Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 357
Filter
Add more filters

Publication year range
1.
Hum Genomics ; 18(1): 19, 2024 Feb 12.
Article in English | MEDLINE | ID: mdl-38347599

ABSTRACT

The causal relationships between plasma metabolites and cholelithiasis/cholecystitis risks remain elusive. Using two-sample Mendelian randomization, we found that genetic proxied plasma campesterol level showed negative correlation with the risk of both cholelithiasis and cholecystitis. Furthermore, the increased risk of cholelithiasis is correlating with the increased level of plasma campesterol. Lastly, genetic colocalization study showed that the leading SNP, rs4299376, which residing at the ABCG5/ABCG8 gene loci, was shared by plasma campesterol level and cholelithiasis, indicating that the aberrant transportation of plant sterol/cholesterol from the blood stream to the bile duct/gut lumen might be the key in preventing cholesterol gallstone formation.


Subject(s)
Cholecystitis , Cholesterol/analogs & derivatives , Gallstones , Phytosterols , Humans , Lipoproteins/genetics , Mendelian Randomization Analysis , ATP Binding Cassette Transporter, Subfamily G, Member 8/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 5/genetics , Cholecystitis/epidemiology , Cholecystitis/genetics , Gallstones/epidemiology , Gallstones/genetics , Gallstones/metabolism
2.
BMC Gastroenterol ; 24(1): 5, 2024 Jan 02.
Article in English | MEDLINE | ID: mdl-38166630

ABSTRACT

INTRODUCTION: Pancreaticobiliary reflux (PBR) can induce gallstone formation; however, its pathogenic mechanism remains unclear. In this study, we explored the mechanism of PBR by the non-targeted metabolomic analysis of bile in patients with PBR. OBJECTIVE: The aim of this study was to investigate the pathogenic mechanism in PBR by the non-targeted metabolomic analysis of bile collected during surgery. METHODS: Sixty patients who underwent gallstone surgery at our center from December 2020 to May 2021 were enrolled in the study. According to the level of bile amylase, 30 patients with increased bile amylase ( > 110 U/L) were classified into the PBR group, and the remaining 30 patients were classified into the control group (≤ 110 U/L). The metabolomic analysis of bile was performed. RESULTS: The orthogonal projections to latent structure-discriminant analysis of liquid chromatography mass spectrometry showed significant differences in bile components between the PBR and control groups, and 40 metabolites were screened by variable importance for the projection value (VIP > 1). The levels of phosphatidylcholine (PC) and PC (20:3(8Z,11Z,14Z)/14:0) decreased significantly, whereas the levels of lysoPC (16:1(9z)/0:0), lysoPC (15:0), lysoPC (16:0), palmitic acid, arachidonic acid, leucine, methionine, L-tyrosine, and phenylalanine increased. CONCLUSIONS: Significant differences in bile metabolites were observed between the PBR and control groups. Changes in amino acids and lipid metabolites may be related to stone formation and mucosal inflammation.


Subject(s)
Bile , Gallstones , Humans , Gallstones/surgery , Gallstones/metabolism , Metabolomics/methods , Liquid Chromatography-Mass Spectrometry , Amylases
3.
World J Surg ; 48(7): 1662-1673, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38777749

ABSTRACT

BACKGROUND: The aim of this study was to establish features of inflammation in histologically normal gallbladders with gallstones and compare the expression of inflammatory markers in acutely and chronically inflamed gallbladders. METHODS: Immunohistochemistry was performed on formalin-fixed paraffin-embedded gallbladders for tumor necrosis factor (TNF)-α, interleukin (IL)-6, IL-2R, and substance p in three groups: Group I (n = 60) chronic cholecystitis, Group II (n = 57) acute cholecystitis and Group III (n = 45) histologically normal gallbladders with gallstones. Expression was quantified using the H-scoring system. RESULTS: Median, interquartile range expression of mucosal IL-2R in Groups I (2.65, 0.87-7.97) and II (12.30, 6.15-25.55) was significantly increased compared with group III (0.40, 0.10-1.35, p < 0.05). Submucosal IL-2R expression in Groups I (2.0, 1.12-4.95) and II (10.0, 5.95-14.30) was also significantly increased compared with Group III (0.50, 0.15-1.05, p < 0.05). There was no difference in the lymphoid cell IL-6 expression between Groups I (5.95, 1.60-18.15), II (6.10, 1.1-36.15) and III (8.30, 2.60-26.35, p > 0.05). Epithelial IL-6 expression of Group III (8.3, 2.6-26.3) was significantly increased compared with group I (0.5, 0-10.2, p < 0.05) as was epithelial TNF-α expression in Group III (85.0, 70.50-92.0) compared with Groups I (72.50, 45.25.0-85.50, p < 0.05) and II (61.0, 30.0-92.0, p < 0.05). Lymphoid cell Substance P expression in Groups I (1.90, 1.32-2.65) and II (5.62, 2.50-20.8) was significantly increased compared with Group III (1.0,1.0-1.30, p < 0.05). Epithelial cell expression of Substance P in Group III (121.7, 94.6-167.8) was significantly increased compared with Groups I (75.7, 50.6-105.3, p < 0.05) and II (78.9, 43.5-118.5, p < 0.05). CONCLUSION: Histologically normal gallbladders with gallstones exhibited features of inflammation on immunohistochemistry.


