Article
Review Article
Korean J Gastroenterol 2025; 85(1): 11-21
Published online January 25, 2025 https://doi.org/10.4166/kjg.2024.135
© The Korean Society of Gastroenterology.
Relationship between Abnormal Lipid Metabolism and Gallstone Formation
Xiang Li1,2,3*, Xiaodan Yin1,2,3*, Jun Xu1,2,3, Lei Geng1,2,3 , Zhengtao Liu4
Correspondence to: Lei Geng, Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, No. 366, wutong Road, Xihu District, Hangzhou City, Zhejiang Province, 310003, China. Tel: +86 13777802607, Fax: +86 571 87236114, E-mail:geng97927@163.com, ORCID: https://orcid.org/0009-0006-8693-5721
Zhengtao Liu, Shulan International Medical College, Zhejiang Shuren University, No. 8 Shuren Street, Gongshu District, Hangzhou, Zhejiang Province, 310000, China. Tel: +86 15289668420, Fax: +86 571 87236570, Email: liuzhengtao@zjsru.edu.cn, ORCID:: https://orcid.org/0000-0002-6232-4778
*These authors contributed equally to this work.
This is an open access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
Abstract
Cholelithiasis is a common biliary system disease with a high incidence worldwide. Abnormal lipid metabolism has been shown to play a key role in the mechanism of gallstones. Therefore, recent research literature on the genes, proteins, and molecular substances involved in lipid metabolism during the pathogenesis of gallstones has been conducted. This study aimed to determine the role of lipid metabolism in the pathogenesis of gallstones and provide insights for future studies using previous research in genomics, metabolomics, transcriptomics, and other fields.
KeywordsGallstones; Lipid metabolism; Triglycerides; Phospholipids; Metabolomics
INTRODUCTION
Gallstone disease (GSD) is a common biliary system disease composed primarily of cholesterol stones, pigment stones (composed mainly of calcium salts and bile pigments), and mixed stones.1 Cholecystolithiasis tends to occur in adults with an overall incidence of 10–15%. The incidence increases with age after 40 years, with a higher incidence in women than men.2
Three main types of gallstones exist: cholesterol, pigmented, and mixed.3 Most gallstones are cholesterol gallstones, accounting for approximately 90%.4 The symptoms of gallstones are usually caused by cholesterol stones.5
Many studies have revealed a close relationship between gallstone formation and human metabolic activity.6 The high expression of beta3-adrenergic receptors and HMG-CoA reductase in obese patients7 can affect gallbladder contraction and inhibit cholesterol excretion in the gallbladder.8,9 In metabolic syndrome, the low high-density lipoprotein,10 elevated triglyceride, and high homocysteine levels lead to a pro-inflammatory state that increases the rate of gallstone formation.11,12 The abnormal metabolism of bile acids in the human body can also affect cholesterol excretion in the bile.13
Gallstone formation is closely related to human metabolism, particularly lipid metabolism. Therefore, studying lipid metabolism will help better understand the mechanism of gallbladder stone formation.
LIPID METABOLISM
Lipids consist of triglycerides (TGs), phospholipids, and cholesterol. They are important nutrients required by the human body, providing energy and essential fatty acids and participating in cell composition. Fatty acids are the most common form of energy storage in the human body. They are stored mainly in the form of TGs.14 Phospholipids and cholesterol are important components of the cell membranes.15
The liver is the main site of lipid synthesis and metabolism.10,14 The synthesis and metabolism of lipids in the normal human body are balanced. Disruption of this balance leads to various diseases such as obesity, atherosclerosis, and type 2 diabetes.16,17
1. Cholesterol metabolism
Cholesterol plays an important role in the cell membrane composition, intracellular signaling, and the synthesis of various hormones.18 Acetyl-CoA is a raw material for cholesterol biosynthesis. It is synthesized in cells through the synergistic action of more than 20 enzymes, e.g., HMG-CoA reductase, exerting its biological functions.19,20 Cholesterol biosynthesis is related to the levels of foam sterol, cholestanol, and desfilin.21,22 The liver is the primary site of cholesterol biosynthesis, transmitting endogenous and exogenous cholesterol as extremely low-density lipoproteins (VLDL) into the bloodstream. Excess cholesterol combines with apolipoprotein A-I (apoA-I) to form high-density lipoprotein (HDL).23,24 Nuclear receptor liver X receptors (LXRs) are important regulatory factors for intracellular cholesterol and lipid homeostasis.25-27
Cholesterol can produce various oxidative sterols via enzymatic and non-enzymatic pathways.28 Some are further metabolized into bile acids in the liver and discharged into the gallbladder, becoming a component of bile.19 The stability of the cholesterol metabolism has a regulatory effect on the inflammatory response and tumor microenvironment.29,30 Moreover, some cholesterol molecules, such as SOAT1, SQLE, and NPC1, have become immunotherapeutic targets for tumors (Fig. 1).31-33
-
Figure 1. In vivo synthesis and metabolic pathways of cholesterol.
2. Triglyceride metabolism
TGs are the main source of energy in the human body and are synthesized mainly in the liver via the diglyceride pathway using sugars, glycerol, and fatty acids.14,34 The liver takes up excess glucose and fructose35-37 through de novo lipogenesis and converts them into TGs.38 The activation of transcription factor 7-like 2 (TCF7L2)39 and liver cell membrane CD36 increases the intake of fatty acids, promoting de novo lipogenesis.40 The action of insulin41,42 and the secretion of the hepatokine oromucoid II limits the progression of de novo lipogenesis.43 Small intestinal mucosal cells can synthesize TGs using glycerol monoesters and fatty acids absorbed through exogenous digestion.
The lipoprotein lipase protein complex (LPL) determines the breakdown of TGs, and liver receptors (RLDL and LRP-1) are responsible for transporting and clearing the decomposed products.44 Dietary habits45,46 and the size of the TG decomposition products and intermediates,47 such as lipid droplets,48 can influence the activity of fatty acid TG lipase, affecting TG metabolism.
3. Phospholipid metabolism
Phospholipids are synthesized in the endoplasmic reticulum, where TGs and phosphatidic acids bind to sphingosine to form a phospholipid sphingosine complex. The complex then combines with phosphate and choline to form phospholipid molecules such as lecithin and cephalin.49 Some fatty acids, inositol, and enzymes can modify phospholipid molecules to perform various biological functions.50 In cells, phospholipids are transported and transmitted through vesicles and cell membranes.51 The roles of phosphoinositol and phospholipase D in vesicular transport reactions are important.52-54
Phospholipids can be hydrolyzed into glycerol, fatty acids, phosphate, and various amino alcohols through phospholipase reactions in living organisms. Phospholipases A1 and A2 cleave fatty acids at the 1,2 positions of phospholipid molecules. In addition, the phospholipase D (PLD) enzyme catalyzes the hydrolysis of phosphodiesterase bonds in glycerol phospholipids.54,55 Sphingosine 1-phosphate (S1P) is a metabolic product of cell membrane sphingolipids, which bind to the S1P receptor (S1PR) coupled with the G protein to regulate embryonic development, organ function, inflammatory response, and multiple system diseases.56-59 The ABCB4 gene, cleavage of lipoproteins, and structural activity of the endoplasmic reticulum have regulatory effects on the biosynthesis and metabolism of phospholipids (Fig. 2).60-63
-
Figure 2. Key enzymes for phospholipid synthesis and metabolism in vivo.
ABNORMAL LIPID METABOLISM AND GALLSTONES
Bile is a three-variable solution; the bile acid, lecithin, and cholesterol concentration ratio in bile affects cholesterol solubility.64 Under normal circumstances, bile salts and lecithin can disperse cholesterol to form soluble microcapsules, which are less likely to precipitate and form stones. Bile cholesterol is more likely to become supersaturated when cholesterol secretion in bile increases or the concentration of bile acids and lecithin decreases.13
1. Cholesterol metabolism and bile biochemistry
Bile acids mainly pass through cholesterol 7 α-hydroxylase (CYP7A1), which oxidizes cholesterol. The regulatory effect of the nuclear factor farnesoid X receptor (FXR) is a key factor.13 FXR downregulates CYP7A1 by inducing a cascade reaction involving human fibroblast growth factor 19-JNK.65,66 Fibrin can increase the risk of gallstones 1.7-fold.67 CYP7A1 polymorphisms also lead to a significantly higher risk of developing gallstones.66 Cholesterol precipitates and forms gallstones when cholesterol synthesis is excessively active and cholesterol in the bile is supersaturated.9 CYP7A1 is inhibited when FXR is overactive, which affects cholesterol breakdown and metabolism into bile acids, further promoting cholesterol accumulation and precipitation.66,68
Some receptors on the gallbladder wall regulate cholesterol absorption, such as scavenger receptor SR-BI.69 This receptor is related to the occurrence of atherosclerosis, and the supersaturation of bile cholesterol appears to be related to SR-BI receptor inhibition.70-72
This study suggests that abnormalities in CYP7A1, FXR, and SR-BI can lead to metabolic disorders of cholesterol in bile, accelerating the formation of gallstones. On the other hand, Chen et al. found through Mendelian randomization analysis that the association between the serum total cholesterol (TC) levels and the risk of gallstones was not linear; lower TC levels lead to a higher risk of stone formation.73
2.Triglyceride metabolism and bile biochemistry
Currently, a large number of clinical studies have shown that patients with hyperlipidemia have a higher risk of developing gallstones. Previous studies have shown that knocking out CD36 on the liver cell membrane, which plays a regulatory role in the de novo lipogenesis pathway, significantly reduces the incidence of gallstones in mice.74 Elevated levels of very low-density lipoprotein (VLDL) related to TG transport can also accelerate the excretion of bile acids.75 Evidence suggests that the gallbladder of high TG patients is less sensitive to CCK. This sensitivity is improved after reversing the high serum TG levels with TG-lowering agents.76 These results confirmed the correlation between the increase in TG and gallstones.
3. Phospholipid metabolism and bile biochemistry
Phospholipids are important biochemical components of bile. The solubility of cholesterol in bile salt phospholipid mixtures is much higher than in pure bile salts.77 Therefore, phospholipids play an important role in the biochemical balance of bile. Phospholipids are generated mainly in the endoplasmic reticulum, with sphingomyelin synthetase being the key enzyme for their synthesis. Abnormal enzyme levels can cause non-alcoholic fatty liver, atherosclerosis, and diabetes.78,79 On the other hand, no reports on the relationship between this enzyme abnormality and the occurrence of gallstones have been published.
The most extensively studied phospholipid transport process is the endoplasmic reticulum-mitochondria encounter structure (ERMES) complex between the endoplasmic reticulum and mitochondria, which is a critical pathway for phospholipid transfer between two organelles.80 It is unclear if there is a connection between this pathway and gallstone formation. Low phospholipid-associated cholelithiasis (LPAC) is an inherited cholelithiasis associated with low phospholipid levels in the bile, accounting for approximately 0.5–2% of symptomatic cholelithiasis.81 LPAC is a genetic disease caused by a single gene mutation associated with mutations in ABCB4, which mainly encodes the carrier protein MDR3 in the biliary tract.61,82 MDR3 exists on the membrane of liver tubules; its main function is to transport phosphatidylcholine from liver cells to the gallbladder. When the MDR3 protein mutates, phospholipid transport disorders lead to easier cholesterol precipitation in bile (Fig. 3).83 In summary, abnormal phospholipid generation and transport caused by various factors can lead to bile cholesterol supersaturation, accelerating gallstone formation.
-
Figure 3. Effects of MDR3 mutations on phospholipid transport on the membrane of hepatic tubular cells.
4. Lipoproteins and bile biochemistry
Many studies have examined the relationship between the HDL and LDL levels in the serum and bile metabolism, as well as the formation of gallstones. A study on the correlation between serum lipoprotein levels and gallstone formation in Western patients with gallstones found that high LDL levels can increase the cholesterol concentration in bile (p<0.01) and increase the incidence of gallstones (p<0.05).84 HDL-C is positively correlated with phospholipids and bile acids in the bile. Hence, high LDL levels can accelerate the formation of gallstones, whereas HDL-C may offer some protection for gallstone formation.85
5. Lipid metabolism and gallbladder contraction disorders
Under normal conditions, the gallbladder can drive the intestinal hepatic circulation of bile acids through regular contraction and relaxation.86 This process is regulated by cholecystokinin (CCK) secreted by small intestinal cells.87 Dietary fat can stimulate CCK secretion to some extent. Active cholesterol synthesis leads to a sustained supersaturation state, resulting in the deposition of cholesterol on the gallbladder wall, COX2-mediated inflammation, and the abnormal contraction and relaxation of the gallbladder.10,88 Gallbladder contraction disorders can affect the excretion of cholesterol and bile acids, increasing the risk of gallstones.
OMICS TECHNIQUES AND GALLSTONES
Omics technology refers to the detection and analysis of data representing the structure and function of a specific biological system, such as genes and proteins, and the acquisition of systematic connections between the data at that level. The technology mainly includes genomics, proteomics, metabolomics, transcriptomics, lipidomics, immunohistochemistry, glycomics, RNA omics, imaging omics, and ultrasound omics.89 The integration of multiple omics has become a common trend in constructing comprehensive causal relationships, with genomics, transcriptomics, proteomics, and metabolomics being the most widely used approaches.
Genomic research measures DNA molecules, whereas epigenomics, transcriptomics, proteomics, and metabolomics research measures the chemical states of DNA and its binding proteins, RNA, proteins, and metabolites.90 Compared to traditional singularity research, the basic status of genes, RNA, proteins, and metabolites in the target sample can be understood by integrating and analyzing genomic, transcriptomic, proteomic, and metabolomic data, providing a comprehensive interpretation of the biological system.
1. Genomic research on gallstones
Research on the gallbladder genome has found that multiple sets of alleles in the gallbladder tissue of patients with stones have undergone mutations, which have been isolated and localized. These include mutations in the liver cholesterol transporter (ABC transporter) ABCG5/ABCG8,91,92 CYP7A1, and changes in the multicellular transmembrane protein NPC1L1 (Niemann Pick C1 Like 1) are associated with the intestinal and liver reabsorption of cholesterol.93
ABCG5/ABCG8 are present in intestinal and liver cells to inhibit the intestinal absorption of cholesterol and promote bile secretion.92 NAPC1L1 is present in intestinal epithelial and liver cells, promoting cholesterol absorption through endocytosis mediated by grid proteins.94 Changes in these genes and proteins promote cholesterol absorption in the human body, inhibit cholesterol transport and bile excretion, and accelerate the nucleation of cholesterol in bile.
The treatment of gallstones is mainly surgical, but early prevention and intervention in gallstones can be achieved by exploring the omics mechanisms of gallstone formation. Currently, in vitro experiments have been conducted to regulate the cholesterol levels in rats through the mediation of NAPC1L1 and ABCG5/ABCG8 proteins by lactic acid bacteria, garlic extract, or Diosgenin.95-97 In addition, NPC1L1 is the target of the lipid-lowering drug ezetimibe in the intestine,98 which may contribute to the early prevention of gallstones.
2. Transcriptomics and Gallstones
Transcriptomic research on gallbladder diseases is progressing rapidly, including the study of genes related to the onset of single diseases such as gallstones, gallbladder polyps, and gallbladder cancer.
In a study of gallstones, a correlation was found between miR-20 (RNA-regulating ABC transporter) and the occurrence of gallstones through the transcriptome analysis of gallbladder tissue.99 This confirms the results of the genomic research on ABC transporter proteins. Aquaporin 3 (AQP3) can reduce gallbladder inflammation and inhibit gallstone formation.100 miR-20 and AQP3 also play important roles in the lipid metabolism. Changes in miR-20 and AQP3 can affect the transportation of cholesterol and TG, promoting the formation of gallstones. A research team from the Netherlands conducted transcriptomic studies on adipose tissue of patients with gallbladder cancer. They revealed a significant increase in apolipoprotein A1 (APOA1) involvement in patients with stones. This can lead to the release of beneficial cholesterol from adipose tissue, resulting in the supersaturation of bile cholesterol.101 Infliximab (IFX) reduces miR-20 expression in children, and research in this area may provide a reference for treating gallstones.102
3. Metabolomics and Gallstones
Metabolomic research on gallstones has focused mainly on differential metabolites. By comparing the plasma or bile metabolism of patients with gallstones and normal individuals, some studies have found that cholesterol and various bile acids are significantly elevated in patients with gallstones.101,103 Relatively little research has been conducted on other metabolites and metabolic pathways related to stone formation.
In an unpublished study, plasma samples were collected from more than 100 patients, and non-targeted metabolomic testing was performed. The detection data were compared with the clinical indicators. The metabolites and metabolic pathways related to GSD generation were obtained using a weighted correlation network analysis (WGCNA) algorithm combined with KEGG pathway enrichment analysis.
The present study found a significant decrease in linoleic acid in the peripheral blood of patients with GSD. Moreover, its corresponding linoleic acid metabolic pathway also showed a significant downregulation trend. The pantothenate and CoA biosynthesis and TCA cycle pathways were significantly upregulated. Linoleic acid is a raw material used to synthesize and convert phospholipids. When the peripheral blood linoleic acid level decreases, there is an obstacle in the production of phospholipids in bile, and the precipitation of cholesterol becomes more pronounced. The pantothenate and CoA bio-synthesis pathways were upregulated in the peripheral blood of patients with GSD. Upregulation of this pathway will increase CoA production, the raw material for cholesterol synthesis, promoting cholesterol production.23,104 The TCA cycle is the oxidation pathway of nutrients in the body. The strong expression of this pathway can also promote the conversion of carbohydrates to lipids during satiety, increase the burden of lipids in the body, affect the lipid structure of the body, and promote the precipitation of cholesterol in bile (Fig. 4).105 Based on this study, GSD can be prevented and treated by affecting the linoleic acid cycle, and the effectiveness of drugs, such as ethyl linoleate, may serve as a direction for future research.
-
Figure 4. Pathway of TCA cycle in the formation of gallstones.
CONCLUSION
The abnormal metabolism of cholesterol, TGs, and phospholipids plays a significant role in gallstone formation. When FXR is overactivated, or CYP7A1 and SR-BI receptors are inhibited to a certain extent, cholesterol in bile becomes supersaturated, and a COX2-mediated inflammatory response is induced in the gallbladder wall, accelerating gallstone formation. The transport of TGs by VLDL and CD36 molecules in the liver cell membrane is affected, increasing the risk of gallstones. In the phospholipid metabolism, abnormalities in the MDR3 protein that regulates phospholipid transfer cause low phospholipid-related cholelithiasis.
In the genomic and transcriptome analysis of gallbladder tissue, changes in the liver cholesterol transporters ABCG5/ ABCG8 accelerate cholesterol nucleation, while abnormalities in aquaporin 3 (AQP3) and apolipoprotein A1 (APOA1) affect the release and transport of cholesterol and TGs (Fig. 5). Through metabolomic techniques, this study discovered that the linoleic acid metabolism pathway, pantothenic acid and coenzyme A biosynthesis pathways, and the TCA cycle pathway play a role in the generation and balance of phospholipids and cholesterol. These omics-related studies provide a new theoretical basis for preventing and treating gallstones and directions for future research.
-
Figure 5. Positions and roles of CD36, APOA-1, SR-BI, and CYP7A1 in the KEGG pathway of cholesterol synthesis.
