CES1G (Carboxylesterase 1G) is a serine hydrolase enzyme belonging to the carboxylesterase family, primarily involved in hydrolyzing ester and amide bonds. In mice, it is encoded by the Ces1g gene (also known as Es-x) and plays critical roles in lipid metabolism, detoxification, and disease pathogenesis. CES1G is expressed in multiple tissues, including the liver, intestine, and macrophages, with its activity linked to cholesterol and triglyceride homeostasis .
Global Ces1g knockout (Ces1g−/−) mice exhibit:
Lowered LDL-C and HDL-C levels, with preserved liver lipid content
Attenuated atherosclerosis when crossed with Ldlr−/− mice (57% reduction in lesions)
Inhibited Intestinal Cholesterol Absorption: Ces1g−/− mice show 33–48% reduced cholesterol absorption via downregulation of Npc1l1 .
Enhanced Macrophage Cholesterol Efflux: Increased expression of ABCA1 and ABCG1 promotes cholesterol export .
M2 Macrophage Polarization: Elevated Arg1, Fizz1, and Ym1 mRNA levels in Ces1g−/− macrophages indicate anti-inflammatory polarization .
Key Discrepancy: Global Ces1g inactivation protects against atherosclerosis, while hepatic-specific deletion exacerbates metabolic dysfunction. This dichotomy highlights tissue-specific roles of CES1G in lipid metabolism .
Model | Purpose | Key Findings |
---|---|---|
Ces1g−/− | Assess systemic CES1G function | Reduced plasma cholesterol, no obesity |
Ces1g−/− Ldlr−/− (DKO) | Study atherosclerosis in hyperlipidemic mice | 57% fewer aortic lesions |
Ces1g−/− Apoe−/− | Evaluate hepatic CES1G’s role in atherogenesis | Aggravated hyperlipidemia and lesions |
Cholesterol Absorption: Measured via fecal cholesterol excretion or plasma radiolabeled cholesterol .
Lipoprotein Profiling: FPLC analysis to quantify VLDL, LDL, and HDL fractions .
Macrophage Polarization: In vitro differentiation with IFNγ/LPS (M1) or IL-4 (M2), followed by qRT-PCR for cytokine markers .
CES1G represents a dual-edged enzyme: its global inactivation may offer therapeutic benefits for atherosclerosis, while hepatic targeting could exacerbate metabolic disorders. Future research should explore tissue-specific CES1G inhibitors and their interactions with lipid-lowering drugs like statins.
Ces1g (also known as Es-x) is a member of the carboxylesterase family in mice that plays significant roles in lipid metabolism. It's important to note that while Ces1g was previously referred to simply as Ces1, it is not the functional ortholog of human CES1. The functional mouse ortholog of human CES1 has been demonstrated to be Ces1d, not Ces1g . The functional human ortholog for Ces1g has not yet been definitively established, which creates an important consideration when attempting to translate findings between mouse models and human applications.
Carboxylesterases are a family of enzymes that hydrolyze esters, amides, and thioesters. In mice, there are multiple Ces genes categorized into different families, with Ces1g belonging to the Ces1 family that includes Ces1a through Ces1g, each with distinct substrate specificities and tissue distribution patterns .
Ces1g plays crucial roles in lipid metabolism and homeostasis. Research has demonstrated that Ces1g is involved in:
Regulation of plasma cholesterol and triglyceride levels
Intestinal cholesterol and fat absorption processes
Macrophage cholesterol efflux
Expression of lipid transport proteins including Niemann-Pick C1 like 1
Regulation of macrophage polarization toward an M2 phenotype
Global inactivation of Ces1g in mice leads to reduced plasma cholesterol levels, suggesting its importance in cholesterol metabolism . Ces1g also appears to influence the expression of ATP-binding cassette subfamily A member 1 (ABCA1) and ABCG1, which are critical for cholesterol efflux from macrophages .
Ces1g expression appears to be regulated by multiple factors, with the Farnesoid X Receptor (FXR) playing a particularly important role. Studies suggest that Ces1g is a direct transcriptional target of FXR and may be involved in the regulation of liver lipid homeostasis by this nuclear receptor .
In FXR knockout mice, alterations in Ces1g expression contribute to changes in lipid metabolism, potentially influencing the development of metabolic disorders such as Metabolic-Associated Fatty Liver Disease (MAFLD) . The regulation of Ces1g may also involve other metabolic pathways, as suggested by transcriptomic analyses comparing FXR-knockout mice and human MAFLD samples .
When generating Ces1g knockout mice, researchers have successfully employed several approaches:
Conventional knockout strategies using homologous recombination
CRISPR/Cas9 system for targeted gene disruption
The CRISPR/Cas9 system has been effectively used to generate FXR knockout mice as described in the literature . A similar approach can be applied to Ces1g. When designing CRISPR guides, researchers should carefully consider potential off-target effects and confirm knockout efficiency through multiple validation methods, including:
Genomic PCR and sequencing to confirm the deletion or mutation
RT-qPCR to verify absence of mRNA transcripts
Western blotting to confirm protein absence
Functional assays to demonstrate loss of Ces1g enzymatic activity
When designing experiments to investigate Ces1g's role in atherosclerosis, researchers should consider the following methodological approaches:
Ces1g-deficient mice exhibit differential responses to dietary challenges, which provides valuable insights into the gene's metabolic functions:
When challenged with a Western diet (high fat, high cholesterol), Ces1g-/- Ldlr-/- double knockout mice demonstrate:
Decreased plasma cholesterol and triglyceride levels compared to Ldlr-/- controls
Reduced development of atherosclerotic lesions
Altered intestinal lipid absorption profiles
Changes in cholesterol efflux from macrophages
These dietary challenge studies reveal that Ces1g deficiency provides protection against diet-induced metabolic disorders, particularly by:
Inhibiting intestinal cholesterol and fat absorption
Reducing the expression of Niemann-Pick C1 like 1, a key cholesterol transporter
Increasing macrophage cholesterol efflux through upregulation of ABCA1 and ABCG1
Promoting M2 macrophage polarization
Inducing the expression of hepatic cholesterol 7α-hydroxylase and sterol 12α-hydroxylase
An intriguing contradiction exists in the literature regarding Ces1g's role in atherosclerosis depending on the model system used:
Global Ces1g knockout: In Ldlr-/- background, global Ces1g deficiency is atheroprotective, leading to:
Reduced plasma lipids
Decreased atherosclerotic lesion development
Enhanced macrophage cholesterol efflux
Inhibited intestinal cholesterol absorption
Liver-specific Ces1g knockdown: In contrast, when Ces1g is specifically knocked down in the liver of ApoE-/- mice, the opposite effect occurs:
Increased hyperlipidemia
Exacerbated Western diet-induced atherogenesis
This contradiction highlights the complex, tissue-specific roles of Ces1g in lipid metabolism. The differential effects may be explained by:
Tissue-specific functions of Ces1g in liver versus intestine
Compensatory mechanisms that may be activated in global knockout but not in tissue-specific knockdown
Potential differences in background strain interactions (Ldlr-/- versus ApoE-/-)
Differences between developmental knockout versus adult knockdown approaches
Researchers must carefully consider these contradictions when designing experiments and interpreting results from Ces1g mouse models.
Ces1g appears to be intimately connected with nuclear receptor signaling, particularly with the Farnesoid X Receptor (FXR) pathway:
Studies suggest that Ces1g is a direct target of FXR and is involved in the regulation of liver lipid homeostasis controlled by this nuclear receptor . When FXR is deleted in mice (FXR-KO), significant hepatic steatosis develops when mice are challenged with a high-fat diet, with increased fat droplet ballooning compared to wild-type mice .
The interplay between Ces1g and FXR may occur through several mechanisms:
Direct transcriptional regulation of Ces1g by FXR
Ces1g-mediated alterations in bile acid metabolism, which in turn affects FXR activation
Shared involvement in metabolic pathways related to fatty liver disease
Interestingly, while FXR knockout exacerbates steatosis in mice, human studies have shown that FXR expression is not significantly different between MAFLD patients and controls . This suggests that changes in FXR protein activity, rather than expression, may be important, or that Ces1g may be regulated by additional factors beyond FXR in humans.
For comprehensive transcriptomic analysis of Ces1g-deficient models, researchers should consider the following methodological approaches:
RNA isolation and quality control:
Extract total RNA from relevant tissues (liver, intestine, adipose)
Assess RNA quality using bioanalyzer (aim for RIN > 8)
Use appropriate RNA extraction methods depending on tissue type
RNA sequencing considerations:
Use paired-end sequencing for better transcript reconstruction
Aim for >20 million reads per sample for adequate coverage
Include appropriate biological replicates (minimum n=3 per group)
Bioinformatic analysis pipeline:
Map trimmed reads to reference genome (mouse GRCm38/mm10)
Perform differential expression analysis using established tools (DESeq2, edgeR)
Use stringent statistical thresholds (adjusted p-value < 0.05)
Validate key findings with RT-qPCR
Pathway analysis strategies:
Integration with public datasets:
Compare findings with publicly available datasets (e.g., GEO database)
Look for overlapping differentially expressed genes between mouse models and human samples
Focus on genes consistently altered across different datasets
The research by Ma et al. illustrates the power of this approach, identifying 134 overlapping genes between FXR-KO mice and MAFLD patients, with enrichment in metabolic pathways, retinol metabolism, oxidation-reduction processes, and lipid metabolic processes .
Validation of gene expression changes identified through transcriptomic analysis requires a multi-faceted approach:
RT-qPCR validation:
Design primers spanning exon-exon junctions to avoid genomic DNA amplification
Use appropriate reference genes (validate stability across experimental conditions)
Include technical and biological replicates
Present data as fold-change relative to control group
Protein-level validation:
Functional validation:
Histological validation:
Perform H&E staining to assess histological changes
Use immunohistochemistry with protein-specific antibodies
For steatosis assessment, employ specialized lipid stains
Translational validation:
Compare findings with human datasets (e.g., GSE48452, GSE63067)
Focus on genes that show consistent changes across species
Prioritize genes with established roles in relevant metabolic pathways
Research on Ces1g mouse models provides several important insights into human metabolic diseases:
The functional human ortholog for Ces1g has not been definitively established
Human carboxylesterase biology may differ significantly from mouse
Complex tissue-specific roles may require targeted therapeutic approaches
Current Ces1g mouse model research faces several important limitations that researchers should consider:
Orthology uncertainty:
The functional human ortholog for Ces1g remains undefined
Research should focus on identifying the human equivalent to better translate findings
Comparative genomics and functional studies across species could help resolve this issue
Strain background effects:
Different mouse strains may show variable Ces1g functions and phenotypes
Studies should be replicated across multiple genetic backgrounds
Backcrossing to congenic backgrounds may help standardize results
Tissue-specific contributions:
The contradictory findings between global and liver-specific Ces1g manipulation highlight the need for comprehensive tissue-specific studies
Development of conditional knockout models for specific tissues (intestine, adipose, macrophage) would help resolve tissue-specific roles
Sex-specific differences:
Many studies use only male mice, potentially missing sex-specific effects
Future research should include both sexes and analyze results separately
Hormonal influences on Ces1g expression and function should be investigated
Diet composition variability:
Different studies use varied "Western diet" or "high-fat diet" compositions
Standardization of diet compositions or systematic testing of different diets would improve comparability between studies
Time-course studies would help understand adaptive responses to dietary challenges
Carboxylesterase 1G (CES1G) is a member of the carboxylesterase family, which is a group of enzymes involved in the hydrolysis of ester and amide bonds. These enzymes play a crucial role in the metabolism of various xenobiotic and endogenous compounds, including drugs, environmental toxicants, and lipids .
Carboxylesterases are classified based on their substrate specificity and sequence homology. In mice, there are multiple carboxylesterase genes, including CES1G, which share significant sequence similarity with human CES1 . CES1G is one of the key enzymes in the mouse liver, involved in lipid metabolism and detoxification processes .
The primary function of CES1G is to hydrolyze ester and amide bonds in xenobiotic and endogenous compounds. This activity is essential for the detoxification of drugs and environmental toxicants, as well as the metabolism of lipids . CES1G also plays a role in energy homeostasis by regulating the hydrolysis of lipid esters .
CES1G catalyzes the hydrolysis of ester and amide bonds through a serine hydrolase mechanism. The enzyme’s active site contains a serine residue that forms a covalent intermediate with the substrate, leading to the cleavage of the ester or amide bond . This reaction results in the formation of an alcohol and an acid, which are then further metabolized by other enzymes .
The expression and activity of CES1G are regulated by various factors, including genetic and environmental influences. In mice, the expression of CES1G can be modulated by dietary factors, hormones, and exposure to xenobiotics . Additionally, genetic variations in the CES1G gene can affect the enzyme’s activity and substrate specificity .
Recombinant CES1G is widely used in research to study the enzyme’s role in drug metabolism and lipid metabolism. The recombinant enzyme is produced in various expression systems, including bacterial and mammalian cells, to ensure high purity and activity . Researchers use recombinant CES1G to investigate the enzyme’s substrate specificity, catalytic mechanism, and potential as a therapeutic target for metabolic diseases .