Subject(s)
Gallstones , Immunohistochemistry , Humans , Gallstones/pathology , Gallstones/metabolism , Male , Female , Middle Aged , Adult , Interleukin-6/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/analysis , Cholecystitis/pathology , Cholecystitis/metabolism , Substance P/metabolism , Gallbladder/pathology , Gallbladder/metabolism , Receptors, Interleukin-2/metabolism , Aged , Chronic Disease , Biomarkers/metabolism , Biomarkers/analysis , Cholecystitis, Acute/pathology , Cholecystitis, Acute/metabolism , Cholecystitis, Acute/surgery
4.
Hepatology ; 75(5): 1081-1094, 2022 05.
Article in English | MEDLINE | ID: mdl-34651315

ABSTRACT

BACKGROUND AND AIMS: Genome-wide association studies (GWAS) have identified several risk loci for gallstone disease. As with most polygenic traits, it is likely that many genetic determinants are undiscovered. The aim of this study was to identify genetic variants that represent new targets for gallstone research and treatment. APPROACH AND RESULTS: We performed a GWAS of 28,627 gallstone cases and 348,373 controls in the UK Biobank, replicated findings in a Scottish cohort (1089 cases, 5228 controls), and conducted a GWA meta-analysis (43,639 cases, 506,798 controls) with the FinnGen cohort. We assessed pathway enrichment using gene-based then gene-set analysis and tissue expression of identified genes in Genotype-Tissue Expression project data. We constructed a polygenic risk score (PRS) and evaluated phenotypic traits associated with the score. Seventy-five risk loci were identified (p < 5 × 10-8 ), of which 46 were new. Pathway enrichment revealed associations with lipid homeostasis, glucuronidation, phospholipid metabolism, and gastrointestinal motility. Anoctamin 1 (ANO1) and transmembrane Protein 147 (TMEM147), both in novel, replicated loci, are expressed in the gallbladder and gastrointestinal tract. Both regulate gastrointestinal motility. The gallstone risk allele rs7599-A leads to suppression of hepatic TMEM147 expression, suggesting that the protein protects against gallstone formation. The highest decile of the PRS demonstrated a 6-fold increased odds of gallstones compared with the lowest decile. The PRS was strongly associated with increased body mass index, serum liver enzymes, and C-reactive protein concentrations, and decreased lipoprotein cholesterol concentrations. CONCLUSIONS: This GWAS demonstrates the polygenic nature of gallstone risk and identifies 46 novel susceptibility loci. We implicate genes influencing gastrointestinal motility in the pathogenesis of gallstones.


Subject(s)
Gallstones , Genome-Wide Association Study , Gallstones/genetics , Gallstones/metabolism , Gastrointestinal Motility , Genetic Predisposition to Disease/genetics , Humans , Polymorphism, Single Nucleotide , White People
5.
J Biomed Sci ; 30(1): 44, 2023 Jun 27.
Article in English | MEDLINE | ID: mdl-37370162

ABSTRACT

BACKGROUND: Cholesterol gallstone disease is a common disease. Reducing cholesterol burden is important to prevent/treat gallstone. In this study, we investigated the application of diosgenin (DG) to prevent the formation of gallstone in mice. METHODS: Adult male C57BL/6J mice were fed with the lithogenic diet (LD) only or LD supplemented with DG or ezetimibe for 8 weeks. Incidences of gallstone formation were documented. Intestine and liver tissues were collected to measure the lipid contents and expression of genes in cholesterol metabolism. Caco2 cells were treated with DG to monitor the regulation on cholesterol absorption and the transcriptional regulation of Npc1l1 gene. Changes of gut microbiota by DG was analyzed. Intraperitoneal injection of LPS on mice was performed to verify its effects on STAT3 activation and Npc1l1 expression in the small intestine. RESULTS: LD led to 100% formation of gallstones in mice. In comparison, dietary DG or ezetimibe supplementary completely prevents gallstones formation. DG inhibited intestinal cholesterol absorption in mice as well as in Caco2 cells by down-regulation of Npc1l1 expression. DG could directly inhibit phosphorylation of STAT3 and its transcriptional regulation of Npc1l1 expression. Furthermore, DG could modulate gut microbiota profiles and LPS mediated STAT3 activation and Npc1l1 expression. CONCLUSION: Our results demonstrated that dietary DG could inhibit intestinal cholesterol absorption through decreasing NPC1L1 expression to prevent cholesterol gallstone formation.


Subject(s)
Diosgenin , Gallstones , Humans , Mice , Male , Animals , Gallstones/prevention & control , Gallstones/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Diosgenin/pharmacology , Diosgenin/metabolism , Caco-2 Cells , Lipopolysaccharides , Mice, Inbred C57BL , Intestines , Cholesterol , Diet , Ezetimibe/pharmacology , Ezetimibe/metabolism , Liver/metabolism
6.
Liver Int ; 42(7): 1585-1592, 2022 07.
Article in English | MEDLINE | ID: mdl-35129276

