Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.816
Filter
Add more filters

Publication year range
1.
Nature ; 626(8000): 852-858, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38326608

ABSTRACT

Bile acids (BAs) are steroid detergents in bile that contribute to the absorption of fats and fat-soluble vitamins while shaping the gut microbiome because of their antimicrobial properties1-4. Here we identify the enzyme responsible for a mechanism of BA metabolism by the gut microbiota involving amino acid conjugation to the acyl-site of BAs, thus producing a diverse suite of microbially conjugated bile acids (MCBAs). We show that this transformation is mediated by acyltransferase activity of bile salt hydrolase (bile salt hydrolase/transferase, BSH/T). Clostridium perfringens BSH/T rapidly performed acyl transfer when provided various amino acids and taurocholate, glycocholate or cholate, with an optimum at pH 5.3. Amino acid conjugation by C. perfringens BSH/T was diverse, including all proteinaceous amino acids except proline and aspartate. MCBA production was widespread among gut bacteria, with strain-specific amino acid use. Species with similar BSH/T amino acid sequences had similar conjugation profiles and several bsh/t alleles correlated with increased conjugation diversity. Tertiary structure mapping of BSH/T followed by mutagenesis experiments showed that active site structure affects amino acid selectivity. These MCBA products had antimicrobial properties, where greater amino acid hydrophobicity showed greater antimicrobial activity. Inhibitory concentrations of MCBAs reached those measured natively in the mammalian gut. MCBAs fed to mice entered enterohepatic circulation, in which liver and gallbladder concentrations varied depending on the conjugated amino acid. Quantifying MCBAs in human faecal samples showed that they reach concentrations equal to or greater than secondary and primary BAs and were reduced after bariatric surgery, thus supporting MCBAs as a significant component of the BA pool that can be altered by changes in gastrointestinal physiology. In conclusion, the inherent acyltransferase activity of BSH/T greatly diversifies BA chemistry, creating a set of previously underappreciated metabolites with the potential to affect the microbiome and human health.


Subject(s)
Acyltransferases , Amidohydrolases , Bile Acids and Salts , Clostridium perfringens , Gastrointestinal Microbiome , Animals , Humans , Mice , Acyltransferases/chemistry , Acyltransferases/metabolism , Alleles , Amidohydrolases/chemistry , Amidohydrolases/metabolism , Amino Acids/metabolism , Anti-Infective Agents/metabolism , Anti-Infective Agents/pharmacology , Bariatric Surgery , Bile Acids and Salts/chemistry , Bile Acids and Salts/metabolism , Catalytic Domain , Clostridium perfringens/enzymology , Clostridium perfringens/metabolism , Feces/chemistry , Gallbladder/metabolism , Gastrointestinal Microbiome/physiology , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Liver/metabolism , Taurocholic Acid/metabolism
2.
Bioorg Chem ; 144: 107132, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38241768

ABSTRACT

The pleiotropic effects of TGR5 make it an appealing target for intervention of metabolic and inflammatory disorders, but systemic activation of TGR5 faces challenges of on-target side effects, especially gallbladder filling. Gut-restricted agonists were proved to be sufficient to circumvent these side effects, but extremely low systemic exposure may not be effective in activating TGR5 since it is located on the basolateral membrane. Herein, to balance potency and physicochemical properties, a series of gut-restricted TGR5 agonists with diversified kinetophores had been designed and synthesized. Compound 22-Na exhibited significant antidiabetic effect, and showed favorable gallbladder safety after 7 days of oral administration in humanized TGR5H88Y mice, confirming that gut-restricted agonism of TGR5 is a viable strategy to alleviate systemic target-related effects.


Subject(s)
Betulinic Acid , Receptors, G-Protein-Coupled , Mice , Animals , Receptors, G-Protein-Coupled/metabolism , Hypoglycemic Agents/pharmacology , Gallbladder/metabolism
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.
Development ; 147(12)2020 06 22.
Article in English | MEDLINE | ID: mdl-32439761

ABSTRACT

The development of the biliary system is a complex yet poorly understood process, with relevance to multiple diseases, including biliary atresia, choledochal cysts and gallbladder agenesis. We present here a crucial role for Hippo-Yap/Taz signaling in this context. Analysis of sav1 mutant zebrafish revealed dysplastic morphology and expansion of both intrahepatic and extrahepatic biliary cells, and ultimately larval lethality. Biliary dysgenesis, but not larval lethality, is driven primarily by Yap signaling. Re-expression of Sav1 protein in sav1-/- hepatocytes is able to overcome these initial deficits and allows sav1-/- fish to survive, suggesting cell non-autonomous signaling from hepatocytes. Examination of sav1-/- rescued adults reveals loss of gallbladder and formation of dysplastic cell masses expressing biliary markers, suggesting roles for Hippo signaling in extrahepatic biliary carcinomas. Deletion of stk3 revealed that the phenotypes observed in sav1 mutant fish function primarily through canonical Hippo signaling and supports a role for phosphatase PP2A, but also suggests Sav1 has functions in addition to facilitating Stk3 activity. Overall, this study defines a role for Hippo-Yap signaling in the maintenance of both intra- and extrahepatic biliary ducts.


