The LIPG gene encodes a 500-amino acid protein synthesized as a 55 kDa precursor. Post-translational glycosylation increases its molecular weight to 68 kDa .
Structural motifs include the catalytic triad (Ser169, Asp193, His274), a heparin-binding domain, and a lid region governing substrate specificity .
LIPG is predominantly expressed in vascular endothelial cells and highly vascularized tissues (e.g., liver, lung, kidney) .
Single-cell RNA sequencing reveals enriched expression in endothelial cells and specific epithelial compartments .
LIPG exhibits phospholipase A1 (PLA1) activity, preferentially hydrolyzing high-density lipoprotein (HDL) phospholipids . Key metabolic roles include:
Atherosclerosis: Elevated LIPG correlates with low HDL-C, a risk factor for cardiovascular disease .
Metabolic Syndrome: High plasma LIPG associates with obesity, hypertriglyceridemia, and insulin resistance .
Inflammation: LIPG upregulation during endotoxemia or chronic inflammation amplifies pro-inflammatory cytokines (IL-6, CRP) and adhesion molecules (VCAM-1) .
Triple-Negative Breast Cancer (TNBC):
Plasma LIPG levels serve as a biomarker for metabolic dysfunction and subclinical inflammation .
In TNBC, high LIPG mRNA expression predicts shorter metastasis-free survival (HR = 2.1, p < 0.01) .
LIPG Human Recombinant (HEK-derived):
Oncology: Validate LIPG inhibitors in clinical trials for TNBC and explore combinatorial therapies targeting lipid metabolism .
Cardiology: Develop LIPG-neutralizing therapies to elevate HDL-C in dyslipidemia .
Structural Biology: Leverage AlphaFold-predicted models (via Human Protein Atlas) to design allosteric inhibitors .
LIPG encodes endothelial lipase, which functions as both a phospholipase and triglyceride lipase, with predominant phospholipase activity. Its primary function is regulating circulating levels of high-density lipoprotein cholesterol (HDL-C), with phospholipid-enriched HDL being its preferred substrate . Beyond enzymatic functions, LIPG can form molecular bridges between endothelial cells and lipoproteins or circulating macrophages through interaction with heparan sulfate proteoglycans. This non-enzymatic action can increase cellular lipoprotein uptake and monocyte adhesion, potentially contributing to atherosclerosis .
LIPG expression has been detected in multiple human tissues, predominantly in the liver, placenta, lung, thyroid, kidney, testis, and in the corpus luteum of the ovary . Notably, LIPG expression is not detected in heart, brain, and muscle tissues . In pathological conditions, LIPG has been identified as a hallmark of Triple-negative breast cancer (TNBC) and shows altered expression in lung adenocarcinoma (LUAD) . This tissue-specific expression pattern suggests differential regulatory mechanisms and potentially diverse functions across various organs.
As a phospholipase with specificity for HDL phospholipids, LIPG hydrolyzes HDL phospholipids, thereby reducing HDL particle size and facilitating HDL catabolism . This activity directly influences HDL-C levels in circulation. Recent research suggests that, beyond its established role in HDL metabolism, LIPG may contribute to very low-density lipoprotein (VLDL) and low-density lipoprotein (LDL) metabolism . The precise mechanisms by which LIPG affects VLDL and LDL metabolism remain an active area of investigation, but this broader impact on lipoprotein profiles highlights the complex role of LIPG in lipid homeostasis.
The human LIPG gene is located on chromosome 18q21.1 . This genomic position places LIPG in a region that contains several other genes involved in various biological processes. Understanding the genomic architecture surrounding LIPG is important for comprehending its evolutionary conservation, regulatory elements, and potential genetic interactions that might influence its function in lipid metabolism and other physiological processes.
The LIPG 584C/T polymorphism has been extensively studied for its association with coronary artery disease (CAD) risk. A meta-analysis of 14 case-control studies involving 9,731 subjects (4,025 cases and 5,706 controls) revealed a significant protective association between this polymorphism and CAD risk . This association was particularly evident in the homozygote comparison model (TT vs. CC) and the allelic comparison model (T vs. C) . The results suggest that individuals carrying the T allele may have a reduced susceptibility to CAD compared to those with the C allele. The protective effect might be mediated through alterations in HDL-C metabolism, as LIPG is known to regulate HDL-C levels.
Various LIPG polymorphisms have been associated with alterations in serum lipid profiles, though findings across studies show some inconsistency. Studies have reported associations between several LIPG SNPs (including rs2156552, rs4939883, and rs7241918) and serum HDL-C concentrations, but these associations vary across different populations . The effects appear to be influenced by multiple factors, including ethnicity, sex, and environmental conditions. For instance, a study comparing Maonan and Han populations in China found population-specific associations between LIPG polymorphisms and lipid parameters . These findings highlight the complex interplay between genetic variants and environmental factors in determining individual lipid profiles.
Significant ethnic variations exist in the distribution of LIPG polymorphisms. Research comparing different populations, such as the Maonan and Han ethnic groups in China, has revealed distinct patterns of LIPG polymorphism frequencies and their associations with serum lipid levels . These differences may reflect evolutionary adaptations to varied environmental conditions, dietary habits, or other population-specific factors. The variation in LIPG polymorphism distributions across ethnic groups can partly explain the inconsistent findings regarding associations between LIPG variants and lipid parameters or disease risks.
Several validated methodologies are commonly employed for genotyping LIPG polymorphisms:
Polymerase Chain Reaction-Restriction Fragment Length Polymorphism (PCR-RFLP): Successfully used for genotyping LIPG SNPs, including rs2156552, rs4939883, and rs7241918 . This involves amplifying the region containing the SNP of interest, followed by digestion with specific restriction enzymes.
TaqMan Assay: This real-time PCR-based method employs allele-specific fluorescent probes and offers high throughput and accuracy for SNP genotyping .
Direct Sequencing: Often used as a confirmation method, direct sequencing provides the most definitive validation of genotypes determined by other methods .
The choice of method depends on laboratory resources, sample size, cost considerations, and specific study requirements. For large-scale epidemiological studies, high-throughput methods like TaqMan may be preferred, while PCR-RFLP might be more suitable for smaller-scale investigations. Confirmation by direct sequencing is recommended for a subset of samples to ensure genotyping accuracy.
Accurate measurement of LIPG expression requires a multi-faceted approach:
Quantitative Real-Time PCR (qRT-PCR): Widely used for measuring LIPG mRNA expression levels with high sensitivity and specificity . Careful selection of reference genes for normalization is crucial.
Immunohistochemistry (IHC): Effectively employed to verify LIPG protein expression in tissue samples, providing information about both expression levels and spatial distribution .
RNA-Sequencing (RNA-Seq): For comprehensive transcriptome analysis, offering advantages in detecting novel transcripts and splice variants of LIPG .
Western Blotting: Allows for semi-quantitative assessment of LIPG protein levels and can complement IHC findings.
For comprehensive analysis, it is advisable to combine multiple methods to assess both mRNA and protein expression. Including appropriate controls and validating findings across different specimen types enhances the reliability of LIPG expression measurements.
The analysis of LIPG polymorphism data requires robust statistical methods:
Hardy-Weinberg Equilibrium (HWE) Testing: Essential to verify genotyping data quality and ensure control populations follow expected genetic distributions .
Odds Ratios (ORs) with 95% Confidence Intervals (CIs): Widely used to evaluate associations between LIPG polymorphisms and disease risks across multiple genetic models :
Heterozygote comparisons (e.g., CT vs. CC)
Homozygote comparisons (e.g., TT vs. CC)
Recessive model (e.g., TT vs. CT + CC)
Dominant model (e.g., TT + CT vs. CC)
Allelic comparisons (e.g., T vs. C)
Stratified Analyses: Conducted by ethnicity, disease subtypes, genotyping methods, and sources of controls to identify potential heterogeneity sources .
Heterogeneity Assessment: Q-test and I² statistics for evaluating heterogeneity across studies or subgroups . High heterogeneity may necessitate random-effects models.
Adjustment for Multiple Testing: When examining multiple SNPs or genetic models, appropriate corrections should be applied to minimize type I errors.
Survival Analysis: For assessing prognostic value, Kaplan-Meier curves and Cox proportional hazards models are appropriate analytical tools .
The selection of statistical methods should be guided by specific research questions, study design, and the nature of outcome variables being investigated.
Several bioinformatics tools and resources are valuable for investigating LIPG and its functional networks:
Gene Expression Analysis Platforms:
Functional Enrichment Analysis Tools:
Gene Ontology (GO) Analysis: Identifies biological processes, cellular components, and molecular functions associated with LIPG and its co-expressed genes .
KEGG Pathway Analysis: Maps LIPG and related genes to established biological pathways .
GSEA (Gene Set Enrichment Analysis): Identifies statistically significant differences in predefined gene sets between biological states .
Immune Infiltration Analysis:
Network Analysis Tools:
STRING: Constructs protein-protein interaction networks involving LIPG.
Cytoscape: Visualizes complex networks and integrates various data types.
Genomic Databases:
These bioinformatics resources, when used in combination, can provide comprehensive insights into the functional significance of LIPG in various biological contexts and disease states.
To implement LIPG as a prognostic biomarker in clinical settings, researchers should consider:
Tissue Analysis: Immunohistochemistry for assessing LIPG protein expression with standardized scoring systems.
Cutoff Determination: Establishing appropriate values for distinguishing high versus low LIPG expression through ROC curve analysis.
Multivariate Analysis: Evaluating LIPG expression alongside established clinicopathological factors through multivariate Cox regression models.
Combined Biomarker Panels: Integrating LIPG with other biomarkers and clinical parameters to enhance predictive accuracy.
Validation in Diverse Cohorts: Ensuring generalizability across different patient populations.
Implementation of LIPG as a prognostic biomarker could improve risk stratification in LUAD patients and inform treatment decisions, contributing to more personalized therapeutic approaches.
LIPG expression exhibits significant correlations with immune cell infiltration in the tumor microenvironment, particularly in lung adenocarcinoma. Research has employed algorithms like CIBERSORT to deconvolute the immune landscape in relation to LIPG expression , while the ESTIMATE algorithm has been used to calculate stromal and immune scores in tumor samples .
Key findings include:
This relationship suggests potential mechanisms by which LIPG might influence cancer progression and treatment responses, possibly through its role in lipid metabolism or direct effects on immune cell behavior. Understanding these interactions could lead to novel therapeutic strategies targeting the LIPG-immune axis in cancer.
While not explicitly discussed in the search results, evidence suggests potential for targeting LIPG in various disease contexts:
Cardiovascular Disease: Given the protective role of the LIPG 584C/T polymorphism in coronary artery disease , modulating LIPG activity might offer a novel approach for cardiovascular disease prevention or treatment.
Cancer Therapy: The association of high LIPG expression with poor prognosis in lung adenocarcinoma suggests therapeutic potential through:
a. Direct LIPG Inhibition: Developing small molecule inhibitors or antibodies.
b. Gene Silencing Approaches: Using RNA interference or CRISPR-based technologies.
c. Combination with Immunotherapy: Given LIPG's correlation with immune infiltration .
Metabolic Disorders: LIPG's role in lipoprotein metabolism suggests applications in treating dyslipidemia.
Challenges to address include:
Specificity: Ensuring selective targeting without affecting related lipases.
Tissue-Specific Delivery: Developing targeted delivery strategies.
Safety Assessment: Evaluating potential side effects on lipid metabolism and immune function.
Biomarkers for Patient Selection: Identifying who would most likely benefit from LIPG-targeted therapy.
Further research into LIPG's structure, function, and regulatory mechanisms is needed to fully realize its therapeutic potential.
LIPG polymorphisms could enhance cardiovascular risk assessment by providing additional genetic information that complements traditional risk factors:
Risk Stratification: The LIPG 584C/T polymorphism, with its protective role in coronary artery disease , could be incorporated into genetic risk scores, potentially modifying preventive intervention strategies.
Population-Specific Assessment: Given ethnic variations in LIPG polymorphism distributions , cardiovascular risk assessment should be tailored to specific populations.
Multi-SNP Analysis: Considering multiple LIPG SNPs simultaneously might provide more comprehensive risk assessment than individual variants .
Gene-Environment Interactions: Incorporating interactions between LIPG polymorphisms and environmental factors could further refine risk prediction models.
Integration with Lipid Biomarkers: Combining genetic information with traditional lipid biomarkers might improve risk prediction accuracy beyond either approach alone.
Implementation requires validation in large, diverse cohorts and development of user-friendly clinical tools that translate genetic information into actionable risk assessments. Cost-effectiveness analyses would determine the value added by including LIPG polymorphism testing in routine cardiovascular risk assessment.
The association between LIPG and immune cell infiltration in tumors likely involves several molecular mechanisms:
Lipid Mediator Production: LIPG's phospholipase activity may generate bioactive lipid mediators that influence immune cell recruitment and function within the tumor microenvironment.
Bridging Function: LIPG can form molecular bridges between endothelial cells and circulating macrophages through interaction with heparan sulfate proteoglycans , potentially facilitating immune cell adhesion and extravasation into tumor tissues.
Altered Lipoprotein Metabolism: LIPG-mediated changes in lipoprotein profiles could affect immune cell metabolism and function, as lipoproteins provide essential cholesterol and fatty acids for immune cell membranes and signaling.
Inflammatory Signaling Pathways: LIPG expression might influence inflammatory signaling cascades affecting immune cell recruitment and activation.
Extracellular Vesicle Modification: LIPG could potentially modify the lipid composition of tumor-derived extracellular vesicles, altering their immunomodulatory effects.
Research using CIBERSORT algorithm has demonstrated correlations between LIPG expression and specific immune cell populations in LUAD . Further mechanistic studies combining targeted LIPG modulation with immune phenotyping would help elucidate the causal relationships in the LIPG-immune axis in cancer.
LIPG functions within a complex network of genes involved in lipid metabolism:
Co-expression Networks: Gene Ontology and KEGG pathway analyses applied to LIPG and its co-expressed genes can reveal functional clusters indicating potential interacting partners.
Lipoprotein Lipase Family Interactions: As a member of the Lipoprotein Lipase family , LIPG likely shares functional relationships with other family members, including LPL and LIPC, with complementary or compensatory roles.
HDL Metabolism Pathway: LIPG interacts functionally with genes involved in HDL metabolism, potentially including ABCA1, ABCG1, SCARB1, CETP, and LCAT.
VLDL and LDL Metabolism: Evidence suggests LIPG may contribute to VLDL and LDL metabolism , implying interactions with genes like APOB, LDLR, and PCSK9.
Regulatory Interactions: LIPG expression may be controlled by transcription factors that also govern other lipid metabolism genes, creating an integrated regulatory network.
Gene Set Enrichment Analysis has been employed to identify gene expression patterns related to LIPG , providing insights into its functional interactions. Further research using protein-protein interaction analyses would help elucidate specific molecular interactions within lipid metabolism pathways.
While the search results don't explicitly discuss epigenetic regulation of LIPG, several mechanisms likely influence its expression:
DNA Methylation: Methylation of CpG islands in the LIPG promoter region could modulate transcriptional activity, potentially explaining tissue-specific expression and changes in pathological conditions.
Histone Modifications: Various histone marks likely play roles in regulating LIPG expression. ChIP-seq could map these modifications across the LIPG locus in different cell types and conditions.
Non-coding RNAs: LIPG is associated with long non-coding RNA DANCR in triple-negative breast cancer , suggesting lncRNAs may be important regulators. Additionally, microRNAs might target LIPG mRNA.
Chromatin Remodeling: Changes in chromatin accessibility could affect the binding of transcription factors to the LIPG promoter and enhancers.
Three-dimensional Genome Organization: Long-range chromatin interactions between the LIPG promoter and distal regulatory elements might contribute to differential expression patterns.
Methodology for investigating these mechanisms would include bisulfite sequencing, ChIP-seq, ATAC-seq, Hi-C, and RNA-seq combined with knockdown/overexpression of potential epigenetic regulators. Understanding epigenetic regulation could provide insights into LIPG dysregulation in diseases and reveal novel therapeutic targets.
Recent evidence suggests LIPG may have broader effects on lipoprotein metabolism beyond HDL processing . Potential mechanisms include:
Direct Enzymatic Action: Although LIPG preferentially acts on HDL phospholipids, its triglyceride lipase activity could potentially affect triglyceride-rich lipoproteins like VLDL, with structural similarities allowing interaction with phospholipids on VLDL and LDL particles.
Indirect Effects via HDL Remodeling: LIPG-mediated modifications of HDL could indirectly influence VLDL and LDL metabolism through effects on CETP activity, which facilitates lipid exchange between HDL and apoB-containing lipoproteins.
Cellular Lipid Homeostasis: By affecting cellular uptake of HDL-derived lipids, LIPG might alter intracellular lipid pools influencing VLDL production in hepatocytes and modulating expression of genes governing VLDL secretion and LDL receptor activity.
Impact on Lipoprotein Clearance: LIPG's bridging function between lipoproteins and cell surfaces might facilitate the binding and uptake of VLDL and LDL particles, affecting their clearance rates.
Inflammatory Modulation: LIPG-generated lipid mediators might influence inflammatory pathways affecting VLDL production and LDL modification.
Research methodologies could include in vitro assays with purified LIPG and different lipoprotein fractions, transgenic animal models, lipidomic analyses, and kinetic studies tracking apolipoprotein B metabolism in subjects with different LIPG genotypes.
Lipase Endothelial (Human Recombinant) is produced in HEK (Human Embryonic Kidney) cells. The recombinant form is a single, glycosylated polypeptide chain consisting of 490 amino acids, with a calculated molecular mass of approximately 55.8 kDa . The enzyme is fused to a 2 amino acid N-terminal linker, a 2 amino acid C-terminal linker, and a 6 amino acid His tag at the C-terminus .
LIPG exhibits extensive phospholipase activity and is more active as a phospholipase than as a triglyceride lipase . It hydrolyzes high-density lipoproteins (HDL) more efficiently than other lipoproteins . This enzyme is involved in the hydrolysis of triglycerides, both with short-chain fatty acyl groups (such as tributyrin) and long-chain fatty acyl groups (such as triolein), showing similar levels of activity toward both types of substrates .
The primary role of LIPG in the body is to regulate lipoprotein metabolism and maintain vascular health. By hydrolyzing HDL, LIPG helps in the remodeling and clearance of plasma lipoproteins . This activity is essential for maintaining lipid homeostasis and preventing the accumulation of lipids in the blood vessels, which can lead to cardiovascular diseases .