ABSTRACT

INTRODUCTION: Gallstones are increasingly common in children. Genetic analyses of adult cohorts demonstrated that the sterol transporter ABCG8 p.D19H and Gilbert UGT1A1*28 variants enhance the odds of developing gallstones. The genetic background of common lithiasis in children remains unknown. METHODS: Overall, 214 children with gallstone disease (1 month-17 years, 107 boys) were inclueded. The control cohorts comprised 214 children (age 6-17 years, 115 boys) and 172 adults (age 40-92 years, 70 men) without gallstones. The ABCG8 p.D19H and UGT1A1*28 polymorphisms as well as ABCB4 (c.504C>T rs1202283, c.711A>T rs2109505) and NPC1L1 variants (p.V1296V rs217434, c.-18C>A rs41279633) were genotyped using TaqMan assays. Serum concentrations of plant sterols and cholesterol precursors were measured by gas chromatography/mass spectrometry. RESULTS: The ABCG8 risk allele was associated with an increased risk of stones (OR = 1.82, p = .03). Children carrying the p.19H allele presented with lower serum concentrations of surrogate markers of intestinal cholesterol absorption and decreased ratios of phytosterols to the cholesterol precursor desmosterol. Carriers of the common NPC1L1 rs217434 allele had an increased gallstone risk compared with stone-free adults (OR 1.90, p < .01). This variant also affected the ratio of phytosterols to cholesterol precursors (p = .03). Other tested variants were not associated with gallstone risk. CONCLUSIONS: The p.D19H ABCG8 and, to a lesser extent, NPC1L1 rs217434 variants increase the risk of early-onset gallstone formation. These results point to the presence of a common lithogenic pathway in children and adults.


Subject(s)
Gallstones , Phytosterols , ATP Binding Cassette Transporter, Subfamily G, Member 8/genetics , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Child , Cholesterol , Gallstones/genetics , Gallstones/metabolism , Genetic Predisposition to Disease , Humans , Male , Membrane Transport Proteins/genetics , Middle Aged , Phytosterols/adverse effects , Phytosterols/genetics , Sterols/metabolism
7.
Int J Mol Sci ; 23(20)2022 Oct 15.
Article in English | MEDLINE | ID: mdl-36293210

ABSTRACT

Women are more prone to develop either hypothyroidism or cholesterol gallstones than men. However, a male predominance in cholesterol gallstones under hypothyroidism was reported. Recently, a novel pathogenic link between thyroid hormone (TH) deficiency and cholesterol gallstones has been described in male mice. Here, we investigate if TH deficiency impacts cholesterol gallstone formation in females by the same mechanism. Three-month-old C57BL/6J mice were randomly divided into a control, a TH deficient, a lithogenic, and a lithogenic + TH deficient group and diet-treated for two, four, and six weeks. Gallstone prevalence, liver function tests, bile composition, hepatic gene expression, and gallbladder aquaporin expression and localization were investigated. Cholesterol gallstones were observed in lithogenic + TH deficient but not lithogenic only female mice. Diminished hydrophilicity of primary bile acids due to decreased gene expression of hepatic detoxification phase II enzymes was observed. A sex-specific expression and localization of hepatobiliary aquaporins involved in transcellular water and glycerol permeability was observed under TH deficient and lithogenic conditions. TH deficiency promotes cholesterol gallstone formation in female C57BL/6J mice by the same mechanism as observed in males. However, cholesterol gallstone prevalence was lower in female than male C57BL/6J mice. Interestingly, the sex-specific expression and localization of hepatobiliary aquaporins could protect female C57BL/6J mice to cholestasis and could reduce biliary water transport in male C57BL/6J mice possibly contributing to the sex-dependent cholesterol gallstone prevalence under TH deficiency.


Subject(s)
Aquaporins , Cholestasis , Gallstones , Hypothyroidism , Female , Male , Mice , Animals , Bile Acids and Salts/metabolism , Mice, Inbred C57BL , Gallstones/genetics , Gallstones/metabolism , Gallstones/pathology , Glycerol/metabolism , Cholesterol/metabolism , Liver/metabolism , Aquaporins/genetics , Aquaporins/metabolism , Cholestasis/metabolism , Cholic Acid/metabolism , Hypothyroidism/metabolism , Hydrophobic and Hydrophilic Interactions , Thyroid Hormones/metabolism , Water/metabolism
8.
J Biol Chem ; 295(20): 7003-7017, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32273342

ABSTRACT

Cholelithiasis is one of the most prevalent gastroenterological diseases and is characterized by the formation of gallstones in the gallbladder. Both clinical and preclinical data indicate that obesity, along with comorbidity insulin resistance, is a predisposing factor for cholelithiasis. Forkhead box O1 (FoxO1) is a key transcription factor that integrates insulin signaling with hepatic metabolism and becomes deregulated in the insulin-resistant liver, contributing to dyslipidemia in obesity. To gain mechanistic insights into how insulin resistance is linked to cholelithiasis, here we determined FoxO1's role in bile acid homeostasis and its contribution to cholelithiasis. We hypothesized that hepatic FoxO1 deregulation links insulin resistance to impaired bile acid metabolism and cholelithiasis. To address this hypothesis, we used the FoxO1LoxP/LoxP-Albumin-Cre system to generate liver-specific FoxO1-knockout mice. FoxO1-knockout mice and age- and sex-matched WT littermates were fed a lithogenic diet, and bile acid metabolism and gallstone formation were assessed in these animals. We showed that FoxO1 affected bile acid homeostasis by regulating hepatic expression of key enzymes in bile acid synthesis and in biliary cholesterol and phospholipid secretion. Furthermore, FoxO1 inhibited hepatic expression of the bile acid receptor farnesoid X receptor and thereby counteracted hepatic farnesoid X receptor signaling. Nonetheless, hepatic FoxO1 depletion neither affected the onset of gallstone disease nor impacted the disease progression, as FoxO1-knockout and control mice of both sexes had similar gallstone weights and incidence rates. These results argue against the notion that FoxO1 is a link between insulin resistance and cholelithiasis.