Financial support
None.
Conflict of interest
None.
References
- Lammert F, Gurusamy K, Ko CW, et al. Gallstones. Nat Rev Dis Primers 2016;2:16024.
- Chen CH, Huang MH, Yang JC, et al. Prevalence and risk factors of gallstone disease in an adult population of Taiwan: an epidemiological survey. J Gastroenterol Hepatol 2006;21:1737-1743.
- Qiao T, Ma RH, Luo XB, Yang LQ, Luo ZL, Zheng PM. The systematic classification of gallbladder stones. PLoS One 2013;8:e74887.
- Tazuma S. Gallstone disease: Epidemiology, pathogenesis, and classification of biliary stones (common bile duct and intra-hepatic). Best Pract Res Clin Gastroenterol 2006;20:1075-1083.
- Schafmayer C, Hartleb J, Tepel J, et al. Predictors of gallstone composition in 1025 symptomatic gallstones from Northern Germany. BMC Gastroenterol 2006;6:36.
- Portincasa P, Moschetta A, Palasciano G. Cholesterol gallstone disease. Lancet 2006;368:230-239.
- Chen L, Yang H, Li H, He C, Yang L, Lv G. Insights into modifiable risk factors of cholelithiasis: A Mendelian randomization study. Hepatology 2022;75:785-796.
- Klass DM, Lauer N, Hay B, Kratzer W, Fuchs M; EMIL Study Group. Arg64 variant of the beta3-adrenergic receptor is associated with gallstone formation. Am J Gastroenterol 2007;102:2482-2487.
- Reshetnyak VI. Concept of the pathogenesis and treatment of cholelithiasis. World J Hepatol 2012;4:18-34.
- Itani M, Dubinsky TJ. Physical chemistry of bile: Detailed pathogenesis of cholelithiasis. Ultrasound Q 2017;33:229-236.
- Santana-Gálvez J, Cisneros-Zevallos L, Jacobo-Velázquez DA. Chlorogenic acid: Recent advances on its dual role as a food additive and a nutraceutical against metabolic syndrome. Molecules 2017;22:358.
- Worthington HV, Hunt LP, McCloy RF, Ubbink JB, Braganza JM. Dietary antioxidant lack, impaired hepatic glutathione reserve, and cholesterol gallstones. Clin Chim Acta 2004;349:157-165.
- Fremont-Rahl JJ, Ge Z, Umana C, et al. An analysis of the role of the indigenous microbiota in cholesterol gallstone pathogenesis. PLoS One 2013;8:e70657.
- Alves-Bezerra M, Cohen DE. Triglyceride metabolism in the liver. Compr Physiol 2017;8:1-8.
- Tamura Y, Sesaki H, Endo T. Phospholipid transport via mitochondria. Traffic 2014;15:933-945.
- DeBose-Boyd RA. Significance and regulation of lipid metabolism. Semin Cell Dev Biol 2018;81:97.
- Jones JG. Hepatic glucose and lipid metabolism. Diabetologia 2016;59:1098-1103.
- Cortes VA, Busso D, Maiz A, Arteaga A, Nervi F, Rigotti A. Physiological and pathological implications of cholesterol. Front Biosci (Landmark Ed) 2014;19:416-428.
- Luo J, Yang H, Song BL. Mechanisms and regulation of cholesterol homeostasis. Nat Rev Mol Cell Biol 2020;21:225-245.
- Cholesterol absorption versus cholesterol synthesis in man. Nutr Rev 1970;28:11-15.
- Márk L, Paragh G. Change in the cholesterol metabolism associated with the combined inhibition of synthesis and absorption. Orv Hetil 2007;148:627-632.
- Schroepfer GJ Jr. Oxysterols: modulators of cholesterol metabolism and other processes. Physiol Rev 2000;80:361-554.
- Phillips MC. Molecular mechanisms of cellular cholesterol efflux. J Biol Chem 2014;289:24020-24029.
- Zhang Q, Ke Y, Hong H. HDL and Lipid Metabolism. Adv Exp Med Biol 2022;1377:49-61.
- Bilotta MT, Petillo S, Santoni A, Cippitelli M. Liver X receptors: Regulators of cholesterol metabolism, inflammation, autoimmunity, and cancer. Front Immunol 2020;11:584303.
- Nazih H, Bard JM. Cholesterol, oxysterols and LXRs in breast cancer pathophysiology. Int J Mol Sci 2020;21:1356.
- Uppal H, Zhai Y, Gangopadhyay A, et al. Activation of liver X receptor sensitizes mice to gallbladder cholesterol crystallization. Hepatology 2008;47:1331-1342.
- Luu W, Sharpe LJ, Capell-Hattam I, Gelissen IC, Brown AJ. Oxysterols: Old tale, new twists. Annu Rev Pharmacol Toxicol 2016;56:447-467.
- Cardoso D, Perucha E. Cholesterol metabolism: a new molecular switch to control inflammation. Clin Sci (Lond) 2021;135:1389-1408.
- Xu H, Zhou S, Tang Q, Xia H, Bi F. Cholesterol metabolism: New functions and therapeutic approaches in cancer. Biochim Biophys Acta Rev Cancer 2020;1874:188394.
- Oni TE, Biffi G, Baker LA, et al. SOAT1 promotes mevalonate pathway dependency in pancreatic cancer. J Exp Med 2020;217:e20192389.
- Sun H, Li L, Li W, et al. p53 transcriptionally regulates SQLE to repress cholesterol synthesis and tumor growth. EMBO Rep 2021;22:e52537.
- O'Neill KI, Kuo LW, Williams MM, et al. NPC1 confers metabolic flexibility in triple negative breast cancer. Cancers (Basel) 2022;14:3543.
- Weiss SB, Kennedy EP, Kiyasu JY. The enzymatic synthesis of triglycerides. J Biol Chem 1960;235:40-44.
- Sanders FW, Griffin JL. De novo lipogenesis in the liver in health and disease: more than just a shunting yard for glucose. Biol Rev Camb Philos Soc 2016;91:452-468.
- Softic S, Cohen DE, Kahn CR. Role of dietary fructose and hepatic de novo lipogenesis in fatty liver disease. Dig Dis Sci 2016;61:1282-1293.
- Geidl-Flueck B, Hochuli M, Németh Á, et al. Fructose- and sucrose- but not glucose-sweetened beverages promote hepatic de novo lipogenesis: A randomized controlled trial. J Hepatol 2021;75:46-54.
- Ameer F, Scandiuzzi L, Hasnain S, Kalbacher H, Zaidi N. De novo lipogenesis in health and disease. Metabolism 2014;63:895-902.
- Lee DS, An TH, Kim H, et al. Tcf7l2 in hepatocytes regulates de novo lipogenesis in diet-induced non-alcoholic fatty liver disease in mice. Diabetologia 2023;66:931-954.
- Zeng H, Qin H, Liao M, et al. CD36 promotes de novo lipogenesis in hepatocytes through INSIG2-dependent SREBP1 processing. Mol Metab 2022;57:101428.
- Kawano Y, Cohen DE. Mechanisms of hepatic triglyceride accumulation in non-alcoholic fatty liver disease. J Gastroenterol 2013;48:434-441.
- Smith U, Kahn BB. Adipose tissue regulates insulin sensitivity: role of adipogenesis, de novo lipogenesis and novel lipids. J Intern Med 2016;280:465-475.
- Zhou B, Luo Y, Ji N, Hu C, Lu Y. Orosomucoid 2 maintains hepatic lipid homeostasis through suppression of de novo lipogenesis. Nat Metab 2022;4:1185-1201.
- Ibarretxe D, Masana L. Triglyceride metabolism and classification of hypertriglyceridemias. Clin Investig Arterioscler 2021;33 Suppl 2:1-6.
- Kersten S. The impact of fasting on adipose tissue metabolism. Biochim Biophys Acta Mol Cell Biol Lipids 2023;1868:159262.
- Barrows BR, Parks EJ. Contributions of different fatty acid sources to very low-density lipoprotein-triacylglycerol in the fasted and fed states. J Clin Endocrinol Metab 2006;91:1446-1452.
- Cerk IK, Wechselberger L, Oberer M. Adipose triglyceride lipase regulation: An overview. Curr Protein Pept Sci 2018;19:221-233.
- Schott MB, Weller SG, Schulze RJ, et al. Lipid droplet size directs lipolysis and lipophagy catabolism in hepatocytes. J Cell Biol 2019;218:3320-3335.
- Fyrst H, Saba JD. An update on sphingosine-1-phosphate and other sphingolipid mediators. Nat Chem Biol 2010;6:489-497.
- Alb JG Jr, Kearns MA, Bankaitis VA. Phospholipid metabolism and membrane dynamics. Curr Opin Cell Biol 1996;8:534-541.
- Moreau P, Cassagne C. Phospholipid trafficking and membrane biogenesis. Biochim Biophys Acta 1994;1197:257-290.
- Dickson EJ. Phosphoinositide transport and metabolism at membrane contact sites. Biochim Biophys Acta Mol Cell Biol Lipids 2022;1867:159107.
- Fruman DA, Meyers RE, Cantley LC. Phosphoinositide kinases. Annu Rev Biochem 1998;67:481-507.
- McDermott MI, Wang Y, Wakelam MJO, Bankaitis VA. Mammalian phospholipase D: Function, and therapeutics. Prog Lipid Res 2020;78:101018.
- Dekker N. Outer-membrane phospholipase A: known structure, unknown biological function. Mol Microbiol 2000;35:711-717.
- Cartier A, Hla T. Sphingosine 1-phosphate: Lipid signaling in pathology and therapy. Science 2019;366:eaar5551.
- Verstockt B, Vetrano S, Salas A, Nayeri S, Duijvestein M, Vande Casteele N; Alimentiv Translational Research Consortium (ATRC). Sphingosine 1-phosphate modulation and immune cell trafficking in inflammatory bowel disease. Nat Rev Gastroenterol Hepatol 2022;19:351-366.
- Jozefczuk E, Guzik TJ, Siedlinski M. Significance of sphingosine-1-phosphate in cardiovascular physiology and pathology. Pharmacol Res 2020;156:104793.
- Obinata H, Hla T. Sphingosine 1-phosphate and inflammation. Int Immunol 2019;31:617-625.
- Erlinger S. Low phospholipid-associated cholestasis and cholelithiasis. Clin Res Hepatol Gastroenterol 2012;36 Suppl 1:S36-40.
- Wang HH, Portincasa P, Liu M, Wang DQ. Genetic analysis of ABCB4 mutations and variants related to the pathogenesis and pathophysiology of low phospholipid-associated cholelithiasis. Genes (Basel) 2022;13:1047.
- Sam PN, Avery E, Claypool SM. Proteolytic control of lipid metabolism. ACS Chem Biol 2019;14:2406-2423.
- Nishimura T, Stefan CJ. Specialized ER membrane domains for lipid metabolism and transport. Biochim Biophys Acta Mol Cell Biol Lipids 2020;1865:158492.
- Coleman R. Bile salts and biliary lipids. Biochem Soc Trans 1987;15 Suppl:68S-80S.
- Portincasa P, Ciaula AD, Bonfrate L, Wang DQ. Therapy of gallstone disease: What it was, what it is, what it will be. World J Gastrointest Pharmacol Ther 2012;3:7-20.
- Claudel T, Zollner G, Wagner M, Trauner M. Role of nuclear receptors for bile acid metabolism, bile secretion, cholestasis, and gallstone disease. Biochim Biophys Acta 2011;1812:867-878.
- Caroli-Bosc FX, Le Gall P, Pugliese P, et al. Role of fibrates and HMG-CoA reductase inhibitors in gallstone formation: epidemiological study in an unselected population. Dig Dis Sci 2001;46:540-544.
- Hu H, Shao W, Liu Q, et al. Gut microbiota promotes cholesterol gallstone formation by modulating bile acid composition and biliary cholesterol secretion. Nat Commun 2022;13:252.
- Yu H. HDL and scavenger receptor class B type I (SRBI). Adv Exp Med Biol 2022;1377:79-93.
- Hermann S, Kuhlmann MT, Starsichova A, et al. Imaging reveals the connection between spontaneous coronary plaque ruptures, atherothrombosis, and myocardial infarctions in HypoE/SRBI-/- Mice. J Nucl Med 2016;57:1420-1427.
- Miquel JF, Moreno M, Amigo L, et al. Expression and regulation of scavenger receptor class B type I (SR-BI) in gall bladder epithelium. Gut 2003;52:1017-1024.
- Rigotti A, Zanlungo S, Miquel JF, Wang DQ. HDL receptor SR-BI and cholesterol gallstones. Hepatology 2002;35:240-242.
- Chen L, Qiu W, Sun X, et al. Novel insights into causal effects of serum lipids and lipid-modifying targets on cholelithiasis. Gut 2024;73:521-532.
- Xie Y, Cifarelli V, Pietka T, et al. Cd36 knockout mice are protected against lithogenic diet-induced gallstones. J Lipid Res 2017;58:1692-1701.
- Castro J, Amigo L, Miquel JF, et al. Increased activity of hepatic microsomal triglyceride transfer protein and bile acid synthesis in gallstone disease. Hepatology 2007;45:1261-1266.
- Smelt AH. Triglycerides and gallstone formation. Clin Chim Acta 2010;411:1625-1631.
- Hofmann AF, Schteingart CD, vanSonnenberg E, Esch O, Zakko SF. Contact dissolution of cholesterol gallstones with organic solvents. Gastroenterol Clin North Am 1991;20:183-199.
- Koh EH, Yoon JE, Ko MS, et al. Sphingomyelin synthase 1 mediates hepatocyte pyroptosis to trigger non-alcoholic steatohepatitis. Gut 2021;70:1954-1964.
- Chen Y, Cao Y. The sphingomyelin synthase family: proteins, diseases, and inhibitors. Biol Chem 2017;398:1319-1325.
- Endo T, Tamura Y, Kawano S. Phospholipid transfer by ERMES components. Aging (Albany NY) 2018;10:528-529.
- Dong C, Condat B, Picon-Coste M, et al. Low-phospholipid-associated cholelithiasis syndrome: Prevalence, clinical features, and comorbidities. JHEP Rep 2020;3:100201.
- Goubault P, Brunel T, Rode A, Bancel B, Mohkam K, Mabrut JY. Low-Phospholipid Associated Cholelithiasis (LPAC) syndrome: A synthetic review. J Visc Surg 2019;156:319-328.
- Gonzales E, Davit-Spraul A, Baussan C, Buffet C, Maurice M, Jacquemin E. Liver diseases related to MDR3 (ABCB4) gene deficiency. Front Biosci (Landmark Ed) 2009;14:4242-4256.
- Atamanalp SS, Keles MS, Atamanalp RS, Acemoglu H, Laloglu E. The effects of serum cholesterol, LDL, and HDL levels on gallstone cholesterol concentration. Pak J Med Sci 2013;29:187-190.
- Fu X, Gong K, Shao X. The relationship between serum lipids, apolipoproteins level and bile lipids level, chemical type of stone. Zhonghua Yi Xue Za Zhi 1995;75:656-659, 708.
- Lavoie B, Nausch B, Zane EA, et al. Disruption of gallbladder smooth muscle function is an early feature in the development of cholesterol gallstone disease. Neurogastroenterol Motil 2012;24:e313-e324.
- Wang HH, Portincasa P, Wang DQ. Update on the molecular mechanisms underlying the effect of cholecystokinin and cholecystokinin-1 receptor on the formation of cholesterol gallstones. Curr Med Chem 2019;26:3407-3423.
- Kanto J, Katevuo K. The effect of drugs with different mechanisms of action on the contraction of the human gallbladder. Int J Clin Pharmacol Ther Toxicol 1981;19:303-309.
- Dai X, Shen L. Advances and trends in omics technology development. Front Med (Lausanne) 2022;9:911861.
- Yamada R, Okada D, Wang J, Basak T, Koyama S. Interpretation of omics data analyses. J Hum Genet 2021;66:93-102.
- Buch S, Schafmayer C, Völzke H, et al. A genome-wide association scan identifies the hepatic cholesterol transporter ABCG8 as a susceptibility factor for human gallstone disease. Nat Genet 2007;39:995-999.
- Yu XH, Qian K, Jiang N, Zheng XL, Cayabyab FS, Tang CK. ABCG5/ABCG8 in cholesterol excretion and atherosclerosis. Clin Chim Acta 2014;428:82-88.
- Lauridsen BK, Stender S, Frikke-Schmidt R, Nordestgaard BG, Tybjærg-Hansen A. Genetic variation in the cholesterol transporter NPC1L1, ischaemic vascular disease, and gallstone disease. Eur Heart J 2015;36:1601-1608.
- Betters JL, Yu L. NPC1L1 and cholesterol transport. FEBS Lett 2010;584:2740-2747.
- Cao K, Zhang K, Ma M, Ma J, Tian J, Jin Y. Lactobacillus mediates the expression of NPC1L1, CYP7A1, and ABCG5 genes to regulate cholesterol. Food Sci Nutr 2021;9:6882-6891.
- Li R, Liu Y, Shi J, et al. Diosgenin regulates cholesterol metabolism in hypercholesterolemic rats by inhibiting NPC1L1 and enhancing ABCG5 and ABCG8. Biochim Biophys Acta Mol Cell Biol Lipids 2019;1864:1124-1133.
- Mohammadi A, Bazrafshani MR, Oshaghi EA. Effect of garlic extract on some serum biochemical parameters and expression of npc1l1, abca1, abcg5 and abcg8 genes in the intestine of hypercholesterolemic mice. Indian J Biochem Biophys 2013;50:500-504.
- Gu J, Zhu N, Li HF, et al. Ezetimibe and cancer: Is there a connection?. Front Pharmacol 2022;13:831657.
- Yang B, Liu B, Bi P, Wu T, Wang Q, Zhang J. An integrated analysis of differential miRNA and mRNA expressions in human gallstones. Mol Biosyst 2015;11:1004-1011.
- Wang G, Zhang H, Zhou Z, et al. AQP3-mediated activation of the AMPK/SIRT1 signaling pathway curtails gallstone formation in mice by inhibiting inflammatory injury of gallbladder mucosal epithelial cells. Mol Med 2023;29:116.
- Guman MSS, Hoozemans JB, Haal S, et al. Adipose tissue, bile acids, and gut microbiome species associated with gallstones after bariatric surgery. J Lipid Res 2022;63:100280.
- Casertano M, Trotta MC, Cenni S, et al. Infliximab therapy decreases the expression of serum and faecal miR-126 and miR-20a in paediatric Crohn's disease: A pilot study. Acta Paediatr 2024;113:590-597.
- Wei J, Chen T, Liu Y, et al. Targeted bile acids metabolomics in cholesterol gallbladder polyps and gallstones: From analytical method development towards application to clinical samples. J Pharm Anal 2023;13:1080-1087.
- Slyshenkov VS, Rakowska M, Wojtczak L. Protective effect of pantothenic acid and related compounds against permeabilization of Ehrlich ascites tumour cells by digitonin. Acta Biochim Pol 1996;43:407-410.
- Akram M. Citric acid cycle and role of its intermediates in metabolism. Cell Biochem Biophys 2014;68:475-478.
Article
Review Article
Korean J Gastroenterol 2025; 85(1): 11-21
Published online January 25, 2025 https://doi.org/10.4166/kjg.2024.135
Copyright © The Korean Society of Gastroenterology.
Relationship between Abnormal Lipid Metabolism and Gallstone Formation
Xiang Li1,2,3*, Xiaodan Yin1,2,3*, Jun Xu1,2,3, Lei Geng1,2,3 , Zhengtao Liu4
Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou1; NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of the diagnosis and treatment of organ Transplantation, CAMS, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou2; Key Laboratory of Organ Transplantation, Zhejiang Province, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou3; Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang4, China
Correspondence to:Lei Geng, Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, No. 366, wutong Road, Xihu District, Hangzhou City, Zhejiang Province, 310003, China. Tel: +86 13777802607, Fax: +86 571 87236114, E-mail:geng97927@163.com, ORCID: https://orcid.org/0009-0006-8693-5721
Zhengtao Liu, Shulan International Medical College, Zhejiang Shuren University, No. 8 Shuren Street, Gongshu District, Hangzhou, Zhejiang Province, 310000, China. Tel: +86 15289668420, Fax: +86 571 87236570, Email: liuzhengtao@zjsru.edu.cn, ORCID:: https://orcid.org/0000-0002-6232-4778
*These authors contributed equally to this work.
This is an open access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
Abstract
Cholelithiasis is a common biliary system disease with a high incidence worldwide. Abnormal lipid metabolism has been shown to play a key role in the mechanism of gallstones. Therefore, recent research literature on the genes, proteins, and molecular substances involved in lipid metabolism during the pathogenesis of gallstones has been conducted. This study aimed to determine the role of lipid metabolism in the pathogenesis of gallstones and provide insights for future studies using previous research in genomics, metabolomics, transcriptomics, and other fields.
Keywords: Gallstones, Lipid metabolism, Triglycerides, Phospholipids, Metabolomics
INTRODUCTION
Gallstone disease (GSD) is a common biliary system disease composed primarily of cholesterol stones, pigment stones (composed mainly of calcium salts and bile pigments), and mixed stones.1 Cholecystolithiasis tends to occur in adults with an overall incidence of 10–15%. The incidence increases with age after 40 years, with a higher incidence in women than men.2
Three main types of gallstones exist: cholesterol, pigmented, and mixed.3 Most gallstones are cholesterol gallstones, accounting for approximately 90%.4 The symptoms of gallstones are usually caused by cholesterol stones.5
Many studies have revealed a close relationship between gallstone formation and human metabolic activity.6 The high expression of beta3-adrenergic receptors and HMG-CoA reductase in obese patients7 can affect gallbladder contraction and inhibit cholesterol excretion in the gallbladder.8,9 In metabolic syndrome, the low high-density lipoprotein,10 elevated triglyceride, and high homocysteine levels lead to a pro-inflammatory state that increases the rate of gallstone formation.11,12 The abnormal metabolism of bile acids in the human body can also affect cholesterol excretion in the bile.13
Gallstone formation is closely related to human metabolism, particularly lipid metabolism. Therefore, studying lipid metabolism will help better understand the mechanism of gallbladder stone formation.
LIPID METABOLISM
Lipids consist of triglycerides (TGs), phospholipids, and cholesterol. They are important nutrients required by the human body, providing energy and essential fatty acids and participating in cell composition. Fatty acids are the most common form of energy storage in the human body. They are stored mainly in the form of TGs.14 Phospholipids and cholesterol are important components of the cell membranes.15
The liver is the main site of lipid synthesis and metabolism.10,14 The synthesis and metabolism of lipids in the normal human body are balanced. Disruption of this balance leads to various diseases such as obesity, atherosclerosis, and type 2 diabetes.16,17
1. Cholesterol metabolism
Cholesterol plays an important role in the cell membrane composition, intracellular signaling, and the synthesis of various hormones.18 Acetyl-CoA is a raw material for cholesterol biosynthesis. It is synthesized in cells through the synergistic action of more than 20 enzymes, e.g., HMG-CoA reductase, exerting its biological functions.19,20 Cholesterol biosynthesis is related to the levels of foam sterol, cholestanol, and desfilin.21,22 The liver is the primary site of cholesterol biosynthesis, transmitting endogenous and exogenous cholesterol as extremely low-density lipoproteins (VLDL) into the bloodstream. Excess cholesterol combines with apolipoprotein A-I (apoA-I) to form high-density lipoprotein (HDL).23,24 Nuclear receptor liver X receptors (LXRs) are important regulatory factors for intracellular cholesterol and lipid homeostasis.25-27
Cholesterol can produce various oxidative sterols via enzymatic and non-enzymatic pathways.28 Some are further metabolized into bile acids in the liver and discharged into the gallbladder, becoming a component of bile.19 The stability of the cholesterol metabolism has a regulatory effect on the inflammatory response and tumor microenvironment.29,30 Moreover, some cholesterol molecules, such as SOAT1, SQLE, and NPC1, have become immunotherapeutic targets for tumors (Fig. 1).31-33
-
Figure 1. In vivo synthesis and metabolic pathways of cholesterol.
2. Triglyceride metabolism
TGs are the main source of energy in the human body and are synthesized mainly in the liver via the diglyceride pathway using sugars, glycerol, and fatty acids.14,34 The liver takes up excess glucose and fructose35-37 through de novo lipogenesis and converts them into TGs.38 The activation of transcription factor 7-like 2 (TCF7L2)39 and liver cell membrane CD36 increases the intake of fatty acids, promoting de novo lipogenesis.40 The action of insulin41,42 and the secretion of the hepatokine oromucoid II limits the progression of de novo lipogenesis.43 Small intestinal mucosal cells can synthesize TGs using glycerol monoesters and fatty acids absorbed through exogenous digestion.
The lipoprotein lipase protein complex (LPL) determines the breakdown of TGs, and liver receptors (RLDL and LRP-1) are responsible for transporting and clearing the decomposed products.44 Dietary habits45,46 and the size of the TG decomposition products and intermediates,47 such as lipid droplets,48 can influence the activity of fatty acid TG lipase, affecting TG metabolism.
3. Phospholipid metabolism
Phospholipids are synthesized in the endoplasmic reticulum, where TGs and phosphatidic acids bind to sphingosine to form a phospholipid sphingosine complex. The complex then combines with phosphate and choline to form phospholipid molecules such as lecithin and cephalin.49 Some fatty acids, inositol, and enzymes can modify phospholipid molecules to perform various biological functions.50 In cells, phospholipids are transported and transmitted through vesicles and cell membranes.51 The roles of phosphoinositol and phospholipase D in vesicular transport reactions are important.52-54
Phospholipids can be hydrolyzed into glycerol, fatty acids, phosphate, and various amino alcohols through phospholipase reactions in living organisms. Phospholipases A1 and A2 cleave fatty acids at the 1,2 positions of phospholipid molecules. In addition, the phospholipase D (PLD) enzyme catalyzes the hydrolysis of phosphodiesterase bonds in glycerol phospholipids.54,55 Sphingosine 1-phosphate (S1P) is a metabolic product of cell membrane sphingolipids, which bind to the S1P receptor (S1PR) coupled with the G protein to regulate embryonic development, organ function, inflammatory response, and multiple system diseases.56-59 The ABCB4 gene, cleavage of lipoproteins, and structural activity of the endoplasmic reticulum have regulatory effects on the biosynthesis and metabolism of phospholipids (Fig. 2).60-63
-
Figure 2. Key enzymes for phospholipid synthesis and metabolism in vivo.
ABNORMAL LIPID METABOLISM AND GALLSTONES
Bile is a three-variable solution; the bile acid, lecithin, and cholesterol concentration ratio in bile affects cholesterol solubility.64 Under normal circumstances, bile salts and lecithin can disperse cholesterol to form soluble microcapsules, which are less likely to precipitate and form stones. Bile cholesterol is more likely to become supersaturated when cholesterol secretion in bile increases or the concentration of bile acids and lecithin decreases.13
1. Cholesterol metabolism and bile biochemistry
Bile acids mainly pass through cholesterol 7 α-hydroxylase (CYP7A1), which oxidizes cholesterol. The regulatory effect of the nuclear factor farnesoid X receptor (FXR) is a key factor.13 FXR downregulates CYP7A1 by inducing a cascade reaction involving human fibroblast growth factor 19-JNK.65,66 Fibrin can increase the risk of gallstones 1.7-fold.67 CYP7A1 polymorphisms also lead to a significantly higher risk of developing gallstones.66 Cholesterol precipitates and forms gallstones when cholesterol synthesis is excessively active and cholesterol in the bile is supersaturated.9 CYP7A1 is inhibited when FXR is overactive, which affects cholesterol breakdown and metabolism into bile acids, further promoting cholesterol accumulation and precipitation.66,68
Some receptors on the gallbladder wall regulate cholesterol absorption, such as scavenger receptor SR-BI.69 This receptor is related to the occurrence of atherosclerosis, and the supersaturation of bile cholesterol appears to be related to SR-BI receptor inhibition.70-72
This study suggests that abnormalities in CYP7A1, FXR, and SR-BI can lead to metabolic disorders of cholesterol in bile, accelerating the formation of gallstones. On the other hand, Chen et al. found through Mendelian randomization analysis that the association between the serum total cholesterol (TC) levels and the risk of gallstones was not linear; lower TC levels lead to a higher risk of stone formation.73
2.Triglyceride metabolism and bile biochemistry
Currently, a large number of clinical studies have shown that patients with hyperlipidemia have a higher risk of developing gallstones. Previous studies have shown that knocking out CD36 on the liver cell membrane, which plays a regulatory role in the de novo lipogenesis pathway, significantly reduces the incidence of gallstones in mice.74 Elevated levels of very low-density lipoprotein (VLDL) related to TG transport can also accelerate the excretion of bile acids.75 Evidence suggests that the gallbladder of high TG patients is less sensitive to CCK. This sensitivity is improved after reversing the high serum TG levels with TG-lowering agents.76 These results confirmed the correlation between the increase in TG and gallstones.
3. Phospholipid metabolism and bile biochemistry
Phospholipids are important biochemical components of bile. The solubility of cholesterol in bile salt phospholipid mixtures is much higher than in pure bile salts.77 Therefore, phospholipids play an important role in the biochemical balance of bile. Phospholipids are generated mainly in the endoplasmic reticulum, with sphingomyelin synthetase being the key enzyme for their synthesis. Abnormal enzyme levels can cause non-alcoholic fatty liver, atherosclerosis, and diabetes.78,79 On the other hand, no reports on the relationship between this enzyme abnormality and the occurrence of gallstones have been published.
The most extensively studied phospholipid transport process is the endoplasmic reticulum-mitochondria encounter structure (ERMES) complex between the endoplasmic reticulum and mitochondria, which is a critical pathway for phospholipid transfer between two organelles.80 It is unclear if there is a connection between this pathway and gallstone formation. Low phospholipid-associated cholelithiasis (LPAC) is an inherited cholelithiasis associated with low phospholipid levels in the bile, accounting for approximately 0.5–2% of symptomatic cholelithiasis.81 LPAC is a genetic disease caused by a single gene mutation associated with mutations in ABCB4, which mainly encodes the carrier protein MDR3 in the biliary tract.61,82 MDR3 exists on the membrane of liver tubules; its main function is to transport phosphatidylcholine from liver cells to the gallbladder. When the MDR3 protein mutates, phospholipid transport disorders lead to easier cholesterol precipitation in bile (Fig. 3).83 In summary, abnormal phospholipid generation and transport caused by various factors can lead to bile cholesterol supersaturation, accelerating gallstone formation.
-
Figure 3. Effects of MDR3 mutations on phospholipid transport on the membrane of hepatic tubular cells.
4. Lipoproteins and bile biochemistry
Many studies have examined the relationship between the HDL and LDL levels in the serum and bile metabolism, as well as the formation of gallstones. A study on the correlation between serum lipoprotein levels and gallstone formation in Western patients with gallstones found that high LDL levels can increase the cholesterol concentration in bile (p<0.01) and increase the incidence of gallstones (p<0.05).84 HDL-C is positively correlated with phospholipids and bile acids in the bile. Hence, high LDL levels can accelerate the formation of gallstones, whereas HDL-C may offer some protection for gallstone formation.85
5. Lipid metabolism and gallbladder contraction disorders
Under normal conditions, the gallbladder can drive the intestinal hepatic circulation of bile acids through regular contraction and relaxation.86 This process is regulated by cholecystokinin (CCK) secreted by small intestinal cells.87 Dietary fat can stimulate CCK secretion to some extent. Active cholesterol synthesis leads to a sustained supersaturation state, resulting in the deposition of cholesterol on the gallbladder wall, COX2-mediated inflammation, and the abnormal contraction and relaxation of the gallbladder.10,88 Gallbladder contraction disorders can affect the excretion of cholesterol and bile acids, increasing the risk of gallstones.
OMICS TECHNIQUES AND GALLSTONES
Omics technology refers to the detection and analysis of data representing the structure and function of a specific biological system, such as genes and proteins, and the acquisition of systematic connections between the data at that level. The technology mainly includes genomics, proteomics, metabolomics, transcriptomics, lipidomics, immunohistochemistry, glycomics, RNA omics, imaging omics, and ultrasound omics.89 The integration of multiple omics has become a common trend in constructing comprehensive causal relationships, with genomics, transcriptomics, proteomics, and metabolomics being the most widely used approaches.
Genomic research measures DNA molecules, whereas epigenomics, transcriptomics, proteomics, and metabolomics research measures the chemical states of DNA and its binding proteins, RNA, proteins, and metabolites.90 Compared to traditional singularity research, the basic status of genes, RNA, proteins, and metabolites in the target sample can be understood by integrating and analyzing genomic, transcriptomic, proteomic, and metabolomic data, providing a comprehensive interpretation of the biological system.
1. Genomic research on gallstones
Research on the gallbladder genome has found that multiple sets of alleles in the gallbladder tissue of patients with stones have undergone mutations, which have been isolated and localized. These include mutations in the liver cholesterol transporter (ABC transporter) ABCG5/ABCG8,91,92 CYP7A1, and changes in the multicellular transmembrane protein NPC1L1 (Niemann Pick C1 Like 1) are associated with the intestinal and liver reabsorption of cholesterol.93
ABCG5/ABCG8 are present in intestinal and liver cells to inhibit the intestinal absorption of cholesterol and promote bile secretion.92 NAPC1L1 is present in intestinal epithelial and liver cells, promoting cholesterol absorption through endocytosis mediated by grid proteins.94 Changes in these genes and proteins promote cholesterol absorption in the human body, inhibit cholesterol transport and bile excretion, and accelerate the nucleation of cholesterol in bile.
The treatment of gallstones is mainly surgical, but early prevention and intervention in gallstones can be achieved by exploring the omics mechanisms of gallstone formation. Currently, in vitro experiments have been conducted to regulate the cholesterol levels in rats through the mediation of NAPC1L1 and ABCG5/ABCG8 proteins by lactic acid bacteria, garlic extract, or Diosgenin.95-97 In addition, NPC1L1 is the target of the lipid-lowering drug ezetimibe in the intestine,98 which may contribute to the early prevention of gallstones.
2. Transcriptomics and Gallstones
Transcriptomic research on gallbladder diseases is progressing rapidly, including the study of genes related to the onset of single diseases such as gallstones, gallbladder polyps, and gallbladder cancer.
In a study of gallstones, a correlation was found between miR-20 (RNA-regulating ABC transporter) and the occurrence of gallstones through the transcriptome analysis of gallbladder tissue.99 This confirms the results of the genomic research on ABC transporter proteins. Aquaporin 3 (AQP3) can reduce gallbladder inflammation and inhibit gallstone formation.100 miR-20 and AQP3 also play important roles in the lipid metabolism. Changes in miR-20 and AQP3 can affect the transportation of cholesterol and TG, promoting the formation of gallstones. A research team from the Netherlands conducted transcriptomic studies on adipose tissue of patients with gallbladder cancer. They revealed a significant increase in apolipoprotein A1 (APOA1) involvement in patients with stones. This can lead to the release of beneficial cholesterol from adipose tissue, resulting in the supersaturation of bile cholesterol.101 Infliximab (IFX) reduces miR-20 expression in children, and research in this area may provide a reference for treating gallstones.102
3. Metabolomics and Gallstones
Metabolomic research on gallstones has focused mainly on differential metabolites. By comparing the plasma or bile metabolism of patients with gallstones and normal individuals, some studies have found that cholesterol and various bile acids are significantly elevated in patients with gallstones.101,103 Relatively little research has been conducted on other metabolites and metabolic pathways related to stone formation.
In an unpublished study, plasma samples were collected from more than 100 patients, and non-targeted metabolomic testing was performed. The detection data were compared with the clinical indicators. The metabolites and metabolic pathways related to GSD generation were obtained using a weighted correlation network analysis (WGCNA) algorithm combined with KEGG pathway enrichment analysis.
The present study found a significant decrease in linoleic acid in the peripheral blood of patients with GSD. Moreover, its corresponding linoleic acid metabolic pathway also showed a significant downregulation trend. The pantothenate and CoA biosynthesis and TCA cycle pathways were significantly upregulated. Linoleic acid is a raw material used to synthesize and convert phospholipids. When the peripheral blood linoleic acid level decreases, there is an obstacle in the production of phospholipids in bile, and the precipitation of cholesterol becomes more pronounced. The pantothenate and CoA bio-synthesis pathways were upregulated in the peripheral blood of patients with GSD. Upregulation of this pathway will increase CoA production, the raw material for cholesterol synthesis, promoting cholesterol production.23,104 The TCA cycle is the oxidation pathway of nutrients in the body. The strong expression of this pathway can also promote the conversion of carbohydrates to lipids during satiety, increase the burden of lipids in the body, affect the lipid structure of the body, and promote the precipitation of cholesterol in bile (Fig. 4).105 Based on this study, GSD can be prevented and treated by affecting the linoleic acid cycle, and the effectiveness of drugs, such as ethyl linoleate, may serve as a direction for future research.
-
Figure 4. Pathway of TCA cycle in the formation of gallstones.
CONCLUSION
The abnormal metabolism of cholesterol, TGs, and phospholipids plays a significant role in gallstone formation. When FXR is overactivated, or CYP7A1 and SR-BI receptors are inhibited to a certain extent, cholesterol in bile becomes supersaturated, and a COX2-mediated inflammatory response is induced in the gallbladder wall, accelerating gallstone formation. The transport of TGs by VLDL and CD36 molecules in the liver cell membrane is affected, increasing the risk of gallstones. In the phospholipid metabolism, abnormalities in the MDR3 protein that regulates phospholipid transfer cause low phospholipid-related cholelithiasis.
In the genomic and transcriptome analysis of gallbladder tissue, changes in the liver cholesterol transporters ABCG5/ ABCG8 accelerate cholesterol nucleation, while abnormalities in aquaporin 3 (AQP3) and apolipoprotein A1 (APOA1) affect the release and transport of cholesterol and TGs (Fig. 5). Through metabolomic techniques, this study discovered that the linoleic acid metabolism pathway, pantothenic acid and coenzyme A biosynthesis pathways, and the TCA cycle pathway play a role in the generation and balance of phospholipids and cholesterol. These omics-related studies provide a new theoretical basis for preventing and treating gallstones and directions for future research.
-
Figure 5. Positions and roles of CD36, APOA-1, SR-BI, and CYP7A1 in the KEGG pathway of cholesterol synthesis.
Financial support
None.
Conflict of interest
None.
Fig 1.

Fig 2.

Fig 3.

Fig 4.

Fig 5.

References
- Lammert F, Gurusamy K, Ko CW, et al. Gallstones. Nat Rev Dis Primers 2016;2:16024.
- Chen CH, Huang MH, Yang JC, et al. Prevalence and risk factors of gallstone disease in an adult population of Taiwan: an epidemiological survey. J Gastroenterol Hepatol 2006;21:1737-1743.
- Qiao T, Ma RH, Luo XB, Yang LQ, Luo ZL, Zheng PM. The systematic classification of gallbladder stones. PLoS One 2013;8:e74887.
- Tazuma S. Gallstone disease: Epidemiology, pathogenesis, and classification of biliary stones (common bile duct and intra-hepatic). Best Pract Res Clin Gastroenterol 2006;20:1075-1083.
- Schafmayer C, Hartleb J, Tepel J, et al. Predictors of gallstone composition in 1025 symptomatic gallstones from Northern Germany. BMC Gastroenterol 2006;6:36.
- Portincasa P, Moschetta A, Palasciano G. Cholesterol gallstone disease. Lancet 2006;368:230-239.
- Chen L, Yang H, Li H, He C, Yang L, Lv G. Insights into modifiable risk factors of cholelithiasis: A Mendelian randomization study. Hepatology 2022;75:785-796.
- Klass DM, Lauer N, Hay B, Kratzer W, Fuchs M; EMIL Study Group. Arg64 variant of the beta3-adrenergic receptor is associated with gallstone formation. Am J Gastroenterol 2007;102:2482-2487.
- Reshetnyak VI. Concept of the pathogenesis and treatment of cholelithiasis. World J Hepatol 2012;4:18-34.
- Itani M, Dubinsky TJ. Physical chemistry of bile: Detailed pathogenesis of cholelithiasis. Ultrasound Q 2017;33:229-236.
- Santana-Gálvez J, Cisneros-Zevallos L, Jacobo-Velázquez DA. Chlorogenic acid: Recent advances on its dual role as a food additive and a nutraceutical against metabolic syndrome. Molecules 2017;22:358.
- Worthington HV, Hunt LP, McCloy RF, Ubbink JB, Braganza JM. Dietary antioxidant lack, impaired hepatic glutathione reserve, and cholesterol gallstones. Clin Chim Acta 2004;349:157-165.
- Fremont-Rahl JJ, Ge Z, Umana C, et al. An analysis of the role of the indigenous microbiota in cholesterol gallstone pathogenesis. PLoS One 2013;8:e70657.
- Alves-Bezerra M, Cohen DE. Triglyceride metabolism in the liver. Compr Physiol 2017;8:1-8.
- Tamura Y, Sesaki H, Endo T. Phospholipid transport via mitochondria. Traffic 2014;15:933-945.
- DeBose-Boyd RA. Significance and regulation of lipid metabolism. Semin Cell Dev Biol 2018;81:97.
- Jones JG. Hepatic glucose and lipid metabolism. Diabetologia 2016;59:1098-1103.
- Cortes VA, Busso D, Maiz A, Arteaga A, Nervi F, Rigotti A. Physiological and pathological implications of cholesterol. Front Biosci (Landmark Ed) 2014;19:416-428.
- Luo J, Yang H, Song BL. Mechanisms and regulation of cholesterol homeostasis. Nat Rev Mol Cell Biol 2020;21:225-245.
- Cholesterol absorption versus cholesterol synthesis in man. Nutr Rev 1970;28:11-15.
- Márk L, Paragh G. Change in the cholesterol metabolism associated with the combined inhibition of synthesis and absorption. Orv Hetil 2007;148:627-632.
- Schroepfer GJ Jr. Oxysterols: modulators of cholesterol metabolism and other processes. Physiol Rev 2000;80:361-554.
- Phillips MC. Molecular mechanisms of cellular cholesterol efflux. J Biol Chem 2014;289:24020-24029.
- Zhang Q, Ke Y, Hong H. HDL and Lipid Metabolism. Adv Exp Med Biol 2022;1377:49-61.
- Bilotta MT, Petillo S, Santoni A, Cippitelli M. Liver X receptors: Regulators of cholesterol metabolism, inflammation, autoimmunity, and cancer. Front Immunol 2020;11:584303.
- Nazih H, Bard JM. Cholesterol, oxysterols and LXRs in breast cancer pathophysiology. Int J Mol Sci 2020;21:1356.
- Uppal H, Zhai Y, Gangopadhyay A, et al. Activation of liver X receptor sensitizes mice to gallbladder cholesterol crystallization. Hepatology 2008;47:1331-1342.
- Luu W, Sharpe LJ, Capell-Hattam I, Gelissen IC, Brown AJ. Oxysterols: Old tale, new twists. Annu Rev Pharmacol Toxicol 2016;56:447-467.
- Cardoso D, Perucha E. Cholesterol metabolism: a new molecular switch to control inflammation. Clin Sci (Lond) 2021;135:1389-1408.
- Xu H, Zhou S, Tang Q, Xia H, Bi F. Cholesterol metabolism: New functions and therapeutic approaches in cancer. Biochim Biophys Acta Rev Cancer 2020;1874:188394.
- Oni TE, Biffi G, Baker LA, et al. SOAT1 promotes mevalonate pathway dependency in pancreatic cancer. J Exp Med 2020;217:e20192389.
- Sun H, Li L, Li W, et al. p53 transcriptionally regulates SQLE to repress cholesterol synthesis and tumor growth. EMBO Rep 2021;22:e52537.
- O'Neill KI, Kuo LW, Williams MM, et al. NPC1 confers metabolic flexibility in triple negative breast cancer. Cancers (Basel) 2022;14:3543.
- Weiss SB, Kennedy EP, Kiyasu JY. The enzymatic synthesis of triglycerides. J Biol Chem 1960;235:40-44.
- Sanders FW, Griffin JL. De novo lipogenesis in the liver in health and disease: more than just a shunting yard for glucose. Biol Rev Camb Philos Soc 2016;91:452-468.
- Softic S, Cohen DE, Kahn CR. Role of dietary fructose and hepatic de novo lipogenesis in fatty liver disease. Dig Dis Sci 2016;61:1282-1293.
- Geidl-Flueck B, Hochuli M, Németh Á, et al. Fructose- and sucrose- but not glucose-sweetened beverages promote hepatic de novo lipogenesis: A randomized controlled trial. J Hepatol 2021;75:46-54.
- Ameer F, Scandiuzzi L, Hasnain S, Kalbacher H, Zaidi N. De novo lipogenesis in health and disease. Metabolism 2014;63:895-902.
- Lee DS, An TH, Kim H, et al. Tcf7l2 in hepatocytes regulates de novo lipogenesis in diet-induced non-alcoholic fatty liver disease in mice. Diabetologia 2023;66:931-954.
- Zeng H, Qin H, Liao M, et al. CD36 promotes de novo lipogenesis in hepatocytes through INSIG2-dependent SREBP1 processing. Mol Metab 2022;57:101428.
- Kawano Y, Cohen DE. Mechanisms of hepatic triglyceride accumulation in non-alcoholic fatty liver disease. J Gastroenterol 2013;48:434-441.
- Smith U, Kahn BB. Adipose tissue regulates insulin sensitivity: role of adipogenesis, de novo lipogenesis and novel lipids. J Intern Med 2016;280:465-475.
- Zhou B, Luo Y, Ji N, Hu C, Lu Y. Orosomucoid 2 maintains hepatic lipid homeostasis through suppression of de novo lipogenesis. Nat Metab 2022;4:1185-1201.
- Ibarretxe D, Masana L. Triglyceride metabolism and classification of hypertriglyceridemias. Clin Investig Arterioscler 2021;33 Suppl 2:1-6.
- Kersten S. The impact of fasting on adipose tissue metabolism. Biochim Biophys Acta Mol Cell Biol Lipids 2023;1868:159262.
- Barrows BR, Parks EJ. Contributions of different fatty acid sources to very low-density lipoprotein-triacylglycerol in the fasted and fed states. J Clin Endocrinol Metab 2006;91:1446-1452.
- Cerk IK, Wechselberger L, Oberer M. Adipose triglyceride lipase regulation: An overview. Curr Protein Pept Sci 2018;19:221-233.
- Schott MB, Weller SG, Schulze RJ, et al. Lipid droplet size directs lipolysis and lipophagy catabolism in hepatocytes. J Cell Biol 2019;218:3320-3335.
- Fyrst H, Saba JD. An update on sphingosine-1-phosphate and other sphingolipid mediators. Nat Chem Biol 2010;6:489-497.
- Alb JG Jr, Kearns MA, Bankaitis VA. Phospholipid metabolism and membrane dynamics. Curr Opin Cell Biol 1996;8:534-541.
- Moreau P, Cassagne C. Phospholipid trafficking and membrane biogenesis. Biochim Biophys Acta 1994;1197:257-290.
- Dickson EJ. Phosphoinositide transport and metabolism at membrane contact sites. Biochim Biophys Acta Mol Cell Biol Lipids 2022;1867:159107.
- Fruman DA, Meyers RE, Cantley LC. Phosphoinositide kinases. Annu Rev Biochem 1998;67:481-507.
- McDermott MI, Wang Y, Wakelam MJO, Bankaitis VA. Mammalian phospholipase D: Function, and therapeutics. Prog Lipid Res 2020;78:101018.
- Dekker N. Outer-membrane phospholipase A: known structure, unknown biological function. Mol Microbiol 2000;35:711-717.
- Cartier A, Hla T. Sphingosine 1-phosphate: Lipid signaling in pathology and therapy. Science 2019;366:eaar5551.