Subject(s)
Biliary Tract/metabolism , Protein Serine-Threonine Kinases/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Zebrafish Proteins/metabolism , Acyltransferases , Animals , Animals, Genetically Modified/growth & development , Animals, Genetically Modified/metabolism , Biliary Tract/anatomy & histology , Biliary Tract/growth & development , CRISPR-Cas Systems/genetics , Carboxylic Ester Hydrolases/metabolism , Gallbladder/anatomy & histology , Gallbladder/growth & development , Gallbladder/metabolism , Larva/growth & development , Larva/metabolism , Liver/anatomy & histology , Liver/metabolism , Phenotype , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Serine-Threonine Kinase 3 , Signal Transduction , Trans-Activators/genetics , Transcription Factors/genetics , YAP-Signaling Proteins , Zebrafish/growth & development , Zebrafish/metabolism , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics
5.
Diabetes Obes Metab ; 25(6): 1632-1637, 2023 06.
Article in English | MEDLINE | ID: mdl-36781820

ABSTRACT

AIM: Liraglutide treatment is associated with gallbladder-related disorders and has been shown to delay postprandial gallbladder refilling. The gut hormones cholecystokinin (CCK), fibroblast growth factor 19 (FGF19) and glucagon-like peptide 2 (GLP-2), are known to regulate gallbladder motility and may be implicated in gallbladder-related disorders associated with liraglutide treatment. MATERIALS AND METHODS: In a double-blind, 12-week trial, 52 participants [50% male, age 47.6 ± 10.0 years, body mass index 32.6 ± 3.4 kg/m2 (mean ± standard deviation)] with obesity were randomized 1:1 to once-daily subcutaneous liraglutide (escalated from 0.6 mg to 3.0 mg once-daily) or placebo. During liquid meal tests performed at baseline, after the first dose and following 12 weeks of treatment, we evaluated postprandial gallbladder dynamics and plasma responses of CCK, FGF19 and GLP-2. RESULTS: Liraglutide reduced postprandial FGF19 after the first dose [area under the curve (AUC)0-240 min 24.8 vs. 48.0 min × ng/ml, treatment ratio (TR) (95% confidence interval) 0.52 (0.39; 0.69)] and following 12 weeks of treatment [AUC0-240 min 33.7 vs. 48.5 ng/ml × min, TR 0.69 (0.52; 0.93)]. Liraglutide also reduced postprandial GLP-2 responses (AUC0-240 min 3650 vs. 4894 min × pmol/L, TR 0.75 (0.62; 0.90)] following the first dose as well as after 12 weeks [AUC0-240 min 3760 vs. 4882 min × pmol/L, TR 0.77 (0.60; 0.99)]. Liraglutide increased postprandial responses of CCK after the first dose [AUC0-240 min 762 vs. 670 min × pmol/L; TR 1.14 (0.97; 1.33)] and following 12 weeks of treatment [AUC0-240 min 873 vs. 628 min × pmol/L; TR 1.39 (1.12; 1.73)]. CONCLUSION: Compared with placebo, treatment with liraglutide decreased postprandial FGF19 and GLP-2 concentrations and increased postprandial CCK concentrations, which may explain the delayed postprandial gallbladder refilling observed in individuals with obesity treated with liraglutide.


Subject(s)
Diabetes Mellitus, Type 2 , Liraglutide , Humans , Male , Adult , Middle Aged , Female , Liraglutide/pharmacology , Liraglutide/therapeutic use , Gallbladder/metabolism , Diabetes Mellitus, Type 2/complications , Obesity/complications , Body Mass Index , Postprandial Period , Double-Blind Method , Blood Glucose/metabolism
6.
Lipids Health Dis ; 21(1): 109, 2022 Oct 27.
Article in English | MEDLINE | ID: mdl-36303150