Subject(s)
Bile Acids and Salts/metabolism , Forkhead Box Protein O1/metabolism , Gallstones/metabolism , Insulin Resistance , Signal Transduction , Animals , Bile Acids and Salts/genetics , Cholesterol/genetics , Cholesterol/metabolism , Female , Forkhead Box Protein O1/genetics , Gallstones/genetics , Gene Deletion , Gene Expression Regulation , Liver , Male , Mice , Mice, Transgenic , Organ Specificity , Phospholipids/genetics , Phospholipids/metabolism , Receptors, Cytoplasmic and Nuclear/biosynthesis , Receptors, Cytoplasmic and Nuclear/genetics
9.
Lipids Health Dis ; 20(1): 97, 2021 Aug 31.
Article in English | MEDLINE | ID: mdl-34465364

ABSTRACT

BACKGROUND: Pancreaticobiliary reflux (PBR) causes chronic inflammation of the gallbladder mucosa and changes in the bile components, which are known to promote gallstone formation. This study aimed to investigate the bile biochemistry changes in gallstone patients with PBR and provide new clues for research on the involvement of PBR in gallstone formation. METHODS: Patients undergoing surgery for gallstones between December 2020 and May 2021 were eligible for inclusion. The bile biochemistry (including amylase, lipase, triglyceride, cholesterol, free fatty acids [FFAs], alanine aminotransferase [ALT], aspartate aminotransferase [AST], alkaline phosphatase [ALP], and γ-glutamyl transferase [γ-GT]) of the included gallstone patients was analysed to determine correlations with PBR. RESULTS: In this study, 144 gallstone patients who underwent surgery were enrolled. Overall, 15.97 % of the patients had an increased bile amylase level, which was associated with older age and significantly higher bile levels of ALP, lipase, triglyceride, and FFAs. Positive correlations were observed between amylase and lipase, triglyceride, FFAs levels in the gallbladder bile. However, the bile levels of triglyceride, FFAs, and lipase were positively correlated with each other only in the PBR group and showed no significant correlation in the control (N) group. In addition, elevated bile FFAs levels were found to be an independent risk factor for gallbladder wall thickening. CONCLUSIONS: In conclusion, PBR-induced increase in FFAs and triglyceride in the gallbladder bile is a cause of gallstone formation, and an increase in bile ALP suggests the presence of cholestasis in PBR.


Subject(s)
Bile Reflux/metabolism , Bile/chemistry , Fatty Acids, Nonesterified/analysis , Gallstones/metabolism , Triglycerides/analysis , Adult , Aged , Fatty Acids, Nonesterified/metabolism , Female , Gallbladder/metabolism , Gallstones/chemistry , Humans , Male , Middle Aged , Mortality , Prospective Studies , Triglycerides/metabolism
10.
Ann Hepatol ; 23: 100289, 2021.
Article in English | MEDLINE | ID: mdl-33217585

ABSTRACT

INTRODUCTION AND OBJECTIVES: The incidence of gallstone-related disease steadily increased in the last few years. Here, we aimed to investigate the effect of tauroursodeoxycholic acid1 (TUDCA) on preventing cholesterol gallstones formation in high-fat fed (HFD) mice. MATERIAL AND METHODS: Specific pathogen-free male C57Bl/6 mice were fed a lithogenic diet2 (LD group) alone or in combination with TUDCA (5g/kg diet) for 8 weeks. Upon sacrifice, serum, gallbladder, liver and small intestine were collected and the formation of gallstones or crystals in the gallbladder was analyzed. Additionally, the intestinal microbiota, and bile acid composition, serum lipids and hepatic lipids were studied. RESULTS: Cholesterol gallstones with cholesterol crystals formed in mice of the LD-fed group (15/15, 100%). However, only cholesterol crystals were found in three mice without the presence of any gallstone in the TUDCA-treated group. Both serum and hepatic total cholesterol levels in the TUDCA group were significantly decreased compared with the LD group. Concomitantly, mRNA expression of Abcg5 and Abcg8 was significantly lower in the liver of the TUDCA group whilst mRNA transcripts for Abcb11, Acat2, and Cyp27 were significantly increased compared with the LD group. Additionally, the gallbladder cholesterol saturation index (1.06±0.15) in the TUDCA group was significantly decreased compared with the LD group. Interestingly, the ratio of Firmicutes/Bacteroides in the TUDCA group was increased 3x fold. CONCLUSIONS: TUDCA can inhibit the absorption and synthesis of lipids in the small intestine by improving the intestinal microbiota in HFD-fed mice, thus reducing gallstone formation.


Subject(s)
Cholagogues and Choleretics/therapeutic use , Gallstones/prevention & control , Gastrointestinal Microbiome/drug effects , Taurochenodeoxycholic Acid/therapeutic use , Animals , Bile Acids and Salts/metabolism , Disease Models, Animal , Gallstones/metabolism , Gallstones/pathology , Lipid Metabolism/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL
11.
J Lipid Res ; 61(5): 767-777, 2020 05.
Article in English | MEDLINE | ID: mdl-32127396

