LIPG (Lipase Endothelial) Human, HEK refers to a recombinant protein produced in human embryonic kidney (HEK293) cells, designed for laboratory research on lipoprotein metabolism and vascular biology. This engineered protein retains native enzymatic activity while enabling controlled experimental conditions .
Phospholipase Dominance: Hydrolyzes HDL more efficiently than other lipoproteins, impacting cholesterol transport .
Triglyceride Lipase Activity: Contributes to lipid storage and droplet formation in cancer cells .
Heparin Binding: Anchors to heparan sulfate proteoglycans (HSPGs) on endothelial cells, critical for cellular localization .
Mutation | Activity Level vs. Wild-Type | Functional Implications |
---|---|---|
T111I | 100% | No change in phospholipase inhibition |
R54C | 79–89% | Reduced HDL remodeling capacity |
N52S | 0% | Complete loss of enzymatic activity |
Cancer Metabolism:
Cardiovascular Studies:
Therapeutic Development:
LIPG (Lipase Endothelial) is a single, glycosylated polypeptide chain protein with extensive phospholipase activity that plays a crucial role in lipoprotein metabolism and vascular biology. LIPG Human, HEK specifically refers to human LIPG that has been recombinantly produced in Human Embryonic Kidney (HEK) cells .
The protein belongs to the triglyceride (TG) lipase gene family with significant sequence identity to other lipases: 46% to lipase member H (LIPH), 45% to lipoprotein lipase (LPL), 40% to hepatic lipase (HL) and 31% to pancreatic lipase (PNLIP) . LIPG is synthesized not only by endothelial cells but also by macrophages and hepatocytes .
Functionally, LIPG serves as a key enzyme that regulates and hydrolyzes serum high-density lipoprotein (HDL), generating free fatty acids and low-lipid apolipoprotein A1 . Animal studies have demonstrated that overexpression of LIPG results in reduced HDL-C levels, while LIPG deficiency leads to marked elevation of HDL-C levels, highlighting its importance in HDL metabolism .
LIPG Human recombinant protein produced in HEK cells consists of:
A single, glycosylated polypeptide chain spanning from Ser21 to Pro500 of the native sequence
A total of 490 amino acids from the native protein
A 2 amino acid N-terminal linker
A 2 amino acid C-terminal linker
A 6 amino acid His tag at the C-Terminus for purification purposes
The protein contains a characteristic lid region, which is a structural feature common to the TG lipase family that provides specificity for the phospholipase activity of the enzyme . The complete amino acid sequence includes specific regions responsible for substrate binding and catalytic activity, making it functionally similar to the native human protein .
For maintaining LIPG activity and stability, researchers should follow these methodological guidelines:
Storage conditions:
Store the lyophilized protein at -20°C for long-term stability
After reconstitution, aliquot the protein to avoid repeated freezing/thawing cycles
Reconstituted protein can be stored at 4°C for a limited period (up to two weeks without significant change)
Reconstitution protocol:
Add approximately 200μl of deionized water to prepare a working stock solution of approximately 0.5 mg/ml
Allow the lyophilized pellet to dissolve completely
Note that the standard preparation is not sterile; filter the solution through an appropriate sterile filter before using it in cell culture applications
Critical handling considerations:
The physical appearance of the product should be a filtered white lyophilized powder
The formulation typically includes phosphate buffered saline pH 7.5 (PBS), 1% (w/v) Sucrose, and 4% (w/v) Mannitol
The product's purity is typically greater than 85.0% as determined by SDS-PAGE analysis
Following these methodological guidelines will help ensure experimental reproducibility and maintain the functional integrity of the protein.
When designing functional assays with LIPG Human, HEK, researchers should optimize several experimental parameters:
Buffer and pH conditions:
Start with phosphate buffered saline (PBS) pH 7.5, as this is used in the protein formulation
Test pH ranges between 7.0-8.0 to determine optimal enzymatic activity
Consider including divalent cations (especially Ca²⁺) which may enhance lipase activity
Temperature optimization:
Conduct enzymatic assays at physiological temperature (37°C) for most applications
For extended incubations, verify temperature stability with time-course experiments
Substrate considerations:
For phospholipase activity assays, HDL particles or phospholipid vesicles can serve as physiologically relevant substrates
Fluorogenic or chromogenic lipase substrates can provide quantitative readouts for high-throughput screening
Consider substrate concentration effects on enzyme kinetics (Km, Vmax)
Methodological controls:
Include heat-inactivated enzyme controls
Use buffer-only reactions to account for non-enzymatic hydrolysis
Consider including known LIPG inhibitors as negative controls
Use fresh reconstitutions of the enzyme for critical experiments
For cell-based assays, start with concentrations in the range of 10-100 ng/ml, while biochemical assays may require higher concentrations (0.1-1 μg/ml). Always perform dose-response experiments to determine the optimal concentration for your specific experimental system .
Accurate measurement of LIPG activity requires carefully selected methodological approaches depending on the experimental context:
Biochemical activity assays:
Substrate hydrolysis assays: Monitor the release of free fatty acids from phospholipid substrates using colorimetric detection methods
Fluorogenic substrate assays: Use specific fluorescent substrates (such as 4-methylumbelliferyl phosphate derivatives) that increase fluorescence upon hydrolysis
Radiometric assays: Employ radiolabeled substrates to track product formation with high sensitivity
HDL-specific functional assays:
HDL remodeling assays: Measure changes in HDL particle size using native gel electrophoresis or NMR spectroscopy
ApoA-I dissociation assays: Quantify the release of ApoA-I from HDL particles after LIPG-mediated phospholipid hydrolysis
Phospholipid composition analysis: Use mass spectrometry to characterize changes in HDL phospholipid profiles after LIPG treatment
Cell-based approaches:
Cellular lipid uptake: Measure LIPG-mediated changes in cellular lipid accumulation
Reporter assays: Monitor LIPG expression and regulation in response to experimental conditions
Fluorescently labeled HDL tracking: Visualize HDL processing in the presence of LIPG
Validation strategies:
Establish assay linearity with respect to enzyme concentration and reaction time
Determine assay sensitivity and limit of detection
Confirm specificity using LIPG inhibitors or genetic knockdown approaches
Include appropriate positive and negative controls in each experiment
For researchers new to LIPG activity assays, it is advisable to begin with established biochemical assays using purified components before advancing to more complex cellular or in vivo systems .
LIPG SNPs have been associated with alterations in serum lipid profiles, particularly HDL-C levels, with significant implications for cardiovascular research:
Key LIPG SNPs affecting lipid metabolism:
rs2000813: Associated with increased HDL-C levels in multiple studies
rs3813082: A polymorphism in the LIPG promoter region associated with plasma HDL-C levels
Methodological approaches for LIPG SNP analysis:
SNP selection strategy:
Genotyping methodologies:
PCR-RFLP (Polymerase Chain Reaction-Restriction Fragment Length Polymorphism): Cost-effective approach involving amplification followed by restriction enzyme digestion
Direct DNA sequencing: Gold standard for SNP confirmation
Consider using specific forward and backward primers designed for the target regions
Experimental design considerations:
Research findings on population differences:
Studies have shown varying effects of LIPG SNPs across different ethnic populations. For example, research on the Maonan nationality (a conservative and isolated minority in Southwest China) revealed specific associations between LIPG SNPs and serum lipid levels that may differ from other populations .
For optimal results, researchers should employ a combination of genotyping approaches, with initial screening by PCR-RFLP followed by confirmation of selected samples with direct sequencing .
To comprehensively investigate LIPG's role in HDL metabolism, researchers should employ multi-level experimental approaches:
In vitro biochemical systems:
Reconstituted HDL particle assays with purified components
Real-time monitoring of HDL remodeling using fluorescent or radioactive labels
Analysis of HDL composition changes using lipidomic and proteomic approaches
Kinetic analyses of LIPG activity against various HDL subfractions
Cellular models:
Primary endothelial cells expressing endogenous LIPG
Cell lines with controlled LIPG expression (overexpression, knockdown, or knockout)
Co-culture systems to study intercellular HDL metabolism
Fluorescently labeled HDL to track cellular processing
Animal model approaches:
LIPG knockout mouse models show marked elevation of HDL-C levels
LIPG overexpression models demonstrate reduced HDL-C levels
Tissue-specific LIPG expression models help identify tissue-specific effects
Adenoviral-mediated acute manipulation of LIPG expression for temporal studies
Translational human studies:
Analysis of LIPG genetic variants and their association with HDL levels
Ex vivo studies using human samples (plasma, cells, or tissues)
Clinical studies correlating LIPG activity with HDL function and cardiovascular outcomes
Methodological integration:
Begin with biochemical characterization of LIPG-HDL interactions
Progress to cellular models to understand physiological context
Validate findings in animal models with altered LIPG expression
Correlate with human genetic and clinical data
This multi-level approach allows researchers to develop a comprehensive understanding of LIPG function in HDL metabolism while addressing potential discrepancies between different experimental systems .
Post-translational modifications (PTMs) of LIPG significantly impact its enzymatic activity, stability, and function, requiring specialized methodological approaches for investigation:
Glycosylation effects and analysis:
LIPG is a glycosylated protein, with N-linked glycosylation sites that can affect protein folding and stability
The pattern and extent of glycosylation may differ depending on the expression system used
Methods to study glycosylation include:
Methodological approaches for studying other PTMs:
Phosphorylation analysis:
Phospho-specific antibodies for Western blotting
Mass spectrometry with phospho-enrichment techniques
In vitro kinase assays to identify potential regulatory kinases
Proteolytic processing:
N-terminal sequencing to identify processing sites
SDS-PAGE and Western blotting to detect multiple forms
Mutation of potential cleavage sites to assess functional significance
Disulfide bond characterization:
Non-reducing versus reducing SDS-PAGE
Mass spectrometry approaches for disulfide mapping
Site-directed mutagenesis of cysteine residues
Experimental considerations:
The choice of expression system (HEK cells) provides human-like post-translational modifications
Consistent sample preparation is essential to maintain PTM integrity
Storage conditions can affect PTM stability
When comparing different LIPG preparations, characterize their PTM profiles
Understanding these modifications is crucial for accurate interpretation of experimental results and for developing strategies to manipulate LIPG activity for research or therapeutic purposes.
The literature on LIPG function contains some discrepancies, particularly regarding its effects on lipoprotein metabolism. Researchers can address these inconsistencies through methodological rigor:
Sources of experimental discrepancies:
Methodological variations:
Biological complexity factors:
Methodological strategies to resolve discrepancies:
Standardization approaches:
Develop and implement standardized protocols for LIPG activity assays
Clearly document and report all experimental conditions in publications
Use multiple complementary assays to validate findings
Consider interlaboratory validation studies for critical findings
Comprehensive experimental design:
Include appropriate positive and negative controls in every experiment
Test hypotheses across multiple experimental systems (in vitro, cellular, in vivo)
Perform dose-response studies rather than single-dose experiments
Account for potential compensatory mechanisms in genetic models
Integrative analysis:
Combine in vitro biochemical data with cellular and in vivo findings
Correlate enzymatic activity with physiological outcomes
Use systems biology approaches to place LIPG in broader metabolic context
Develop computational models to predict LIPG effects in complex systems
By addressing these factors systematically, researchers can help resolve apparent discrepancies and develop a more coherent understanding of LIPG function in lipoprotein metabolism .
Inhibition studies are valuable for understanding LIPG function and exploring its potential as a therapeutic target. Here are methodological considerations for designing effective LIPG inhibition studies:
Types of inhibition approaches:
Small molecule inhibitors:
Screen compound libraries for potential LIPG inhibitors
Characterize inhibition mechanisms (competitive, non-competitive, etc.)
Test for selectivity against other lipase family members (LPL, HL)
Determine structure-activity relationships to optimize potency
Biological inhibitors:
Develop specific antibodies against LIPG's active site
Design peptide inhibitors based on substrate recognition sequences
Evaluate neutralizing antibodies for specificity and potency
Genetic approaches:
RNA interference (siRNA, shRNA) for transient or stable LIPG knockdown
CRISPR-Cas9 genome editing for complete LIPG knockout
Antisense oligonucleotides targeting LIPG mRNA
Experimental design considerations:
Controls: Include vehicle controls, positive controls (known inhibitors if available), and negative controls
Dose-response: Perform careful dose-response studies to determine IC50 values
Time-course: Establish optimal incubation times for inhibition assays
Specificity testing: Evaluate effects on related lipases to determine inhibitor specificity
Validation across multiple systems:
Begin with purified protein for direct biochemical assays
Progress to cell-based assays to confirm intracellular efficacy
Test in ex vivo systems (e.g., human plasma samples)
Validate promising inhibitors in animal models
Readouts for inhibition:
Direct measurement of LIPG enzymatic activity using appropriate substrates
Assessment of HDL remodeling and phospholipid hydrolysis
Monitoring of downstream effects on lipoprotein metabolism
Evaluation of potential physiological consequences (e.g., HDL levels, function)
Carefully designed inhibition studies with appropriate controls and validation steps can provide valuable insights into LIPG function and its potential as a therapeutic target.
Based on current understanding and technological capabilities, several research directions show particular promise for advancing LIPG research:
Structure-function relationships:
High-resolution structural studies of LIPG, particularly focusing on the lid region and active site
Molecular dynamics simulations to understand substrate specificity and catalytic mechanism
Structure-guided design of specific inhibitors or activity modulators
Translational research opportunities:
Investigation of LIPG as a potential therapeutic target for dyslipidemia
Development of LIPG inhibitors or activators for modulating HDL metabolism
Exploration of LIPG genetic variants as biomarkers for cardiovascular risk assessment
Systems biology approaches:
Integration of LIPG into comprehensive models of lipoprotein metabolism
Network analysis of LIPG interactions with other proteins in lipid metabolism
Multi-omics studies to understand LIPG regulation across different physiological states
Emerging methodologies:
Application of CRISPR-Cas9 technology for precise genetic manipulation of LIPG
Development of novel biosensors for real-time monitoring of LIPG activity
Advanced imaging techniques to visualize LIPG-mediated HDL remodeling in living cells
Expanding physiological contexts:
Investigation of LIPG's role in inflammation and immune response
Exploration of tissue-specific functions beyond vascular endothelium
Studies of LIPG in pathological conditions beyond dyslipidemia
These research directions could significantly advance our understanding of LIPG biology and potentially lead to novel therapeutic strategies for lipid-related disorders .
Integrating LIPG research into the broader context of cardiovascular disease requires multidisciplinary approaches:
Mechanistic integration:
Investigate how LIPG-mediated HDL remodeling affects reverse cholesterol transport
Explore the relationship between LIPG activity and endothelial function
Examine how LIPG interacts with other risk factors for atherosclerosis
Genetic approaches:
Conduct genome-wide association studies to identify genetic variants that interact with LIPG SNPs
Perform Mendelian randomization studies to establish causal relationships between LIPG activity and cardiovascular outcomes
Clinical translation:
Correlate LIPG activity or genetic variants with clinical cardiovascular outcomes
Investigate LIPG as a potential biomarker for cardiovascular risk assessment
Explore therapeutic strategies targeting LIPG in cardiovascular disease prevention
Technological integration:
Utilize advanced imaging techniques to visualize LIPG effects on vascular function
Apply proteomics and metabolomics to understand LIPG's broader metabolic impact
Develop computational models integrating LIPG into cardiovascular risk prediction
Collaborative approaches:
Establish multidisciplinary research teams combining expertise in biochemistry, genetics, cardiology, and computational biology
Participate in large-scale consortia studying cardiovascular biomarkers and genetic risk factors
Share standardized protocols and resources to enhance reproducibility
By integrating LIPG research into this broader context, researchers can enhance the relevance and impact of their findings while contributing to our understanding of cardiovascular disease mechanisms .
Researchers working with LIPG Human, HEK should prioritize several methodological considerations to ensure robust and reproducible results:
Protein quality and handling: Store lyophilized protein at -20°C, prepare fresh working solutions, and avoid repeated freeze-thaw cycles to maintain activity .
Experimental design: Include appropriate controls, standardize experimental conditions, and validate findings across multiple experimental systems.
Technical expertise: Familiarize yourself with the specific characteristics of LIPG, including its glycosylation pattern, substrate specificity, and enzymatic properties.
Methodological rigor: Employ multiple complementary approaches to address research questions, as single methodologies may provide incomplete insights into LIPG function.
Contextual understanding: Consider the broader biological context of LIPG function in lipoprotein metabolism and cardiovascular biology when interpreting results.
By attending to these considerations, researchers can maximize the value of their work with LIPG Human, HEK and contribute meaningfully to our understanding of lipid metabolism and cardiovascular biology .
Lipase Endothelial (LIPG) is produced as a recombinant protein in HEK 293 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 protein is fused to a 6 amino acid His tag at the C-terminus, which facilitates its purification .
LIPG exhibits extensive phospholipase activity and is more active as a phospholipase than a triglyceride lipase . It hydrolyzes high-density lipoproteins (HDL) more efficiently than other lipoproteins, which is significant for its role in lipoprotein metabolism . Additionally, LIPG binds heparin, which may influence its activity and interactions within the vascular system .