ABSTRACT

BACKGROUND: Hepatic caveolin-1 (CAV1) is reduced in cholesterol gallstone disease (CGD). Mice with CAV1 deficiency were prone to develop CGD. However, it remains unknown whether restored hepatic CAV1 expression prevents the development of CGD. METHODS: C57BL/6 mice were injected with adeno-associated virus 2/8 (AAV2/8) vectors carrying the CAV1 gene (AAV2/8CAV1) via intravenous (i.v.) or intraperitoneal (i.p.) route and then subjected to a lithogenic diet (LD) for 8 weeks. Uninjected mice were used as controls. The functional consequences of rescuing CAV1 expression by either i.v. or i.p. AAV2/8CAV1 treatment for CGD prevention and its subsequent molecular mechanisms were examined. RESULTS: CAV1 expression was reduced in the liver and gallbladder of LD-fed CGD mice. We discovered that AAV2/8CAV1 i.p. delivery results in higher transduction efficiency in the gallbladder than tail vein administration. Although either i.v. or i.p. injection of AAV2/8CAV1 improved liver lipid metabolic abnormalities in CGD mice but did not affect LD feeding-induced bile cholesterol supersaturation. In comparison with i.v. administration route, i.p. administration of AAV2/8CAV1 obviously increased CAV1 protein levels in the gallbladder of LD-fed mice, and i.p. delivery of AAV2/8CAV1 partially improved gallbladder cholecystokinin receptor (CCKAR) responsiveness and impeded bile cholesterol nucleation via the activation of adenosine monophosphate-activated protein kinase (AMPK) signaling, which induced a reduction in gallbladder mucin-1 (MUC1) and MUC5ac expression and gallbladder cholesterol accumulation. CONCLUSION: CGD prevention by i.p. AAV2/8CAV1 injection in LD-fed mice was associated with the improvement of gallbladder stasis, which again supported the notion that supersaturated bile is required but not sufficient for the formation of cholesterol gallstones. Additionally, AAV treatment via the local i.p. injection offers particular advantages over the systemic i.v. route for much more effective gallbladder gene delivery, which will be an excellent tool for conducting preclinical functional studies on the maintenance of normal gallbladder function to prevent CGD.


Subject(s)
Caveolin 1 , Gallstones , Animals , Mice , Caveolin 1/genetics , Caveolin 1/metabolism , Cholesterol/metabolism , Cholesterol, Dietary , Dependovirus/genetics , Dependovirus/metabolism , Gallbladder/metabolism , Gallstones/genetics , Gallstones/prevention & control , Liver/metabolism , Mice, Inbred C57BL
7.
Surg Today ; 52(2): 215-223, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34132887

ABSTRACT

PURPOSES: Congenital biliary dilatation (CBD), defined as pancreaticobiliary maljunction (PBM) with biliary dilatation, is a high risk factor for biliary tract cancer (BTC). KRAS and p53 mutations reportedly affect this process, but the mechanisms are unclear, as is the likelihood of BTC later in life in children with CBD. We investigated potential carcinogenetic pathways in children with CBD compared with adults. METHODS: The subjects of this study were nine children with CBD and 13 adults with PBM (10 dilated, 3 non-dilated) without BTC who underwent extrahepatic bile duct resections, as well as four control patients who underwent pancreaticoduodenectomy for non-biliary cancer. We evaluated expressions of Ki-67, KRAS, p53, histone deacetylase (HDAC) and activation-induced cytidine deaminase (AID) in the biliary tract epithelium immunohistochemically. RESULTS: The Ki-67 labeling index (LI) and expressions of KRAS, p53, HDAC, and AID in the gallbladder epithelium were significantly higher or tended to be higher in both the children with CBD and the adults with PBM than in the controls. CONCLUSIONS: BTC may develop later in children with CBD and in adults with PBM, via HDAC and AID expression and through epigenetic and genetic regulation.


Subject(s)
Biliary Tract Neoplasms/etiology , Biliary Tract Neoplasms/genetics , Choledochal Cyst/complications , Choledochal Cyst/genetics , Epithelium/metabolism , Gene Expression Regulation, Neoplastic/genetics , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Adult , Aged , Child , Child, Preschool , Female , Gallbladder/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Infant , Male , Middle Aged , Pancreaticobiliary Maljunction/surgery , Risk
8.
Br Poult Sci ; 63(4): 548-551, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35187989

ABSTRACT

1. The proximate composition and, amino acid, fatty acid and mineral profiles of gallbladder bile of 35 d old broiler chickens were determined.2. The dry matter (DM) content of the bile was determined to be 19.8 g/100 g. Fat, protein and ash contents in the bile were 24.9, 23.0 and 6.6 g/100 g DM, respectively.3. The content of total fatty acids (FA) in the bile was determined to be 17.68 g/100 g DM. Unsaturated FA dominated the profile in the bile, with an unsaturated: saturated FA ratio of 1.36. Oleic, linoleic, and arachidonic acids were the main unsaturated FA, whereas palmitic and stearic acids were the major saturated FA.4. A major finding was that the data established taurine as the primary amino acid in chicken bile and not glycine, as previously assumed. Taurine was the dominant amino acid, constituting 62% of bile protein.5. Among the major minerals, sodium (2.56 g/100 g DM) was in the greatest concentration. The concentrations of other major minerals were relatively low.6. The present work provided, for the first time, preliminary data on reference values for nutrients in the chicken gallbladder bile.