ABSTRACT

Many clinical studies and epidemiological investigations have clearly demonstrated that women are twice as likely to develop cholesterol gallstones as men, and oral contraceptives and other estrogen therapies dramatically increase that risk. Further, animal studies have revealed that estrogen promotes cholesterol gallstone formation through the estrogen receptor (ER) α, but not ERß, pathway. More importantly, some genetic and pathophysiological studies have found that G protein-coupled estrogen receptor (GPER) 1 is a new gallstone gene, Lith18, on chromosome 5 in mice and produces additional lithogenic actions, working independently of ERα, to markedly increase cholelithogenesis in female mice. Based on computational modeling of GPER, a novel series of GPER-selective antagonists were designed, synthesized, and subsequently assessed for their therapeutic effects via calcium mobilization, cAMP, and ERα and ERß fluorescence polarization binding assays. From this series of compounds, one new compound, 2-cyclohexyl-4-isopropyl-N-(4-methoxybenzyl)aniline (CIMBA), exhibits superior antagonism and selectivity exclusively for GPER. Furthermore, CIMBA reduces the formation of 17ß-estradiol-induced gallstones in a dose-dependent manner in ovariectomized mice fed a lithogenic diet for 8 weeks. At 32 µg/day/kg CIMBA, no gallstones are found, even in ovariectomized ERα (-/-) mice treated with 6 µg/day 17ß-estradiol and fed the lithogenic diet for 8 weeks. In conclusion, CIMBA treatment protects against the formation of estrogen-induced cholesterol gallstones by inhibiting the GPER signaling pathway in female mice. CIMBA may thus be a new agent for effectively treating cholesterol gallstone disease in women.


Subject(s)
Cholesterol/metabolism , Estrogens/pharmacology , Gallstones/chemically induced , Gallstones/prevention & control , Receptors, Estrogen/antagonists & inhibitors , Receptors, G-Protein-Coupled/antagonists & inhibitors , Animals , Calcium/metabolism , Cyclic AMP/metabolism , Female , Gallstones/metabolism , HL-60 Cells , Humans , Mice , Receptors, Estrogen/metabolism , Signal Transduction/drug effects
12.
BMC Gastroenterol ; 20(1): 59, 2020 Mar 06.
Article in English | MEDLINE | ID: mdl-32143645

ABSTRACT

BACKGROUND: The gut microbiota participates in the metabolism of substances and energy, promotes the development and maturation of the immune system, forms the mucosal barrier, and protects the host from pathogen attacks. Although the pathogenesis of cholesterol gallstones is still not clear, studies have suggested that gut microbiota dysbiosis plays an important role in their formation. METHODS: Microbial DNA from faeces of normal control patients and those of patients with calculi was subjected to 16S rRNA gene sequencing to detect gene expression changes in intestinal microbes. ELISA kits were used to measure free bile acids, secondary bile acids and coprostanol according to the manufacturer's instructions. The relationship between flora and their metabolites was then analysed. RESULTS: In the gallstone group, the diversity of intestinal bacteria and the abundances of certain phylogroups were significantly decreased (p < 0.05), especially Firmicutes (p < 0.05), the largest phylum represented by the gut microbiota. This study found an increase in free bile acids (p < 0.001) and secondary bile acids (p < 0.01) in the enterohepatic circulation. Bile salt hydrolase activity was not related to the abundances of BSH-active bacteria. 7a-dehydroxylating gut bacteria were significantly increased (p < 0.01), whereas cholesterol-lowering bacteria were significantly reduced (p < 0.05). The Ruminococcus gnavus group could be used as a biomarker to distinguish the gallstone group from the control group. CONCLUSION: We conclude that intestinal flora imbalance affects bile acid and cholesterol metabolism and is associated with gallstone formation.


Subject(s)
Bile Acids and Salts/metabolism , Gallstones/metabolism , Gallstones/microbiology , Gastrointestinal Microbiome , Adult , Bacteria/classification , Bacteria/genetics , Cholesterol/metabolism , DNA, Bacterial/analysis , Dysbiosis/microbiology , Enterohepatic Circulation , Feces/microbiology , Female , Gastrointestinal Microbiome/genetics , Gene Expression , Humans , Male , Middle Aged , RNA, Ribosomal, 16S/genetics , Sequence Analysis, RNA
13.
J Lipid Res ; 60(3): 498-505, 2019 03.
Article in English | MEDLINE | ID: mdl-30610083

ABSTRACT

Gallstone (GS) formation requires that bile is supersaturated with cholesterol, which is estimated by a cholesterol saturation index (CSI) calculated from gallbladder (GB) total lipids and the mol% (mole percent) of bile acids (BAs), cholesterol, and phospholipids (PLs). Whereas CSI indicates GS risk, we hypothesized that additional comparisons of GB lipid mol% data are inappropriate to identify why CSI is increased in GS disease. We anticipated that GB lipid mmol/l (millimole per liter) levels should instead identify that, and therefore retrieved GB mmol/l data for BAs, cholesterol, and PLs from a study on 145 GS and 87 GS-free patients and compared them with the corresponding mol% data. BA and PL mmol/l levels were 33% and 31% lower in GS patients, while cholesterol was unaltered. CSI was higher in GS patients and correlated inversely with GB levels of BAs and PLs, but not with cholesterol. A literature search confirmed, in 13 studies from 11 countries, that GB BA levels and, to a certain extent, PLs are strongly reduced in GS patients, while cholesterol levels are not elevated. Our findings show that a shortage of BAs is a major reason why GB bile is supersaturated with cholesterol in GS patients. These results are sustainable because they are also valid from a global perspective.