- Verstockt B, Vetrano S, Salas A, Nayeri S, Duijvestein M, Vande Casteele N; Alimentiv Translational Research Consortium (ATRC). Sphingosine 1-phosphate modulation and immune cell trafficking in inflammatory bowel disease. Nat Rev Gastroenterol Hepatol 2022;19:351-366.
- Jozefczuk E, Guzik TJ, Siedlinski M. Significance of sphingosine-1-phosphate in cardiovascular physiology and pathology. Pharmacol Res 2020;156:104793.
- Obinata H, Hla T. Sphingosine 1-phosphate and inflammation. Int Immunol 2019;31:617-625.
- Erlinger S. Low phospholipid-associated cholestasis and cholelithiasis. Clin Res Hepatol Gastroenterol 2012;36 Suppl 1:S36-40.
- Wang HH, Portincasa P, Liu M, Wang DQ. Genetic analysis of ABCB4 mutations and variants related to the pathogenesis and pathophysiology of low phospholipid-associated cholelithiasis. Genes (Basel) 2022;13:1047.
- Sam PN, Avery E, Claypool SM. Proteolytic control of lipid metabolism. ACS Chem Biol 2019;14:2406-2423.
- Nishimura T, Stefan CJ. Specialized ER membrane domains for lipid metabolism and transport. Biochim Biophys Acta Mol Cell Biol Lipids 2020;1865:158492.
- Coleman R. Bile salts and biliary lipids. Biochem Soc Trans 1987;15 Suppl:68S-80S.
- Portincasa P, Ciaula AD, Bonfrate L, Wang DQ. Therapy of gallstone disease: What it was, what it is, what it will be. World J Gastrointest Pharmacol Ther 2012;3:7-20.
- Claudel T, Zollner G, Wagner M, Trauner M. Role of nuclear receptors for bile acid metabolism, bile secretion, cholestasis, and gallstone disease. Biochim Biophys Acta 2011;1812:867-878.
- Caroli-Bosc FX, Le Gall P, Pugliese P, et al. Role of fibrates and HMG-CoA reductase inhibitors in gallstone formation: epidemiological study in an unselected population. Dig Dis Sci 2001;46:540-544.
- Hu H, Shao W, Liu Q, et al. Gut microbiota promotes cholesterol gallstone formation by modulating bile acid composition and biliary cholesterol secretion. Nat Commun 2022;13:252.
- Yu H. HDL and scavenger receptor class B type I (SRBI). Adv Exp Med Biol 2022;1377:79-93.
- Hermann S, Kuhlmann MT, Starsichova A, et al. Imaging reveals the connection between spontaneous coronary plaque ruptures, atherothrombosis, and myocardial infarctions in HypoE/SRBI-/- Mice. J Nucl Med 2016;57:1420-1427.
- Miquel JF, Moreno M, Amigo L, et al. Expression and regulation of scavenger receptor class B type I (SR-BI) in gall bladder epithelium. Gut 2003;52:1017-1024.
- Rigotti A, Zanlungo S, Miquel JF, Wang DQ. HDL receptor SR-BI and cholesterol gallstones. Hepatology 2002;35:240-242.
- Chen L, Qiu W, Sun X, et al. Novel insights into causal effects of serum lipids and lipid-modifying targets on cholelithiasis. Gut 2024;73:521-532.
- Xie Y, Cifarelli V, Pietka T, et al. Cd36 knockout mice are protected against lithogenic diet-induced gallstones. J Lipid Res 2017;58:1692-1701.
- Castro J, Amigo L, Miquel JF, et al. Increased activity of hepatic microsomal triglyceride transfer protein and bile acid synthesis in gallstone disease. Hepatology 2007;45:1261-1266.
- Smelt AH. Triglycerides and gallstone formation. Clin Chim Acta 2010;411:1625-1631.
- Hofmann AF, Schteingart CD, vanSonnenberg E, Esch O, Zakko SF. Contact dissolution of cholesterol gallstones with organic solvents. Gastroenterol Clin North Am 1991;20:183-199.
- Koh EH, Yoon JE, Ko MS, et al. Sphingomyelin synthase 1 mediates hepatocyte pyroptosis to trigger non-alcoholic steatohepatitis. Gut 2021;70:1954-1964.
- Chen Y, Cao Y. The sphingomyelin synthase family: proteins, diseases, and inhibitors. Biol Chem 2017;398:1319-1325.
- Endo T, Tamura Y, Kawano S. Phospholipid transfer by ERMES components. Aging (Albany NY) 2018;10:528-529.
- Dong C, Condat B, Picon-Coste M, et al. Low-phospholipid-associated cholelithiasis syndrome: Prevalence, clinical features, and comorbidities. JHEP Rep 2020;3:100201.
- Goubault P, Brunel T, Rode A, Bancel B, Mohkam K, Mabrut JY. Low-Phospholipid Associated Cholelithiasis (LPAC) syndrome: A synthetic review. J Visc Surg 2019;156:319-328.
- Gonzales E, Davit-Spraul A, Baussan C, Buffet C, Maurice M, Jacquemin E. Liver diseases related to MDR3 (ABCB4) gene deficiency. Front Biosci (Landmark Ed) 2009;14:4242-4256.
- Atamanalp SS, Keles MS, Atamanalp RS, Acemoglu H, Laloglu E. The effects of serum cholesterol, LDL, and HDL levels on gallstone cholesterol concentration. Pak J Med Sci 2013;29:187-190.
- Fu X, Gong K, Shao X. The relationship between serum lipids, apolipoproteins level and bile lipids level, chemical type of stone. Zhonghua Yi Xue Za Zhi 1995;75:656-659, 708.
- Lavoie B, Nausch B, Zane EA, et al. Disruption of gallbladder smooth muscle function is an early feature in the development of cholesterol gallstone disease. Neurogastroenterol Motil 2012;24:e313-e324.
- Wang HH, Portincasa P, Wang DQ. Update on the molecular mechanisms underlying the effect of cholecystokinin and cholecystokinin-1 receptor on the formation of cholesterol gallstones. Curr Med Chem 2019;26:3407-3423.
- Kanto J, Katevuo K. The effect of drugs with different mechanisms of action on the contraction of the human gallbladder. Int J Clin Pharmacol Ther Toxicol 1981;19:303-309.
- Dai X, Shen L. Advances and trends in omics technology development. Front Med (Lausanne) 2022;9:911861.
- Yamada R, Okada D, Wang J, Basak T, Koyama S. Interpretation of omics data analyses. J Hum Genet 2021;66:93-102.
- Buch S, Schafmayer C, Völzke H, et al. A genome-wide association scan identifies the hepatic cholesterol transporter ABCG8 as a susceptibility factor for human gallstone disease. Nat Genet 2007;39:995-999.
- Yu XH, Qian K, Jiang N, Zheng XL, Cayabyab FS, Tang CK. ABCG5/ABCG8 in cholesterol excretion and atherosclerosis. Clin Chim Acta 2014;428:82-88.
- Lauridsen BK, Stender S, Frikke-Schmidt R, Nordestgaard BG, Tybjærg-Hansen A. Genetic variation in the cholesterol transporter NPC1L1, ischaemic vascular disease, and gallstone disease. Eur Heart J 2015;36:1601-1608.
- Betters JL, Yu L. NPC1L1 and cholesterol transport. FEBS Lett 2010;584:2740-2747.
- Cao K, Zhang K, Ma M, Ma J, Tian J, Jin Y. Lactobacillus mediates the expression of NPC1L1, CYP7A1, and ABCG5 genes to regulate cholesterol. Food Sci Nutr 2021;9:6882-6891.
- Li R, Liu Y, Shi J, et al. Diosgenin regulates cholesterol metabolism in hypercholesterolemic rats by inhibiting NPC1L1 and enhancing ABCG5 and ABCG8. Biochim Biophys Acta Mol Cell Biol Lipids 2019;1864:1124-1133.
- Mohammadi A, Bazrafshani MR, Oshaghi EA. Effect of garlic extract on some serum biochemical parameters and expression of npc1l1, abca1, abcg5 and abcg8 genes in the intestine of hypercholesterolemic mice. Indian J Biochem Biophys 2013;50:500-504.
- Gu J, Zhu N, Li HF, et al. Ezetimibe and cancer: Is there a connection?. Front Pharmacol 2022;13:831657.
- Yang B, Liu B, Bi P, Wu T, Wang Q, Zhang J. An integrated analysis of differential miRNA and mRNA expressions in human gallstones. Mol Biosyst 2015;11:1004-1011.
- Wang G, Zhang H, Zhou Z, et al. AQP3-mediated activation of the AMPK/SIRT1 signaling pathway curtails gallstone formation in mice by inhibiting inflammatory injury of gallbladder mucosal epithelial cells. Mol Med 2023;29:116.
- Guman MSS, Hoozemans JB, Haal S, et al. Adipose tissue, bile acids, and gut microbiome species associated with gallstones after bariatric surgery. J Lipid Res 2022;63:100280.
- Casertano M, Trotta MC, Cenni S, et al. Infliximab therapy decreases the expression of serum and faecal miR-126 and miR-20a in paediatric Crohn's disease: A pilot study. Acta Paediatr 2024;113:590-597.
- Wei J, Chen T, Liu Y, et al. Targeted bile acids metabolomics in cholesterol gallbladder polyps and gallstones: From analytical method development towards application to clinical samples. J Pharm Anal 2023;13:1080-1087.
- Slyshenkov VS, Rakowska M, Wojtczak L. Protective effect of pantothenic acid and related compounds against permeabilization of Ehrlich ascites tumour cells by digitonin. Acta Biochim Pol 1996;43:407-410.
- Akram M. Citric acid cycle and role of its intermediates in metabolism. Cell Biochem Biophys 2014;68:475-478.