Subject(s)
Bile , Chickens , Amino Acids , Animals , Bile/metabolism , Chickens/metabolism , Fatty Acids/metabolism , Fatty Acids, Unsaturated , Gallbladder/metabolism , Minerals , Taurine
9.
Cancer ; 127(8): 1293-1300, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33289918

ABSTRACT

BACKGROUND: Biliary tract cancer (BTC) has a poor prognosis despite treatment with first-line gemcitabine and cisplatin. In BTC, PI3K/AKT pathway activation has been shown to increase resistance to chemotherapy, which may be overcome with PI3K inhibition. This phase 2 study evaluated the safety and efficacy of copanlisib, a PI3K inhibitor, with gemcitabine and cisplatin in advanced BTCs. The role of PTEN expression in outcomes was also explored. METHODS: Patients with advanced/unresectable BTC received gemcitabine, cisplatin, and copanlisib as their first-line treatment. The primary endpoint was progression-free survival (PFS) at 6 months. Secondary endpoints were the response rate (RR), median overall survival (OS)/PFS, and safety profile. An assessment of PTEN expression by immunohistochemistry was also performed along with molecular profiling. RESULTS: Twenty-four patients received at least 1 dose of the study drug. The PFS rate at 6 months was 51%; the median OS was 13.7 months (95% CI, 6.8-18.0 months), and the median PFS was 6.2 months (95% CI, 2.9-10.1 months). Nineteen patients were evaluable for RR: 6 patients achieved a partial response (31.6%), and 11 (57.9%) had stable disease. The most common grade 3/4 adverse events were a decreased neutrophil count (45.83%), anemia (25%), increased lipase (25%), and hypertension (20.8%). Twenty patients had tissue evaluable for the PTEN status. The PFS for low (n = 9) and high PTEN expression (n = 11) was 8.5 and 4.6 months, respectively (P = .19). The median OS for low and high PTEN expression groups was 17.9 and 7.0 months, respectively (P = .19). CONCLUSIONS: The addition of copanlisib to gemcitabine and cisplatin does not improve PFS at 6 months. However, future studies using PTEN as a potential biomarker should be considered. LAY SUMMARY: The addition of copanlisib, a PI3K inhibitor, to standard chemotherapy for advanced biliary tract cancers was assessed for efficacy and safety. Twenty-four patients with advanced biliary tract cancer received treatment in this study. There was no difference in survival with the addition of copanlisib in comparison with standard chemotherapy. Copanlisib may be more effective and increase survival in patients with low PTEN expression levels. Further studies are needed to confirm this. No unexpected adverse events occurred.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biliary Tract Neoplasms/drug therapy , Cholangiocarcinoma/drug therapy , Cisplatin/therapeutic use , Deoxycytidine/analogs & derivatives , Pyrimidines/therapeutic use , Quinazolines/therapeutic use , Adult , Aged , Aged, 80 and over , Biliary Tract Neoplasms/metabolism , Biliary Tract Neoplasms/pathology , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/pathology , Deoxycytidine/therapeutic use , Disease-Free Survival , Female , Gallbladder/drug effects , Gallbladder/metabolism , Gallbladder/pathology , High-Throughput Nucleotide Sequencing , Humans , Male , Middle Aged , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphoinositide-3 Kinase Inhibitors/therapeutic use , Precision Medicine , Progression-Free Survival , Gemcitabine
10.
Am J Physiol Gastrointest Liver Physiol ; 321(4): G243-G251, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34259574