Subject(s)
Bile Acids and Salts/metabolism , Cholesterol/metabolism , Gallbladder/metabolism , Gallstones/metabolism , Adult , Female , Humans , Male
14.
J Hepatol ; 69(6): 1308-1316, 2018 12.
Article in English | MEDLINE | ID: mdl-30213590

ABSTRACT

BACKGROUND & AIMS: Most cholesterol gallstones have a core consisting of inorganic and/or organic calcium salts, although the mechanisms of core formation are poorly understood. We examined whether the paracellular permeability of ions at hepatic tight junctions is involved in the core formation of cholesterol gallstones, with particular interest in the role of phosphate ion, a common food additive and preservative. METHODS: We focused on claudin-3 (Cldn3), a paracellular barrier-forming tight junction protein whose expression in mouse liver decreases with age. Since Cldn3-knockout mice exhibited gallstone diseases, we used them to assess the causal relationship between paracellular phosphate ion permeability and the core formation of cholesterol gallstones. RESULTS: In the liver of Cldn3-knockout mice, the paracellular phosphate ion permeability through hepatic tight junctions was significantly increased, resulting in calcium phosphate core formation. Cholesterol overdose caused cholesterol gallstone disease in these mice. CONCLUSION: We revealed that in the hepatobiliary system, Cldn3 functions as a paracellular barrier for phosphate ions, to help maintain biliary ion homeostasis. We provide in vivo evidence that elevated phosphate ion concentrations play a major role in the lifestyle- and age-related risks of developing cholesterol gallstone disease under cholesterol overdose. LAY SUMMARY: Herein, we reveal a new mechanism for cholesterol gallstone formation, in which increased paracellular phosphate ion permeability across hepatobiliary epithelia causes calcium phosphate core formation and cholesterol gallstones. Thus, altered phosphate ion metabolism under cholesterol overdose plays a major role in the lifestyle- and age-related risks of developing cholesterol gallstone disease.


Subject(s)
Bile Canaliculi/metabolism , Cell Membrane Permeability/physiology , Cholesterol/metabolism , Claudin-3/metabolism , Gallstones/metabolism , Aging/physiology , Animals , Aquaporins/metabolism , Calcium/metabolism , Calcium Phosphates/metabolism , Claudin-3/genetics , Claudins/genetics , Claudins/metabolism , Female , Gene Knockout Techniques , Liver/metabolism , Male , Mice , Mice, Knockout , Phosphorus/metabolism , Tight Junctions/metabolism
15.
Gastroenterology ; 152(6): 1521-1535.e8, 2017 05.
Article in English | MEDLINE | ID: mdl-28088462

ABSTRACT

BACKGROUND & AIMS: Hypoxia-inducible factor 1α subunit (HIF1A) is a transcription factor that controls the cellular response to hypoxia and is activated in hepatocytes of patients with nonalcoholic fatty liver disease (NAFLD). NAFLD increases the risk for cholesterol gallstone disease by unclear mechanisms. We studied the relationship between HIF1A and gallstone formation associated with liver steatosis. METHODS: We performed studies with mice with inducible disruption of Hif1a in hepatocytes via a Cre adenoviral vector (inducible hepatocyte-selective HIF1A knockout [iH-HIFKO] mice), and mice without disruption of Hif1a (control mice). Mice were fed a diet rich in cholesterol and cholate for 1 or 2 weeks; gallbladders were collected and the number of gallstones was determined. Livers and biliary tissues were analyzed by histology, quantitative reverse-transcription polymerase chain reaction, immunohistochemistry, and immunoblots. We measured concentrations of bile acid, cholesterol, and phospholipid in bile and rates of bile flow. Primary hepatocytes and cholangiocytes were isolated and analyzed. HIF1A was knocked down in Hepa1-6 cells with small interfering RNAs. Liver biopsy samples from patients with NAFLD, with or without gallstones, were analyzed by quantitative reverse-transcription polymerase chain reaction. RESULTS: Control mice fed a diet rich in cholesterol and cholate developed liver steatosis with hypoxia; levels of HIF1A protein were increased in hepatocytes around central veins and 90% of mice developed cholesterol gallstones. Only 20% of the iH-HIFKO mice developed cholesterol gallstones. In iH-HIFKO mice, the biliary lipid concentration was reduced by 36%, compared with control mice, and bile flow was increased by 35%. We observed increased water secretion from hepatocytes into bile canaliculi to mediate these effects, resulting in suppression of cholelithogenesis. Hepatic expression of aquaporin 8 (AQP8) protein was 1.5-fold higher in iH-HIFKO mice than in control mice. Under hypoxic conditions, cultured hepatocytes increased expression of Hif1a, Hmox1, and Vegfa messenger RNAs (mRNAs), and down-regulated expression of AQP8 mRNA and protein; AQP8 down-regulation was not observed in cells with knockdown of HIF1A. iH-HIFKO mice had reduced inflammation and mucin deposition in the gallbladder compared with control mice. Liver tissues from patients with NAFLD with gallstones had increased levels of HIF1A, HMOX1, and VEGFA mRNAs, compared with livers from patients with NAFLD without gallstones. CONCLUSIONS: In steatotic livers of mice, hypoxia up-regulates expression of HIF1A, which reduces expression of AQP8 and concentrates biliary lipids via suppression of water secretion from hepatocytes. This promotes cholesterol gallstone formation. Livers from patients with NAFLD and gallstones express higher levels of HIF1A than livers from patients with NAFLD without gallstones.


Subject(s)
Cholesterol/metabolism , Gallstones/genetics , Gallstones/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Animals , Aquaporins/genetics , Aquaporins/metabolism , Bile/metabolism , Bile Acids and Salts/metabolism , Cholates/administration & dosage , Cholesterol, Dietary/administration & dosage , Cholesterol, Dietary/metabolism , Down-Regulation/genetics , Female , Gallbladder/pathology , Gallstones/pathology , Heme Oxygenase-1/genetics , Hepatocytes/metabolism , Humans , Hypoxia/metabolism , Inflammation/etiology , Liver/metabolism , Male , Membrane Proteins/genetics , Mice , Mice, Knockout , Mucins/metabolism , Non-alcoholic Fatty Liver Disease/complications , RNA, Messenger/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/genetics , Water/metabolism
16.
Am J Pathol ; 187(4): 808-818, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28283178

ABSTRACT

Cholesterol gallstone disease (CGD) is one of the most common gastrointestinal diseases. Lithogenic hepatic bile secretion precedes the formation of cholesterol gallstones. Constitutive androstane receptor (CAR), a member of nuclear family, plays an important role in cholesterol and bile acid metabolism. To examine whether activation of CAR can prevent cholesterol gallstone formation, we treated C57BL6/J mice maintained on a lithogenic diet with CAR agonist 1,4-bis-[2-(3, 5-dichlorpyridyloxy)] benzene and performed bile duct cannulation to study the dynamics of biliary lipids. We report that activation of CAR decreases the biliary cholesterol concentration and prevents CGD formation. The lower biliary cholesterol level was largely attributed to suppressed Abcg5 and Abcg8 expression in CAR-activated mice. CAR activation also promoted cholesterol conversion into bile acids by increasing the expression of Cyp7a1, a rate-limiting enzyme in bile acid biosynthesis. Activation of CAR enhanced bile acid re-absorption via increasing the expression of bile acid transporters Asbt and Ostß in the ileum. The hepatic steatosis was also improved in the liver of CAR-activated mice. Furthermore, activation of CAR protected the mice against the liver X receptor α-sensitized CGD through suppressing the expression of Abcg5/8. Collectively, CAR plays an important role in maintaining the homeostasis of cholesterol, bile acids, and triglycerides levels, and it might be a promising therapeutic target for preventing or treating CGD.


Subject(s)
Cholesterol/adverse effects , Gallstones/metabolism , Gallstones/prevention & control , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Bile/metabolism , Bile Canaliculi/metabolism , Biological Transport/genetics , Cholesterol 7-alpha-Hydroxylase , Constitutive Androstane Receptor , Fatty Liver/genetics , Fatty Liver/pathology , Gallbladder/metabolism , Gallbladder/pathology , Gallstones/pathology , Gene Expression Regulation , Lipogenesis/genetics , Liver/metabolism , Liver/pathology , Liver X Receptors/metabolism , Male , Mice, Inbred C57BL , Phospholipids/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
17.
J Lipid Res ; 58(8): 1692-1701, 2017 08.
Article in English | MEDLINE | ID: mdl-28634191

ABSTRACT

The scavenger receptor and multiligand transporter CD36 functions to promote cellular free fatty acid uptake and regulates aspects of both hepatic and intestinal cholesterol metabolism. However, the role of CD36 in regulating canalicular and biliary cholesterol transport and secretion is unknown. Here, we show that germline Cd36 knockout (KO) mice are protected against lithogenic diet (LD)-induced gallstones compared with congenic (C57BL6/J) controls. Cd36 KO mice crossed into congenic L-Fabp KO mice (DKO mice) demonstrated protection against LD-induced gallstones, reversing the susceptibility phenotype observed in L-Fabp KO mice. DKO mice demonstrated reduced biliary cholesterol secretion and a shift into more hydrophophilic bile acid species, without changes in either BA pool size or fecal excretion. In addition, we found that the mean and maximum force of gallbladder contraction was increased in germline Cd36 KO mice, and gallbladder lipid content was reduced compared with wild-type controls. Finally, whereas germline Cd36 KO mice were protected against LD-induced gallstones, neither liver- nor intestine-specific Cd36 KO mice were protected. Taken together, our findings show that CD36 plays an important role in modifying gallstone susceptibility in mice, at least in part by altering biliary lipid composition, but also by promoting gallbladder contractility.


Subject(s)
CD36 Antigens/deficiency , CD36 Antigens/genetics , Diet/adverse effects , Gallstones/genetics , Animals , Bile Acids and Salts/chemistry , Bile Acids and Salts/metabolism , Cholesterol/metabolism , Gallbladder/metabolism , Gallbladder/physiopathology , Gallstones/etiology , Gallstones/metabolism , Gallstones/physiopathology , Gene Knockout Techniques , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle Contraction/genetics
18.
Hepatology ; 64(3): 853-64, 2016 09.
Article in English | MEDLINE | ID: mdl-27014967

ABSTRACT

UNLABELLED: The adenosine triphosphate-binding cassette (ABC) sterol transporter, Abcg5/g8, is Lith9 in mice, and two gallstone-associated variants in ABCG5/G8 have been identified in humans. Although ABCG5/G8 plays a critical role in determining hepatic sterol secretion, cholesterol is still secreted to bile in sitosterolemic patients with a defect in either ABCG5 or ABCG8 and in either Abcg5/g8 double- or single-knockout mice. We hypothesize that in the defect of ABCG5/G8, an ABCG5/G8-independent pathway is essential for regulating hepatic secretion of biliary sterols, which is independent of the lithogenic mechanism of the ABCG5/G8 pathway. To elucidate the effect of the ABCG5/G8-independent pathway on cholelithogenesis, we investigated the biliary and gallstone characteristics in male wild-type (WT), ABCG5(-/-)/G8(-/-), and ABCG8 (-/-) mice fed a lithogenic diet or varying amounts of cholesterol, treated with a liver X receptor (LXR) agonist, or injected intravenously with [(3) H]sitostanol- and [(14) C]cholesterol-labeled high-density lipoprotein (HDL). We found that ABCG5(-/-)/G8(-/-) and ABCG8 (-/-) mice displayed the same biliary and gallstone phenotypes. Although both groups of knockout mice showed a significant reduction in hepatic cholesterol output compared to WT mice, they still formed gallstones. The LXR agonist significantly increased biliary cholesterol secretion and gallstones in WT, but not ABCG5(-/-)/G8(-/-) or ABCG8 (-/-), mice. The 6-hour recovery of [(14) C]cholesterol in hepatic bile was significantly lower in both groups of knockout mice than in WT mice and [(3) H]sitostanol was detected in WT, but not ABCG5(-/-)/G8(-/-) or ABCG8 (-/-), mice. CONCLUSIONS: The ABCG5/G8-independent pathway plays an important role in regulating biliary cholesterol secretion, the transport of HDL-derived cholesterol from plasma to bile, and gallstone formation, which works independently of the ABCG5/G8 pathway. Further studies are needed to observe whether this pathway is also operational in humans. (Hepatology 2016;64:853-864).


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 5/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 8/genetics , Cholesterol/metabolism , Gallstones/etiology , Lipoproteins/genetics , Animals , Bile/metabolism , Bile Acids and Salts/metabolism , Female , Gallbladder/physiology , Gallstones/metabolism , Hydrocarbons, Fluorinated , Lipid Metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Sulfonamides
19.
Eur J Clin Invest ; 47(4): 328-333, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28186337

ABSTRACT

BACKGROUND: Coeliac disease is a chronic, small intestinal, immune-mediated enteropathy caused by a permanent intolerance to dietary gluten in genetically predisposed individuals. Clinical studies have found that intestinal cholecystokinin secretion and gallbladder emptying in response to a fatty meal are impaired before coeliac patients start the gluten-free diet (GFD). DESIGN: However, it was never really appreciated whether coeliac disease is associated with gallstones because there were very few studies investigating the mechanism underlying the impact of coeliac disease on the pathogenesis of gallstones. RESULTS: We summarize recent progress on the relationship between coeliac disease and gallstones and propose that coeliac disease is an important risk factor for gallstone formation because defective intestinal cholecystokinin secretion markedly increases susceptibility to cholesterol gallstones via a mechanism involving dysmotility of both the gallbladder and the small intestine. Because GFD can significantly improve the coeliac enteropathy, early diagnosis and therapy in coeliac patients is crucial for preventing the long-term impact of cholecystokinin deficiency on the biliary and intestinal consequences. When gluten is reintroduced, clinical and histologic relapse often occurs in coeliac patients. Moreover, some of the coeliac patients do not respond well to GFD. CONCLUSIONS: It is imperative to routinely examine by ultrasonography whether gallbladder motility function is preserved in coeliac patients and monitor whether biliary sludge (a precursor of gallstones) appears in the gallbladder, regardless of whether they are under the GFD programme. To prevent gallstones in coeliac patients, it is urgently needed to investigate the prevalence and pathogenesis of gallstones in these patients.


Subject(s)
Celiac Disease/complications , Cholecystokinin/metabolism , Gallstones/etiology , Animals , Celiac Disease/metabolism , Disease Models, Animal , Forecasting , Gallbladder Emptying/physiology , Gallstones/metabolism , Humans , Intestine, Small/metabolism , Mice, Knockout , Receptors, Cholecystokinin/agonists , Receptors, Cholecystokinin/metabolism , Risk Factors
20.
Eur J Clin Invest ; 47(9): 667-674, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28722105

ABSTRACT

BACKGROUND AND AIMS: This study aimed to investigate the relationship between circulating soluble C-X-C chemokine ligand 16 (CXCL16) levels and clinical characteristics of gallstone. METHODS: 93 subjects including 53 subjects with gallstone, 25 subjects with nonalcoholic fatty liver disease (NAFLD), and 40 control subjects were recruited. All gallstone subjects underwent ultrasounds to confirm the gallstone patients. Serum CXCL16 levels and other clinical and biochemical parameters in all subjects were obtained based on standard clinical examination methods. Liver tissues from patients with gallstone undergoing cholecystotomy and healthy subjects were also used to determine the hepatic CXCL16 profiles by IHC staining and real-time quantitative PCR. RESULTS: Serum CXCL16 levels were significantly increased in patients with gallstone and NAFLD as compared to healthy controls (P < 0·001). Hepatic CXCL16 mRNA and protein levels were also significantly increased in gallstone patients following with elevation of hepatic triglycerides and free fatty acid concentration, as compared to those in healthy subjects (P < 0·001). Otherwise, serum CXCL16 levels positively correlated with nonalcoholic fatty liver disease (NAFLD), alanine aminotransferase, aspartate aminotransferase, alkaline phosphatase, gamma-glutamyl transpeptidase (GGT) and direct bilirubin (P < 0·05), but negatively with total protein and albumin after adjustment with age and gender. Multiple stepwise regression analyses indicated that CXCL16 was independently associated with AST, NAFLD and albumin (P < 0·05, respectively). CONCLUSIONS: Serum CXCL16 levels are significantly increased in patients with gallstone, and are independently associated with liver injury in Chinese population, suggesting that CXCL16 may be a biomarker of liver injury in subjects with gallstone or NAFLD.


Subject(s)
Chemokine CXCL16/genetics , Gallstones/genetics , Liver/metabolism , RNA, Messenger/metabolism , Adult , Alanine Transaminase/metabolism , Alkaline Phosphatase/metabolism , Asian People , Aspartate Aminotransferases/metabolism , Case-Control Studies , Chemokine CXCL16/metabolism , Fatty Acids, Nonesterified/metabolism , Female , Gallstones/metabolism , Humans , Immunohistochemistry , Male , Middle Aged , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Real-Time Polymerase Chain Reaction , Triglycerides/metabolism , gamma-Glutamyltransferase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL