions of genetic and traditional factors; (3) Explore potential interaction effects between genetic variants and environmental or physiological risk factors; and (4) Consider stratified analyses to identify potential differential genetic effects based on the presence of specific traditional risk factors .
Based on the proteomic findings, LGALS7 may contribute to cerebrovascular pathology in ICH through several potential mechanisms: (1) Influence on cerebrovascular wall-related proteins - altered galectin-7 expression affects proteins involved in vascular structure and function; (2) Impact on blood-brain barrier integrity - galectin-7-associated proteins like alpha-1-acid glycoprotein 2 can modulate blood-brain barrier permeability; (3) Effects on smooth muscle cell function - galectin-7 appears to influence proteins like α-actin that are essential for contractile functions of vascular smooth muscle cells; (4) Regulation of nitric oxide activity - through interaction with proteins like myoglobin, galectin-7 may affect nitric oxide regulation in cerebral blood vessels; and (5) Modulation of inflammatory processes - silencing of LGALS7 led to increased levels of serum amyloid A (SAA), a protein involved in inflammatory cell recruitment.
These mechanisms suggest that altered LGALS7 expression could inhibit actin-driven angiogenesis, affect endothelial cell activities, and influence inflammatory processes that contribute to cerebral amyloid angiopathy (CAA), which in turn can lead to vessel wall weakening and hemorrhage risk .
Proteomic analysis has identified several key protein interactions of galectin-7 that may relate to cerebrovascular function. Among the 28 known differentially expressed proteins detected, several have direct implications for vascular health:
Proteins increased in galectin-7 knockdown mice: "Serum amyloid A protein" and "Actin 1"
Proteins decreased in galectin-7 knockdown mice: "Myosin regulatory light chain 2," "Keratin (type I cytoskeletal 13)," "Troponin C," "Creatine kinase M-type," "Myosin light chain 2," and "Myosin, heavy polypeptide 8"
Proteins progressively increased in galectin-7-overexpressing mice: "Calsequestrin," "Myosin light chain 1/3," "Myoglobin," "Myosin regulatory light chain 2," "Myosin-1," and "Keratin (type II cytoskeletal 4)"
These interactions suggest galectin-7 influences contractile proteins (myosins), structural proteins (keratins, actin), calcium-handling proteins (calsequestrin, troponin C), and inflammatory markers (serum amyloid A). Functionally, these interactions may affect vascular contractility, structural integrity, calcium signaling, and inflammatory responses in cerebral vessels, all of which are critical for maintaining cerebrovascular health and preventing hemorrhage .
For analyzing differential protein expression data related to LGALS7, researchers should implement a comprehensive bioinformatics approach that includes: (1) Hierarchical cluster analysis using tools like Cluster 3.0 to group proteins based on expression profiles; (2) Pathway classification using the Kyoto Encyclopedia of Genes and Genomes (KEGG) database and Reactome pathway database to assign proteins to biochemical pathways; (3) Protein-protein interaction analysis using the STRING database to detect functional interactions between identified proteins; (4) Gene ontology (GO) analysis to categorize proteins into biological processes, cellular components, and molecular functions; (5) Statistical analysis to determine significance of differential expression, with requirement for replication; and (6) Integrative analysis that combines proteomic findings with genetic data to develop comprehensive models of galectin-7 function.
This multilayered approach allowed researchers to determine that the most highly enriched GO functional categories associated with differentially expressed proteins were related to signal transduction and molecule metabolic processes, providing insight into the biological significance of galectin-7 interactions .
To effectively validate protein interactions identified through proteomics studies of LGALS7, researchers should implement a multi-method validation strategy: (1) Confirm differential expression using orthogonal techniques such as Western blotting, as demonstrated for verification of galectin-7 knockdown; (2) Perform co-immunoprecipitation assays to verify direct protein-protein interactions; (3) Utilize immunohistochemistry or immunofluorescence to visualize co-localization of galectin-7 with candidate interacting proteins in relevant tissues; (4) Apply functional assays to assess the biological significance of identified interactions (e.g., contractility assays for smooth muscle-related proteins); (5) Use knockout or knockdown models of interacting proteins to assess reciprocal effects on galectin-7 function; and (6) Validate findings in human tissues or primary cell cultures to ensure relevance to human pathophysiology.
This comprehensive validation approach helps ensure that proteomics-identified interactions represent biologically significant relationships rather than technical artifacts, and provides insight into the functional consequences of these interactions .
Based on the research findings, LGALS7 genetic variations could potentially affect response to immunosuppressant treatments in CAA through several mechanisms: (1) Influence on inflammatory pathways - given that galectin-7 appears to regulate inflammatory processes and silencing leads to increased levels of inflammatory markers like serum amyloid A; (2) Impact on vascular remodeling - as galectin-7 affects proteins involved in vascular structure and function; and (3) Modulation of immune cell recruitment and activation in cerebrovascular tissues.
The research noted that in CAA patients, those receiving immunosuppressant treatment had a recurrence rate of 26% compared to 71% in those not receiving such treatment. This significant difference suggests that inflammatory and immune processes are critical in CAA pathogenesis. Methodologically, researchers investigating this relationship should: (1) Genotype CAA patients for LGALS7 variants; (2) Compare treatment outcomes based on genotype; (3) Assess inflammatory markers before and after treatment; and (4) Consider prospective clinical trials stratified by LGALS7 genotype to determine if genetic status predicts treatment response .
To elucidate the role of LGALS7 in blood-brain barrier (BBB) function and cerebrovascular integrity, researchers should consider these advanced experimental approaches: (1) In vitro BBB models using endothelial cells with modulated LGALS7 expression, assessing barrier integrity through transendothelial electrical resistance (TEER) and permeability assays; (2) Live cell imaging to visualize the dynamics of tight junction proteins in response to altered galectin-7 levels; (3) 3D cerebral organoids with LGALS7 variants to study vascular development in a complex tissue context; (4) Intravital microscopy in transgenic mouse models to directly visualize BBB integrity in vivo; (5) Single-cell RNA sequencing of neurovascular unit components to identify cell-specific responses to altered galectin-7 expression; and (6) Analysis of cerebrospinal fluid and plasma biomarkers of BBB integrity in individuals with different LGALS7 genotypes.
These approaches would build upon the finding that galectin-7 interacts with proteins that influence BBB permeability, such as alpha-1-acid glycoprotein 2, which can modulate the blood-brain barrier by adding negative charges to its matrix component .
To investigate potential ethnic variations in LGALS7-associated ICH risk, researchers should design studies that: (1) Include diverse ethnic populations with adequate sample sizes for each group to ensure statistical power; (2) Implement standardized case definitions and sample collection protocols across all populations; (3) Use population-specific controls matched for demographic factors; (4) Conduct comprehensive genotyping of the LGALS7 promoter and coding regions to identify both common and population-specific variants; (5) Apply haplotype analysis to detect population differences in linkage disequilibrium patterns; (6) Perform meta-analyses when multiple studies are available; and (7) Investigate gene-environment interactions that may differ between ethnic groups.
The current research noted that "further studies with expanded case numbers that include subjects of other ethnic populations are needed to elucidate mechanisms underlying associations between these SNPs and ICH risk." This acknowledges that the identified associations in the Chinese Han population may differ in other ethnic groups due to genetic background, environmental factors, and differing prevalence of traditional risk factors .
Galectin-7 was first reported by Celis in 1995 while searching for keratinocyte proteins that may play a role in maintaining the normal phenotype and various skin diseases . The protein is expressed in stratified epithelia and is involved in apoptotic responses, proliferation, differentiation, cell adhesion, and migration .
The expression of Galectin-7 is induced by the tumor suppressor protein p53 and is associated with apoptosis . It has diverse effects on many cellular functions, including:
Recombinant human Galectin-7 is produced using an E. coli expression system. The target protein is expressed with the sequence Met1-Phe136 of human LGALS7 (Accession #P47929) . The recombinant protein is typically purified to a high degree of purity (>95%) and is used in various research applications .
Recombinant human Galectin-7 is used in research to study its role in various pathological states, including autoimmune diseases, allergic reactions, inflammation, tumor cell metastasis, atherosclerosis, and diabetic complications . It is also used to investigate its potential as a therapeutic target in cancer and other diseases .