ABSTRACT

The gallbladder is considered an important organ in maintaining digestive and metabolic homeostasis. Given that therapeutic options for gallbladder diseases are often limited to cholecystectomy, understanding gallbladder pathophysiology is essential in developing novel therapeutic strategies. Since liver X receptor ß (LXRß), an oxysterol-activated transcription factor, is strongly expressed in gallbladder cholangiocytes, the aim was to investigate LXRß physiological function in the gallbladder. Thus, we studied the gallbladders of WT and LXRß-/- male mice using immunohistochemistry, electron microscopy, qRT-PCR, bile duct cannulation, bile and blood biochemistry, and duodenal pH measurements. LXRß-/- mice presented a large gallbladder bile volume with high duodenal mRNA levels of the vasoactive intestinal polypeptide (VIP), a strong mediator of gallbladder relaxation. LXRß-/- gallbladders showed low mRNA and protein expression of Aquaporin-1, Aquaporin-8, and cystic fibrosis transmembrane conductance regulator (CFTR). A cystic fibrosis-resembling phenotype was evident in the liver showing high serum cholestatic markers and the presence of reactive cholangiocytes. For LXRß being a transcription factor, we identified eight putative binding sites of LXR on the promoter and enhancer of the Cftr gene, suggesting Cftr as a novel LXRß regulated gene. In conclusion, LXRß was recognized as a regulator of gallbladder bile volume through multiple mechanisms involving CFTR and aquaporins.NEW & NOTEWORTHY This report reveals a novel and specific role of the nuclear receptor liver X receptor ß (LXRß) in controlling biliary tree pathophysiology. LXRß-/- mice have high gallbladder bile volume and are affected by a cholangiopathy that resembles cystic fibrosis. We found LXRß to regulate the expression of both aquaporins water channels and the cystic fibrosis transmembrane conductance regulator. This opens a new field in biliary tree pathophysiology, enlightening a possible transcription factor controlling CFTR expression.


Subject(s)
Aquaporin 1/metabolism , Aquaporins/metabolism , Bile/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Gallbladder/metabolism , Liver X Receptors/metabolism , Animals , Aquaporin 1/genetics , Aquaporins/genetics , Binding Sites , Cell Proliferation , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Duodenum/metabolism , Gallbladder/ultrastructure , Liver X Receptors/genetics , Male , Mice, Knockout , Promoter Regions, Genetic , Vasoactive Intestinal Peptide/genetics , Vasoactive Intestinal Peptide/metabolism
11.
Mol Syst Biol ; 16(7): e9610, 2020 07.
Article in English | MEDLINE | ID: mdl-32715618

ABSTRACT

The novel SARS-coronavirus 2 (SARS-CoV-2) poses a global challenge on healthcare and society. For understanding the susceptibility for SARS-CoV-2 infection, the cell type-specific expression of the host cell surface receptor is necessary. The key protein suggested to be involved in host cell entry is angiotensin I converting enzyme 2 (ACE2). Here, we report the expression pattern of ACE2 across > 150 different cell types corresponding to all major human tissues and organs based on stringent immunohistochemical analysis. The results were compared with several datasets both on the mRNA and protein level. ACE2 expression was mainly observed in enterocytes, renal tubules, gallbladder, cardiomyocytes, male reproductive cells, placental trophoblasts, ductal cells, eye, and vasculature. In the respiratory system, the expression was limited, with no or only low expression in a subset of cells in a few individuals, observed by one antibody only. Our data constitute an important resource for further studies on SARS-CoV-2 host cell entry, in order to understand the biology of the disease and to aid in the development of effective treatments to the viral infection.


Subject(s)
Peptidyl-Dipeptidase A/metabolism , Respiratory System/metabolism , Angiotensin-Converting Enzyme 2 , Betacoronavirus , Blood Vessels/metabolism , Conjunctiva/metabolism , Enterocytes/metabolism , Female , Gallbladder/metabolism , Host Microbial Interactions , Humans , Immunohistochemistry , Kidney Tubules, Proximal/metabolism , Male , Mass Spectrometry , Myocytes, Cardiac/metabolism , Organ Specificity , Peptidyl-Dipeptidase A/genetics , Placenta/metabolism , Pregnancy , RNA-Seq , SARS-CoV-2 , Single-Cell Analysis , Testis/metabolism
12.
PLoS Biol ; 16(7): e2006682, 2018 07.
Article in English | MEDLINE | ID: mdl-30048457

ABSTRACT

The gut-to-brain axis exhibits significant control over motivated behavior. However, mechanisms supporting this communication are poorly understood. We reveal that a gut-based bariatric surgery chronically elevates systemic bile acids and attenuates cocaine-induced elevations in accumbal dopamine. Notably, this surgery reduces reward-related behavior and psychomotor sensitization to cocaine. Utilizing a knockout mouse model, we have determined that a main mediator of these post-operative effects is the Takeda G protein-coupled bile acid receptor (TGR5). Viral restoration of TGR5 in the nucleus accumbens of TGR5 knockout animals is sufficient to restore cocaine reward, centrally localizing this TGR5-mediated modulation. These findings define TGR5 and bile acid signaling as pharmacological targets for the treatment of cocaine abuse and reveal a novel mechanism of gut-to-brain communication.


Subject(s)
Bariatric Surgery , Bile/metabolism , Cocaine/pharmacology , Reward , Signal Transduction , Animals , Behavior, Animal , Choice Behavior/drug effects , Dopamine/metabolism , Gallbladder/metabolism , Ileum/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Nucleus Accumbens/metabolism
13.
Pharm Res ; 38(12): 2035-2046, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34862570

ABSTRACT

PURPOSE: To estimate hepatobiliary clearances of rosuvastatin via simultaneously fitting to reported human positron emission tomography (PET) data in the liver and gallbladder. METHODS: A hepatobiliary model incorporating five intrinsic hepatobiliary clearances (active uptake clearance at the sinusoidal membrane, efflux clearance by passive diffusion through the sinusoidal membrane, influx clearance by passive diffusion through sinusoidal membrane, clearance of biliary excretion at the canalicular membrane, and intercompartment clearance from the intrahepatic bile duct to the gallbladder) and three compartments (liver, intrahepatic bile duct, and gallbladder) was developed to simultaneously fit rosuvastatin liver and gallbladder data from a representative subject reported by Billington et al. (1). Two liver blood supply input functions, arterial input function and dual input function (using peripheral venous as an alternative to portal vein), were assessed. Additionally, the predictive performance between the established model and four reported models trained with only systemic exposure data, was evaluated by comparing simulated liver and gallbladder profiles with observations. RESULTS: The established hepatobiliary model well captured the kinetic profiles of rosuvastatin in the liver and gallbladder during the PET scans. Application of dual input function led to a marked underestimation of liver concentrations at the initial stage after i.v. dosing which cannot be offset by altering model parameter values. The simulated hepatobiliary profiles from three of the reported models demonstrated substantial deviation from the observed data. CONCLUSIONS: The present study highlights the necessity of using hepatobiliary data to verify and improve the predictive performance of hepatic disposition of rosuvastatin.


Subject(s)
Gallbladder/metabolism , Hepatobiliary Elimination , Liver/metabolism , Rosuvastatin Calcium/pharmacokinetics , Datasets as Topic , Gallbladder/diagnostic imaging , Humans , Liver/diagnostic imaging , Models, Biological , Positron-Emission Tomography , Tissue Distribution
14.
Acta Haematol ; 144(2): 229-235, 2021.
Article in English | MEDLINE | ID: mdl-33017829

ABSTRACT

Histiocytic sarcoma (HS) is a rare, malignant, and aggressive subtype of histiocytosis. We present an unusual case of aggressive HS presenting in the gastrointestinal tract and gallbladder that progressed after several lines of chemotherapy with a leukemic phase. We review the clinical, pathological, and molecular characteristics of HS in this case and review the literature on HS involving the digestive system as well as on overt leukemic phase of this disease. HS is often diagnosed at an advanced stage, and mortality is high. We discuss the therapeutic approach to patients with HS. We highlight the role of overexpression and somatic alterations in the RAF-MEK-ERK pathway in the pathogenesis of HS and discuss potential targeted approaches to treat these rare tumors.


Subject(s)
Gastrointestinal Neoplasms/diagnosis , Histiocytic Sarcoma/diagnosis , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cholangiopancreatography, Magnetic Resonance , Cholecystectomy , Gallbladder/metabolism , Gallbladder/pathology , Gastrointestinal Neoplasms/diagnostic imaging , Gastrointestinal Neoplasms/drug therapy , Histiocytic Sarcoma/diagnostic imaging , Histiocytic Sarcoma/drug therapy , Humans , Male , Positron-Emission Tomography , Tomography, X-Ray Computed
15.
Lipids Health Dis ; 20(1): 26, 2021 Mar 22.
Article in English | MEDLINE | ID: mdl-33752687

ABSTRACT

BACKGROUND: The purpose of this study was to assess the risk factors for cholesterol polyp formation in the gallbladder. METHODS: This was a multicenter retrospective study based on pathology. From January 2016 to December 2019, patients who underwent cholecystectomy and non-polyp participants confirmed by continuous ultrasound follow-ups were reviewed. Patients in the cholesterol polyp group were recruited from three high-volume centers with a diagnosis of pathologically confirmed cholesterol polyps larger than 10 mm. Population characteristics and medical data were collected within 24 h of admission before surgery. The non-polyp group included participants from the hospital physical examination center database. They had at least two ultrasound examinations with an interval longer than 180 days. Data from the final follow-up of the non-polyp group were analyzed. The risk factors for cholesterol polyp formation were analyzed by comparing the two groups. RESULTS: A total of 4714 participants were recruited, including 376 cholesterol polyp patients and 4338 non-polyp participants. In univariate analysis, clinical risk factors for cholesterol polyps were age, male sex, higher body mass index (BMI), higher low-density lipoprotein (LDL), lower high-density lipoprotein (HDL), and higher aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels. In multivariate logistic analysis, independent risk factors were age > 50 years (odds ratio [OR] = 3.02, 95% confidence interval [CI] 2.33-3.91, P < 0.001], LDL > 2.89 mmol/L (OR = 1.38, 95% CI 1.08-1.78, P = 0.011), lower HDL (OR = 1.78 95% CI 1.32-2.44, P < 0.001), AST > 40 IU/L (OR = 3.55, 95% CI 2.07-6.07, P < 0.001), and BMI > 25 kg/m 2 (OR = 1.32, 95% CI 1.01-1.72, P = 0.037). CONCLUSIONS: Age, LDL, HDL, AST, and BMI are strong risk factors for cholesterol polyp formation. Older overweight patients with polyps, accompanied by abnormal lipid levels, are at high risk for cholesterol polyps.


Subject(s)
Cholesterol/metabolism , Gallbladder/metabolism , Gallbladder/pathology , Lipid Metabolism , Polyps/metabolism , Polyps/pathology , Female , Humans , Logistic Models , Male , Middle Aged , Risk Factors
16.
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
17.
Ann Hepatol ; 26: 100563, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34653690

ABSTRACT

INTRODUCTION AND OBJECTIVES: Long non-coding RNA (lncRNA) EPIC1 (epigenetically-induced lncRNA1) is likely involved in human cancer by promoting cell cycle progression. Our study was carried out to investigate the involvement of EPIC1 in gallbladder cancer (GBC). METHODS: Expression levels of EPIC1 in two types of tissues (GBC and paracancerous) and plasma were measured by performing qPCR. GBC-SD and SGC-996 cells were transfected with low expression in tumor (LET) and EPIC1 expression vectors. RESULTS: The present study found that EPIC1 was upregulated in tumor tissues than in paracancerous tissues of GBC patients, and plasma levels of EPIC1 were significantly correlated with levels of EPIC1 in tumor tissues. LncRNA LET was downregulated in tumor tissues than in paracancerous tissues and was inversely correlated with EPIC1 in both tumor tissues and paracancerous tissues. Overexpression of EPIC1 led to downregulated LET, and LET overexpression also mediated the downregulation of EPIC1. EPIC1 led to accelerated GBC cell proliferation and inhibited apoptosis. Overexpression of LET played opposites roles. In addition, LET overexpression attenuated the effects of EPIC1 overexpression on cancer cell proliferation and apoptosis. CONCLUSIONS: LncRNA EPIC1 promoted proliferation and inhibited apoptosis of GBC cells by interacting with LET.


Subject(s)
Apoptosis/genetics , Down-Regulation , Gallbladder Neoplasms/genetics , Gallbladder/pathology , RNA, Long Noncoding/genetics , Up-Regulation , Aged , Cell Proliferation/genetics , Female , Follow-Up Studies , Gallbladder/metabolism , Gallbladder Neoplasms/metabolism , Gallbladder Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Retrospective Studies , Tumor Cells, Cultured
18.
Int J Mol Sci ; 22(4)2021 Feb 10.
Article in English | MEDLINE | ID: mdl-33579036

ABSTRACT

Bile acids (BAs) are facial amphiphiles synthesized in the body of all vertebrates. They undergo the enterohepatic circulation: they are produced in the liver, stored in the gallbladder, released in the intestine, taken into the bloodstream and lastly re-absorbed in the liver. During this pathway, BAs are modified in their molecular structure by the action of enzymes and bacteria. Such transformations allow them to acquire the chemical-physical properties needed for fulling several activities including metabolic regulation, antimicrobial functions and solubilization of lipids in digestion. The versatility of BAs in the physiological functions has inspired their use in many bio-applications, making them important tools for active molecule delivery, metabolic disease treatments and emulsification processes in food and drug industries. Moreover, moving over the borders of the biological field, BAs have been largely investigated as building blocks for the construction of supramolecular aggregates having peculiar structural, mechanical, chemical and optical properties. The review starts with a biological analysis of the BAs functions before progressively switching to a general overview of BAs in pharmacology and medicine applications. Lastly the focus moves to the BAs use in material science.


Subject(s)
Bile Acids and Salts/metabolism , Animals , Bile Acids and Salts/analysis , Bile Acids and Salts/chemical synthesis , Cholesterol/metabolism , Gallbladder/metabolism , Humans , Intestinal Mucosa/metabolism , Lipid Metabolism , Liver/metabolism
19.
Int J Mol Sci ; 22(4)2021 Feb 06.
Article in English | MEDLINE | ID: mdl-33562048

ABSTRACT

The expression of monocarboxylate transporters (MCTs) is linked to pathophysiological changes in diseases, including cancer, such that MCTs could potentially serve as diagnostic markers or therapeutic targets. We recently developed [18F]FACH as a radiotracer for non-invasive molecular imaging of MCTs by positron emission tomography (PET). The aim of this study was to evaluate further the specificity, metabolic stability, and pharmacokinetics of [18F]FACH in healthy mice and piglets. We measured the [18F]FACH plasma protein binding fractions in mice and piglets and the specific binding in cryosections of murine kidney and lung. The biodistribution of [18F]FACH was evaluated by tissue sampling ex vivo and by dynamic PET/MRI in vivo, with and without pre-treatment by the MCT inhibitor α-CCA-Na or the reference compound, FACH-Na. Additionally, we performed compartmental modelling of the PET signal in kidney cortex and liver. Saturation binding studies in kidney cortex cryosections indicated a KD of 118 ± 12 nM and Bmax of 6.0 pmol/mg wet weight. The specificity of [18F]FACH uptake in the kidney cortex was confirmed in vivo by reductions in AUC0-60min after pre-treatment with α-CCA-Na in mice (-47%) and in piglets (-66%). [18F]FACH was metabolically stable in mouse, but polar radio-metabolites were present in plasma and tissues of piglets. The [18F]FACH binding potential (BPND) in the kidney cortex was approximately 1.3 in mice. The MCT1 specificity of [18F]FACH uptake was confirmed by displacement studies in 4T1 cells. [18F]FACH has suitable properties for the detection of the MCTs in kidney, and thus has potential as a molecular imaging tool for MCT-related pathologies, which should next be assessed in relevant disease models.


Subject(s)
Drug Evaluation, Preclinical/methods , Lactic Acid/metabolism , Monocarboxylic Acid Transporters/metabolism , Positron-Emission Tomography/methods , Radiopharmaceuticals/pharmacology , Animals , Cell Line, Tumor , Female , Fluorine Radioisotopes/chemistry , Gallbladder/metabolism , Kidney/metabolism , Liver/metabolism , Mice , Monocarboxylic Acid Transporters/antagonists & inhibitors , Rats , Swine
20.
Fish Physiol Biochem ; 47(4): 1199-1209, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34173184

ABSTRACT

This study aimed to examine the effects of dietary digested soybean protein (DSP) and taurine on bile acid (BA) level, lipase activity, lipid apparent digestibility coefficient (ADC), and growth performance of pompano (Trachinotus blochii). Five diets were formulated with fish meal (FM), defatted soybean meal (SBM), and the DSP as main dietary protein sources. The diets were denoted as follows: FMD (FM-based diet), SBMD (SBM-based diet), SBM+TD (SBM-based diet plus taurine), DSPD (DSP-based diet), and DSP+TD (DSP-based diet plus taurine). Fingerling pompano with an initial body weight (BW) of 21.4 g were stocked in 500-L tanks, with triplicate tanks per dietary treatment. For 8 weeks, the fish were hand-fed the experimental diets to apparent satiation twice daily. The results showed that the DSPD and DSP+TD groups had significantly higher final BW, weight gain, and specific growth rate, but lower feed conversion ratio, than the SBMD and SBM+TD groups, respectively (P < 0.05). There were no significant differences in growth and feed performances between fish fed DSP+TD and FMD. The gallbladder and anterior intestinal BA levels, anterior intestinal lipase activity, and lipid and protein ADCs were markedly increased in fish fed DSPD and DSP+TD compared to those fed SBMD (P < 0.05), and no significant differences were detected between the DSP+TD and FMD groups. The findings of the present study suggested that dietary DSP inclusion with taurine supplementation might effectively improve lipid digestion and this contributed to growth enhancement in pompano fed a soybean protein-based diet.


Subject(s)
Fishes , Soybean Proteins/pharmacology , Taurine/pharmacology , Animal Feed , Animals , Bile Acids and Salts/metabolism , Diet , Fish Proteins/metabolism , Fishes/growth & development , Fishes/metabolism , Gallbladder/drug effects , Gallbladder/metabolism , Lipase/metabolism , Lipid Metabolism/drug effects , Liver/drug effects , Liver/metabolism , Muscles/drug effects , Muscles/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL