Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 123
Filter
1.
J Appl Toxicol ; 44(6): 863-873, 2024 06.
Article in English | MEDLINE | ID: mdl-38311468

ABSTRACT

Although the medicinal properties of colchicine (COL) have been widely known for centuries, its toxicity has been the subject of controversy. The narrow therapeutic window causes COL to induce gastrointestinal adverse effects even when taken at recommended doses, mainly manifested as nausea, vomiting, and diarrhea. However, the mechanism of COL-induced gastrointestinal toxic reactions remains obscure. In the present study, the mice were dosed with COL (2.5 mg/kg b.w./day) for a week to explore the effect of COL on bile acid metabolism and the mechanism of COL-induced diarrhea. The results showed that COL treatment affected liver biochemistry in mice, resulting in a significant down-regulation of the mRNA expression levels of bile acid biosynthesis regulators Cyp7a1, Cyp8b1, Cyp7b1, and Cyp27a1 in liver tissues. The mRNA expression levels of bile acid transporters Ntcp, Oatp1, Mrp2, Ibabp, Mrp3, Osta, and Ostb in liver and ileum tissues were also significantly down-regulated. In addition, COL treatment significantly inhibited the mRNA expression levels of Fxr and its downstream target genes Shp, Lrh1, and Fgf15 in liver and ileum tissues, affecting the feedback regulation of bile acid biosynthesis. More importantly, the inhibition of COL on bile acid transporters in ileal and hepatic tissues affected bile acid recycling in the ileum as well as their reuptake in the liver, leading to a significantly increased accumulation of bile acids in the colon, which may be an important cause of diarrhea. In conclusion, our study revealed that COL treatment affected bile acid biosynthesis and enterohepatic circulation, thereby disrupting bile acid metabolic homeostasis in mice.


Subject(s)
Bile Acids and Salts , Colchicine , Enterohepatic Circulation , Homeostasis , Liver , Animals , Bile Acids and Salts/metabolism , Enterohepatic Circulation/drug effects , Colchicine/toxicity , Homeostasis/drug effects , Liver/drug effects , Liver/metabolism , Mice , Male , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Cytoplasmic and Nuclear/drug effects , Receptors, Cytoplasmic and Nuclear/genetics , Ileum/drug effects , Ileum/metabolism , Diarrhea/chemically induced
2.
Yakugaku Zasshi ; 143(2): 101-104, 2023.
Article in Japanese | MEDLINE | ID: mdl-36724921

ABSTRACT

The evaluation and prediction of pharmacokinetics in humans is important in the field of drug discovery and development. Generally, human pharmacokinetics is predicted using physiologically based pharmacokinetic models that include physiological and physicochemical (drug) parameters obtained from in vitro assays. Specific organ dysfunction, such as liver disease, also affects the functions of other organs, causing unexpected pharmacokinetic fluctuations. I investigated the effect of cholestasis on intestinal drug absorption in mice subjected to bile duct ligation (BDL). The intestinal absorption and permeability of imatinib was decreased in BDL mice compared with sham-operated mice, and this may be attributed to the up-regulation of the efflux transporter, breast cancer resistance protein. However, a single-organ experimental system cannot predict such pharmacokinetic changes. To overcome this challenge, I investigated a microphysiological system (MPS) equipped with intestinal and hepatic cells for pharmacokinetic evaluation. The glucuronidation of triazolam was significantly increased in an enterohepatic MPS compared with a single-culture system. These results suggested that the elucidation of organ interactions requires the use of an MPS loaded with human cells in combination with laboratory animal studies. In this review, I present the results of my evaluation of organ interactions using animal models and MPSs in the Award for Young Scientists from the Pharmaceutical Society of Japan, Hokuriku Branch.


Subject(s)
Cholestasis , Enterohepatic Circulation , Liver , Animals , Humans , Mice , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Cholestasis/metabolism , Intestines , Liver/drug effects , Liver/metabolism , Neoplasm Proteins/metabolism , Pharmacokinetics , Enterohepatic Circulation/drug effects , Enterohepatic Circulation/physiology
3.
Gut Microbes ; 13(1): 1949095, 2021.
Article in English | MEDLINE | ID: mdl-34313539

ABSTRACT

Farnesoid X receptor (FXR) is a nuclear receptor for bile acids (BAs) that is widely expressed in the intestine, liver and kidney. FXR has important regulatory impacts on a wide variety of metabolic pathways (such as glucose, lipid, and sterol metabolism) and has been recognized to ameliorate obesity, liver damage, cholestasis and chronic inflammatory diseases. The types of BAs are complex and diverse. BAs link the intestine with the liver through the enterohepatic circulation. BAs derivatives have entered clinical trials for liver disease. In addition to the liver, the intestine is also targeted by BAs. This article reviews the effects of different BAs on the intestinal tract through the enterohepatic circulation from the perspective of FXR, aiming to elucidate the effects of different BAs on the intestinal tract and lay a foundation for new treatment methods.


Subject(s)
Bile Acids and Salts/metabolism , Bile Acids and Salts/pharmacokinetics , Bile Acids and Salts/therapeutic use , Enterohepatic Circulation/drug effects , Gastrointestinal Microbiome/drug effects , Intestinal Diseases/drug therapy , Intestines/drug effects , Animals , Disease Models, Animal , Humans
4.
Dig Dis Sci ; 66(2): 568-576, 2021 02.
Article in English | MEDLINE | ID: mdl-32198567

ABSTRACT

BACKGROUND: A Western diet is a risk factor for the development of inflammatory bowel disease (IBD). High levels of fecal deoxycholic acid (DCA) in response to a Western diet contribute to bowel inflammatory injury. However, the mechanism of DCA in the natural course of IBD development remains unanswered. AIMS: The aim of this study is to investigate the effect of DCA on the induction of gut dysbiosis and its roles in the development of intestinal inflammation. METHODS: Wild-type C57BL/6J mice were fed an AIN-93G diet, either supplemented with or without 0.2% DCA, and killed at 24 weeks. Distal ileum and colon tissues were assessed by histopathological analysis. Hepatic and ileal gene expression was examined by qPCR, and the gut microbiota was analyzed by high-throughput 16S rRNA gene sequencing. HPLC-MS was used for fecal bile acid quantification. RESULTS: Mice fed the DCA-supplemented diet developed focal areas of ileal and colonic inflammation, accompanied by alteration of the composition of the intestinal microbiota and accumulation of fecal bile acids. DCA-induced dysbiosis decreased the deconjugation of bile acids, and this regulation was associated with the repressed expression of target genes in the enterohepatic farnesoid X receptor-fibroblast growth factor (FXR-FGF15) axis, leading to upregulation of hepatic de novo bile acid synthesis. CONCLUSIONS: These results suggest that DCA-induced gut dysbiosis may act as a key etiologic factor in intestinal inflammation, associated with bile acid metabolic disturbance and downregulation of the FXR-FGF15 axis.


Subject(s)
Bile Acids and Salts/metabolism , Deoxycholic Acid/toxicity , Diet, Western/adverse effects , Dysbiosis/metabolism , Enterohepatic Circulation/physiology , Inflammatory Bowel Diseases/metabolism , Animals , Deoxycholic Acid/administration & dosage , Dysbiosis/chemically induced , Dysbiosis/pathology , Enterohepatic Circulation/drug effects , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/pathology , Mice , Mice, Inbred C57BL
5.
Hepatology ; 73(6): 2577-2585, 2021 06.
Article in English | MEDLINE | ID: mdl-33222321

ABSTRACT

Bile salts play a pivotal role in lipid homeostasis, are sensed by specialized receptors, and have been implicated in various disorders affecting the gut or liver. They may play a role either as culprit or as potential panacea. Four very efficient transporters mediate most of the hepatic and intestinal bile salt uptake and efflux, and are each essential for the efficient enterohepatic circulation of bile salts. Starting from the intestinal lumen, conjugated bile salts cross the otherwise impermeable lipid bilayer of (primarily terminal ileal) enterocytes through the apical sodium-dependent bile acid transporter (gene SLC10A2) and leave the enterocyte through the basolateral heteromeric organic solute transporter, which consists of an alpha and beta subunit (encoded by SLC51A and SLC51B). The Na+ -taurocholate cotransporting polypeptide (gene SLC10A1) efficiently clears the portal circulation of bile salts, and the apical bile salt export pump (gene ABCB11) pumps the bile salts out of the hepatocyte into primary bile, against a very steep concentration gradient. Recently, individuals lacking either functional Na+ -taurocholate cotransporting polypeptide or organic solute transporter have been described, completing the quartet of bile acid transport deficiencies, as apical sodium-dependent bile acid transporter and bile salt export pump deficiencies were already known for years. Novel pathophysiological insights have been obtained from knockout mice lacking functional expression of these genes and from pharmacological transporter inhibition in mice or humans. Conclusion: We provide a concise overview of the four main bile salt transport pathways and of their status as possible targets of interventions in cholestatic or metabolic disorders.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 11 , Bile Acids and Salts/metabolism , Enterohepatic Circulation/physiology , Membrane Transport Proteins , Organic Anion Transporters, Sodium-Dependent , Receptors, G-Protein-Coupled , Symporters , ATP Binding Cassette Transporter, Subfamily B, Member 11/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 11/metabolism , Animals , Biological Transport, Active/drug effects , Biological Transport, Active/physiology , Drug Development , Enterohepatic Circulation/drug effects , Humans , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Organic Anion Transporters, Sodium-Dependent/antagonists & inhibitors , Organic Anion Transporters, Sodium-Dependent/genetics , Organic Anion Transporters, Sodium-Dependent/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Symporters/antagonists & inhibitors , Symporters/genetics , Symporters/metabolism
6.
Nutrients ; 12(5)2020 May 05.
Article in English | MEDLINE | ID: mdl-32380648

ABSTRACT

Chorioamnionitis can lead to inflammation and injury of the liver and gut, thereby predisposing patients to adverse outcomes such as necrotizing enterocolitis (NEC). In addition, intestinal bile acids (BAs) accumulation is causally linked to NEC development. Plant sterols are a promising intervention to prevent NEC development, considering their anti-inflammatory properties in the liver. Therefore, we investigated whether an intra-amniotic (IA) Ureaplasma parvum (UP) infection affected the liver and enterohepatic circulation (EHC) and evaluated whether an IA administered plant sterol mixture dissolved in ß-cyclodextrin exerted prophylactic effects. An ovine chorioamnionitis model was used in which liver inflammation and the EHC were assessed following IA UP exposure in the presence or absence of IA prophylactic plant sterols (a mixture of ß-sitosterol and campesterol dissolved in ß-cyclodextrin (carrier)) or carrier alone. IA UP exposure caused an inflammatory reaction in the liver, histologically seen as clustered and conflated hepatic erythropoiesis in the parenchyma, which was partially prevented by IA administration of sterol + ß-cyclodextrin, or ß-cyclodextrin alone. In addition, IA administration of ß-cyclodextrin prior to UP caused changes in the expression of several hepatic BAs transporters, without causing alterations in other aspects of the EHC. Thereby, the addition of plant sterols to the carrier ß-cyclodextrin did not have additional effects.


Subject(s)
Cholesterol/analogs & derivatives , Chorioamnionitis/drug therapy , Chorioamnionitis/microbiology , Drug Carriers , Enterocolitis, Necrotizing/microbiology , Enterocolitis, Necrotizing/prevention & control , Enterohepatic Circulation/drug effects , Fetus/blood supply , Liver/blood supply , Phytosterols/administration & dosage , Phytotherapy , Post-Exposure Prophylaxis/methods , Sitosterols/administration & dosage , Ureaplasma Infections , Ureaplasma , beta-Cyclodextrins , Animals , Cholesterol/administration & dosage , Cholesterol/pharmacology , Disease Models, Animal , Female , Inflammation , Injections, Intralesional , Phytosterols/pharmacology , Pregnancy , Sheep , Sitosterols/pharmacology
7.
Alcohol Alcohol ; 55(3): 264-277, 2020 Apr 16.
Article in English | MEDLINE | ID: mdl-32232424

ABSTRACT

AIMS: To investigate the effect of ethanol intake on the whole enterohepatic circulation (EHC) of bile acids (BAs) and, more importantly, on pharmacokinetics of irinotecan. METHODS: The present study utilized a mouse model administered by gavage with 0 (control), 240 mg/100 g (30%, v/v) and 390 mg/100 g (50%, v/v) ethanol for 6 weeks, followed by BA profiles in the whole EHC (including liver, gallbladder, intestine and plasma) and colon using ultra-high performance liquid chromatography with tandem mass spectrometry analysis. Pharmacokinetic parameters of irinotecan were measured after administration of irinotecan (i.v. 5 mg/kg) on alcohol-treated mice. RESULTS: The results showed that compared with the control group, concentrations of most free-BAs, total amount of the three main forms of BAs (free-BA, taurine-BA and glycine-BA) and total BAs (TBAs) in 50% ethanol intake group were significantly increased, which are mostly attributed to the augmentation of free-BAs and taurine-BAs. Additionally, the TBAs in liver and gallbladder and the BA pool were markedly increased in the 30% ethanol intake group. Importantly, ethanol intake upregulated the expression of BA-related enzymes (Cyp7a1, Cyp27a1, Cyp8b1 and Baat) and transporters (Bsep, Mrp2, P-gp and Asbt) and downregulated the expression of transporter Ntcp and nuclear receptor Fxr in the liver and ileum, respectively. Additionally, 50% ethanol intake caused fairly distinct liver injury. Furthermore, the AUC0-24 h of irinotecan and SN38 were significantly reduced but their clearance was significantly increased in the disrupted EHC of BA by 50% ethanol intake. CONCLUSIONS: The present study demonstrated that ethanol intake altered the expression of BA-related synthetases and transporters. The BA levels, especially the toxic BAs (chenodeoxycholic acid, deoxycholic acid and lithocholic acid), in the whole EHC were significantly increased by ethanol intake, which may provide a potential explanation to illuminate the pathogenesis of alcoholic liver injury. Most importantly, chronic ethanol consumption had a significant impact on the pharmacokinetics (AUC0-24 h and clearance) of irinotecan and SN38; hence colon cancer patients with chronic alcohol consumption treated with irinotecan deserve our close attention.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Bile Acids and Salts/metabolism , Enterohepatic Circulation/drug effects , Irinotecan/pharmacokinetics , Alcohol Drinking , Animals , Bile Acids and Salts/blood , Blotting, Western , Colon/drug effects , Colon/pathology , Disease Models, Animal , Gallbladder/drug effects , Gallbladder/pathology , Irinotecan/pharmacology , Liver/drug effects , Liver/pathology , Male , Mice
8.
Am J Emerg Med ; 37(9): 1809.e1-1809.e3, 2019 09.
Article in English | MEDLINE | ID: mdl-31257124

ABSTRACT

Half-life of the antipsychotic vegetamin is very long, partially due to the presence of phenobarbital, and mortality due to phenobarbital poisoning is high. Here, we present the case of a 22-year-old female admitted to the emergency department with disturbed consciousness due to vegetamin overdose. Her blood phenobarbital level was elevated to 123 µg/ml. Phenobarbital undergoes enterohepatic circulation, and its retention in the intestine causes its blood levels to remain sustained. The utility of hemodialysis for drug poisoning has been previously reported; however, its efficiency is not yet established and its efficacy is low for drugs with long half-lives such as phenobarbital. Therefore, we performed a two-tube approach to adsorb phenobarbital in the intestines with activated charcoal delivered via a gastric tube and to remove the phenobarbital-adsorbed activated charcoal using whole bowel irrigation via an ileus tube 2 h later. The patient successfully eliminated the charcoal via stool, the blood phenobarbital level decreased drastically without hemodialysis, and the clinical course improved. We propose that this two-tube approach is suitable for treatment of poisoning with drugs that undergo enterohepatic circulation and have long half-lives.


Subject(s)
Charcoal/therapeutic use , Drug Overdose/therapy , Hypnotics and Sedatives/poisoning , Phenobarbital/poisoning , Enterohepatic Circulation/drug effects , Female , Hemoperfusion/methods , Humans , Hypnotics and Sedatives/pharmacokinetics , Phenobarbital/pharmacokinetics , Young Adult
9.
Biomed Pharmacother ; 116: 109044, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31174088

ABSTRACT

BACKGROUND: Sanwei Ganjiang Powder (SWGJ), derived from traditional Chinese medicine (TCM), has long demonstrated its effectiveness in long-term liver damage therapy. Recent studies indicated that it can also regulate the intestinal tract, although the underlying molecular mechanisms of this remain mysterious. The aim of the study is to investigate the mechanisms of SWGJ against dysbacteriosis and carbon tetrachloride (CCl4)-induced gut-liver axis damage underlying bile acid enterohepatic circulation. METHODS: To observe the regulatory effects of SWGJ on Liver and Intestinal Damage, we explored two animal models. In model 1, sixty BALB/c mice were subjected to oral gavage with 12 g/kg of ceftriaxone sodium for 10d; during this time, SWGJ, bifendate and bifico were sequentially administered over 7d. In model 2, the model of chronic liver injury was induced by subcutaneous injection of 40% CCl4 oil solution twice per week for 8 weeks. From the 3rd week, SWGJ, bifendate and bifico were sequentially administered for 6 weeks. Intestinal flora (16S rDNA analysis), histology (H&E staining), tight connections (Immunohistochemistry, IHC), ultrastructure (Transmission electron microscopy, TEM), inflammatory cytokines and LPS (Enzyme-linked immunosorbent assay, ELISA) of the intestines were assessed, and liver function was also evaluated by methods including ALT, AST and H&E staining. The levels of protein associated with bile acid metabolism were assessed by western blot. RESULTS: In model 1, SWGJ significantly decreased the activity of inflammatory cytokines and LPS compared with the ceftriaxone sodium group. In addition, SWGJ improved symptoms of intestinal flora imbalance; further, ZO-1 and occludin in the cytoplasm of intestinal villus epithelial cells was increased, and the histopathology of the ileum was improved. Notably, the expression of ALT and AST was significant increased, and disordered hepatic lobule structures were clearly observed in liver histopathology in model group; SWGJ can significantly improve these changes. Furthermore, the levels of proteins related to bile acid synthesis, such as CYP7A1, were significantly upregulated in the SWGJ group compared with the model, and proteins related to excretion and reabsorption, such as NTCP, Mrp2 and BESP, were also upregulated. Importantly, SWGJ increased the nuclear expression of nuclear factor-E2-related factor-2 (Nrf2). Similar results appeared in model 2. CONCLUSION: This study suggests that SWGJ may elicit significant effects on the treatment of gut-liver axis damage, potential mechanisms at least partially involve bile acid enterohepatic, and increasing of the nuclear Nrf2 levels.


Subject(s)
Bile Acids and Salts/metabolism , Drugs, Chinese Herbal/pharmacology , Homeostasis , Intestines/pathology , Liver/pathology , Animals , Body Weight/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cytokines/metabolism , Drugs, Chinese Herbal/chemistry , Enterohepatic Circulation/drug effects , Female , Gastrointestinal Microbiome/drug effects , Homeostasis/drug effects , Inflammation Mediators/metabolism , Intestinal Mucosa/drug effects , Intestines/drug effects , Intestines/microbiology , Lipopolysaccharides , Liver/drug effects , Liver/physiopathology , Male , Mice, Inbred BALB C , NF-E2-Related Factor 2/metabolism , Organ Size/drug effects , Rats
10.
J Clin Pharmacol ; 59(10): 1351-1365, 2019 10.
Article in English | MEDLINE | ID: mdl-31062373

ABSTRACT

Tacrolimus or cyclosporine is prescribed with mycophenolic acid posttransplant and contributes to interpatient variability in mycophenolic acid pharmacokinetics and response. Cyclosporine inhibits enterohepatic circulation of the metabolite mycophenolic acid glucuronide, which is not described with tacrolimus. This study investigated mycophenolic acid pharmacokinetics and adverse effects in stable renal transplant recipients and the association with calcineurin inhibitors, sex, and race. Mycophenolic acid and mycophenolic acid glucuronide area under the concentration-time curve from 0 to 12 hours (AUC0-12h ) and apparent clearance were determined at steady state in 80 patients receiving cyclosporine with mycophenolate mofetil and 67 patients receiving tacrolimus with mycophenolate sodium. Gastrointestinal adverse effects and hematologic parameters were evaluated. Statistical models evaluated mycophenolic acid pharmacokinetics and adverse effects. Mycophenolic acid AUC0-12h was 1.70-fold greater with tacrolimus (68.9 ± 30.9 mg·h/L) relative to cyclosporine (40.8 ± 17.6 mg·h/L); P < .001. Target mycophenolic acid AUC0-12h of 30-60 mg·h/L was achieved in 56.3% on cyclosporine compared with 34.3% receiving tacrolimus (P < .001). Mycophenolic acid clearance was 48% slower with tacrolimus (10.6 ± 4.7 L/h) relative to cyclosporine (20.5 ± 10.0 L/h); P < .001. Enterohepatic circulation occurred less frequently with cyclosporine (45%) compared with tacrolimus (78%); P < 0.001; with a 2.9-fold greater mycophenolic acid glucuronide AUC0-12h to mycophenolic acid AUC0-12h ratio (P < .001). Race did not affect mycophenolic acid pharmacokinetics. Gastrointestinal adverse effect scores were 2.2-fold higher with tacrolimus (P < .001) and more prominent in women (P = .017). Lymphopenia was more prevalent with tacrolimus (52.2%) than cyclosporine (22.5%); P < 0.001. Calcineurin inhibitors and sex contributed to interpatient variability in mycophenolic acid pharmacokinetics and adverse effects post-renal transplant, which could be attributed to differences in enterohepatic circulation.


Subject(s)
Antibiotics, Antineoplastic/pharmacokinetics , Calcineurin Inhibitors/adverse effects , Drug Interactions/physiology , Mycophenolic Acid/pharmacokinetics , Area Under Curve , Cyclosporine/adverse effects , Enterohepatic Circulation/drug effects , Female , Humans , Immunosuppressive Agents/adverse effects , Kidney/drug effects , Kidney Transplantation/methods , Lymphopenia/drug therapy , Male , Middle Aged , Tacrolimus/adverse effects , Transplant Recipients
11.
J Agric Food Chem ; 67(14): 3956-3965, 2019 Apr 10.
Article in English | MEDLINE | ID: mdl-30905157

ABSTRACT

The heavy metal cadmium (Cd) is a contaminant widely distributed in the food chain. In the present study, 8-week oral administration of a probiotic strain, Lactobacillus plantarum CCFM8610, markedly decreased blood Cd levels in volunteers. Further animal study showed that three L. plantarum strains administered orally exhibited significantly different effects on the regulation of bile acid (BA) metabolism and Cd excretion in mice. Among the strains, L. plantarum CCFM8610 showed the most significant effects on enhancing hepatic BA synthesis, biliary glutathione output, and fecal BA excretion. Biliary Cd output and fecal Cd excretion were markedly increased after L. plantarum CCFM8610 administration, resulting in a marked reduction in tissue Cd levels. The regulation of BA homeostasis and Cd excretion was due to the suppression of the enterohepatic farnesoid X receptor-fibroblast growth factor 15 (FXR-FGF15) axis by L. plantarum CCFM8610 and could be abolished by treatment with the FXR agonist GW4064. The regulatory effects were also related to the gut microbiota, as antibiotic pretreatment reversed L. plantarum CCFM8610-induced effects in BA and Cd metabolism.


Subject(s)
Cadmium/metabolism , Enterohepatic Circulation/drug effects , Lactobacillus plantarum/physiology , Probiotics/administration & dosage , Adult , Animals , Bile Acids and Salts/metabolism , Cadmium/blood , Feces/chemistry , Female , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Gastrointestinal Microbiome/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
12.
Drugs R D ; 19(1): 67-71, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30689138

ABSTRACT

A recent multi-state outbreak of life-threatening bleeding following inhalation of synthetic cannabinoids has been attributed to contamination with the long-acting anticoagulant rodenticide (LAAR) brodifacoum, a second-generation, highly potent, long-acting derivative of the commonly used blood thinner warfarin. While long-term treatment with high-dose vitamin K1 restores coagulation, it does not affect brodifacoum metabolism or clearance, and, consequently, brodifacoum remains in the human body for several months, thereby predisposing to risk of bleeding recurrence and development of coagulation-independent injury in extrahepatic tissues and fetuses. This has prompted the evaluation of pharmacological measures that accelerate brodifacoum clearance from poisoned patients. Since the induction of certain cytochrome P450 (CYP) enzymes accelerates warfarin metabolism, using CYP inducers, such as phenobarbital, to accelerate brodifacoum clearance seems plausible. However, unlike warfarin, brodifacoum does not undergo significant metabolism in the liver, nor have the effects of phenobarbital on vitamin K1 metabolism been previously determined. In addition, the safety of phenobarbital in brodifacoum-poisoned patients has not been established. Therefore, we propose that CYP inducers should not be used to accelerate the clearance of brodifacoum from poisoned patients, but that alternative approaches such as reducing enterohepatic recirculation of brodifacoum, or using lipid emulsions to scavenge brodifacoum throughout the body, be considered.


Subject(s)
4-Hydroxycoumarins/pharmacokinetics , 4-Hydroxycoumarins/poisoning , Cytochrome P-450 Enzyme Inducers/administration & dosage , Anticoagulants/administration & dosage , Blood Coagulation/drug effects , Enterohepatic Circulation/drug effects , Fat Emulsions, Intravenous , Half-Life , Hemorrhage/chemically induced , Hemorrhage/drug therapy , Hemorrhage/metabolism , Humans , Inactivation, Metabolic/drug effects , Vitamin K/administration & dosage
13.
J Ethnopharmacol ; 235: 472-480, 2019 May 10.
Article in English | MEDLINE | ID: mdl-30528660

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Polygonum multiflorum Thunb (Heshouwu, HSW) is commonly used in clinical medicine, while the hepatotoxicities of HSW are reported increasingly in recent years. Currently, researchers have demonstrated an essential role of Bile Acids (BAs) in liver diseases. The occurrence of hepatotoxicity cases linked to HSW are characterized by jaundice and cholestasis, suggesting an interaction that between BAs and HSW AIM OF THE STUDY: This study was designed to investigate the HSW-induced liver functional and histological changes in mice and the role of HSW on bile acid synthesis, metabolism, clearance and intestinal absorption. MATERIALS AND METHODS: The mice were intragastrically (i.g.) given HSW at doses of 1.275 and 3.825 g/kg (Crude extracts /body weight) once a day for seven days. Liver function was evaluated by measuring the serum levels of enzymes and analyzing the liver histology. The LC/MS analysis was performed to quantify BAs from liver, ileum and serum. Moreover, the expression of bile metabolic-related transporters and metabolic enzymes at both protein and mRNA levels were observed to elucidate the underlying mechanisms. RESULTS: Oral administration of HSW for 7 days could not cause liver damage. A significant change was observed for the concentrations of liver and serum BAs in treatment groups compared with normal control. The mRNA expression levels of bile acid excretory transporter (Bsep) and basolateral uptake transporter (Ntcp) were increased with the development of HSW. The concentrations of unconjugated BAs increased in mice intestines after the administration of HSW. Western blot and qRT-PCR analyses showed that HSW upregulated the protein and mRNA expression of Shp and Fgf15 in the ileum of the mice. CONCLUSION: HSW treatment for 7days did not cause liver damage. HSW accelerated bile acid enterohepatic circulation and changed the composition of intestinal BAs, leding to the activation of Fxr-Fgf15 signal in intestines, and further inhibited the expression of Cyp7a1 in the liver.


Subject(s)
Bile Acids and Salts/metabolism , Fallopia multiflora/chemistry , Intestines/drug effects , Plant Extracts/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 11/genetics , Animals , Bile Acids and Salts/biosynthesis , Blotting, Western , Dose-Response Relationship, Drug , Enterohepatic Circulation/drug effects , Fibroblast Growth Factors/metabolism , Gene Expression Regulation/drug effects , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Plant Extracts/administration & dosage , RNA, Messenger/drug effects , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction/drug effects
14.
Xenobiotica ; 49(4): 446-456, 2019 Apr.
Article in English | MEDLINE | ID: mdl-29629619

ABSTRACT

Ezetimibe is a potent cholesterol absorption inhibitor, with an erratic pharmacokinetic (PK) profile, attributed to an extensive enterohepatic recirculation (EHC). The aim of this study was to develop a population PK model able to adequately characterize the complex distribution processes of total ezetimibe. The analysis was performed on the individual concentration-time data obtained from 28 healthy subjects who participated in a bioequivalence study comparing two oral ezetimibe formulations. The population PK analysis was performed using nonlinear mixed effect modeling, where different EHC models were developed and evaluated for their performance. Total ezetimibe pharmacokinetics was best described by a four-compartment model featuring EHC through the inclusion of an additional gallbladder compartment, which was assumed to release drug at specific time-intervals consistent with food intake. The final PK model was able to adequately estimate the population pharmacokinetic parameters and to allow for a formal characterization of the pharmacokinetic profile and the secondary peaks due to enterohepatic recirculation.


Subject(s)
Enterohepatic Circulation , Ezetimibe/pharmacokinetics , Administration, Oral , Adult , Dose-Response Relationship, Drug , Enterohepatic Circulation/drug effects , Ezetimibe/administration & dosage , Ezetimibe/pharmacology , Female , Humans , Male
15.
J Cyst Fibros ; 18(2): 286-293, 2019 03.
Article in English | MEDLINE | ID: mdl-30279125

ABSTRACT

OBJECTIVE: Disruption of the enterohepatic circulation of bile acids (BAs) is part of the gastrointestinal phenotype of cystic fibrosis (CF). Ivacaftor (VX-770), a cystic fibrosis transmembrane conductance regulator (CFTR) potentiator, improves pulmonary function in CF patients with class III gating mutations. We studied the effect of ivacaftor on the enterohepatic circulation by assessing markers of BA homeostasis and their changes in CF patients. METHODS: In CF patients with an S1251N mutation (N = 16; age 9-35 years S125N study/NTR4873) or a G551D mutation (N = 101; age 10-24 years; GOAL study/ NCT01521338) we analyzed plasma fibroblast growth factor 19 (FGF19) and 7α-hydroxy-4-cholesten-3-one (C4) levels, surrogate markers for intestinal BA absorption and hepatic synthesis, respectively, before and after treatment with ivacaftor. RESULTS: At baseline, median FGF19 was lower (52% and 53%, P < .001) and median C4 higher (350% and 364%, P < .001), respectively, for the S1251 N and G551D mutation patient groups compared to healthy controls. Treatment with ivacaftor significantly increased FGF19 and reduced C4 levels towards normalization in both cohorts but this did not correlate with CFTR function in other organs, as measured by sweat chloride levels or pulmonary function. CONCLUSIONS: We demonstrate that patients with CFTR gating mutations display interruption of the enterohepatic circulation of BAs reflected by lower FGF19 and elevated C4 levels. Treatment with ivacaftor partially restored this disruption of BA homeostasis. The improvement did not correlate with established outcome measures of CF, suggesting involvement of modulating factors of CFTR correction in different organs.


Subject(s)
Bile Acids and Salts , Cholestenones/blood , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis , Enterohepatic Circulation/drug effects , Fibroblast Growth Factors/blood , Adolescent , Adult , Aminophenols/pharmacokinetics , Aminophenols/therapeutic use , Bile Acids and Salts/biosynthesis , Bile Acids and Salts/metabolism , Biological Availability , Child , Chloride Channel Agonists/pharmacokinetics , Chloride Channel Agonists/therapeutic use , Cystic Fibrosis/drug therapy , Cystic Fibrosis/genetics , Cystic Fibrosis/metabolism , Cystic Fibrosis/physiopathology , Female , Homeostasis/drug effects , Humans , Male , Mutation , Netherlands , Quinolones/pharmacokinetics , Quinolones/therapeutic use
16.
Diabetes ; 67(9): 1720-1728, 2018 09.
Article in English | MEDLINE | ID: mdl-30135133

ABSTRACT

Bariatric surgery procedures, such as Roux-en-Y gastric bypass (RYGB) and vertical sleeve gastrectomy (VSG), are the most effective interventions available for sustained weight loss and improved glucose metabolism. Bariatric surgery alters the enterohepatic bile acid circulation, resulting in increased plasma bile levels as well as altered bile acid composition. While it remains unclear why both VSG and RYGB can alter bile acids, it is possible that these changes are important mediators of the effects of surgery. Moreover, a molecular target of bile acid synthesis, the bile acid-activated transcription factor FXR, is essential for the positive effects of VSG on weight loss and glycemic control. This Perspective examines the relationship and sequence of events between altered bile acid levels and composition, FXR signaling, and gut microbiota after bariatric surgery. We hypothesize that although bile acids and FXR signaling are potent mediators of metabolic function, unidentified downstream targets are the main mediators behind the benefits of weight-loss surgery. One of these targets, the gut-derived peptide FGF15/19, is a potential molecular and therapeutic marker to explain the positive metabolic effects of bariatric surgery. Focusing research efforts on identifying these complex molecular mechanisms will provide new opportunities for therapeutic strategies to treat obesity and metabolic dysfunction.


Subject(s)
Fibroblast Growth Factors/antagonists & inhibitors , Hypoglycemic Agents/therapeutic use , Metabolic Diseases/drug therapy , Models, Biological , Obesity, Morbid/physiopathology , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Signal Transduction/drug effects , Animals , Bariatric Surgery/adverse effects , Bile Acids and Salts/metabolism , Combined Modality Therapy/adverse effects , Enterohepatic Circulation/drug effects , Fibroblast Growth Factors/agonists , Fibroblast Growth Factors/metabolism , Gastrointestinal Microbiome , Humans , Liver/drug effects , Liver/metabolism , Metabolic Diseases/etiology , Metabolic Diseases/metabolism , Metabolic Diseases/microbiology , Molecular Targeted Therapy/adverse effects , Obesity, Morbid/metabolism , Obesity, Morbid/surgery , Obesity, Morbid/therapy , Organ Specificity , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/metabolism
17.
J Clin Pharmacol ; 58(11): 1452-1460, 2018 11.
Article in English | MEDLINE | ID: mdl-29873817

ABSTRACT

AZD3241 is a potent and selective myeloperoxidase inhibitor potentially for the treatment of a number of neurodegenerative disorders, including multiple system atrophy (MSA). The objectives of this work were to develop a population pharmacokinetic (PopPK) model for AZD3241 and to investigate the correlation between AZD3241 exposure and myeloperoxidase inhibition. The PopPK model was developed using AZD3241 data from one phase 1 study in healthy subjects and one phase 2 study in patients with MSA. A one-compartment model incorporating a gallbladder compartment for enterohepatic circulation, sequential zero-first order absorption, and first-order elimination adequately described the AZD3241 concentration profiles. The apparent clearance and central volume of distribution were 63.1 L/h (interindividual variability: 34.8%) and 121.9 L (interindividual variability: 44.0%), respectively. The enterohepatic circulation model reasonably captured the second peak of AZD3241, and high-fat food increased the absorption rate by 69%. A linear regression model was applied to describe the relationship between AZD3241 exposure and percentage change from baseline in myeloperoxidase-specific activity. The developed PopPK model was consistent with known pharmacokinetic characteristics of AZD3241. This model can be used to estimate AZD3241 exposure in patients with MSA and could be applied to future pharmacokinetic-pharmacodynamic analyses of AZD3241 in clinical development.


Subject(s)
Enterohepatic Circulation/drug effects , Multiple System Atrophy/drug therapy , Peroxidase/antagonists & inhibitors , Pyrimidinones/pharmacology , Pyrimidinones/pharmacokinetics , Pyrroles/pharmacology , Pyrroles/pharmacokinetics , Adult , Aged , Bile , Female , Healthy Volunteers , Humans , Male , Middle Aged , Models, Biological , Multiple System Atrophy/metabolism , Pyrimidinones/metabolism , Pyrroles/metabolism , Random Allocation
18.
Drug Deliv ; 25(1): 1224-1233, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29791242

ABSTRACT

Liver is the primary acting site of insulin. In this study, we developed innovative nanoparticles for oral and liver-targeted delivery of insulin by using enterohepatic circulation of bile acids. The nanoparticles were produced from cholic acid and quaternary ammonium modified chitosan derivative and hydroxypropyl methylcellulose phthalate (HPMCP). The nanoparticles had a diameter of 239 nm, an insulin loading efficiency of 90.9%, and a loading capacity of 18.2%. Cell culture studies revealed that the cholic acid groups effectively enhanced the transport of the nanoparticles through Caco-2 cell monolayer and greatly increased the absorption of the nanoparticles in HepG-2 cells via bile acid transporter mechanism. Ex vivo fluorescence images of ileum section, gastrointestinal tract, and liver demonstrated that the HPMCP increased the mucoadhesion of the nanoparticles in ileum, and the cholic acid groups facilitated the absorptions of the nanoparticles in both ileum and liver by use of bile acid transporters via enterohepatic circulation of bile acids. The therapy for diabetic mice displayed that the oral nanoparticle group could maintain hypoglycemic effect for more than 24 h and its pharmacological availability was about 30% compared with the insulin injection group. For the first time, this study demonstrates that using enterohepatic circulation of bile acids is an effective strategy for oral delivery of insulin.


Subject(s)
Bile Acids and Salts/metabolism , Enterohepatic Circulation/drug effects , Insulin/pharmacology , Liver/drug effects , Nanoparticles/chemistry , Animals , Blood Glucose/drug effects , Caco-2 Cells , Cell Line, Tumor , Chitosan/chemistry , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Drug Carriers/chemistry , Drug Delivery Systems/methods , Female , Hep G2 Cells , Humans , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , Insulin/chemistry , Liver/metabolism , Male , Methylcellulose/analogs & derivatives , Methylcellulose/chemistry , Mice, Inbred ICR
19.
Sci Rep ; 7(1): 5921, 2017 07 19.
Article in English | MEDLINE | ID: mdl-28725001

ABSTRACT

2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is a persistent environmental contaminant which elicits hepatotoxicity through activation of the aryl hydrocarbon receptor (AhR). Male C57BL/6 mice orally gavaged with TCDD (0.01-30 µg/kg) every 4 days for 28 days exhibited bile duct proliferation and pericholangitis. Mass spectrometry analysis detected a 4.6-fold increase in total hepatic bile acid levels, despite the coordinated repression of genes involved in cholesterol and primary bile acid biosynthesis including Cyp7a1. Specifically, TCDD elicited a >200-fold increase in taurolithocholic acid (TLCA), a potent G protein-coupled bile acid receptor 1 (GPBAR1) agonist associated with bile duct proliferation. Increased levels of microbial bile acid metabolism loci (bsh, baiCD) are consistent with accumulation of TLCA and other secondary bile acids. Fecal bile acids decreased 2.8-fold, suggesting enhanced intestinal reabsorption due to induction of ileal transporters (Slc10a2, Slc51a) and increases in whole gut transit time and intestinal permeability. Moreover, serum bile acids were increased 45.4-fold, consistent with blood-to-hepatocyte transporter repression (Slco1a1, Slc10a1, Slco2b1, Slco1b2, Slco1a4) and hepatocyte-to-blood transporter induction (Abcc4, Abcc3). These results suggest that systemic alterations in enterohepatic circulation, as well as host and microbiota bile acid metabolism, favor bile acid accumulation that contributes to AhR-mediated hepatotoxicity.


Subject(s)
Bacteria/metabolism , Bile Acids and Salts/metabolism , Enterohepatic Circulation/drug effects , Homeostasis/drug effects , Polychlorinated Dibenzodioxins/toxicity , Animals , Bacteria/drug effects , Bacteria/genetics , Bile Acids and Salts/blood , Cholesterol/metabolism , Feces/chemistry , Female , Genes, Bacterial , Hydrophobic and Hydrophilic Interactions , Intestinal Absorption/drug effects , Liver/drug effects , Liver/metabolism , Male , Mice, Inbred C57BL
20.
Drug Des Devel Ther ; 10: 2173-80, 2016.
Article in English | MEDLINE | ID: mdl-27462142

ABSTRACT

According to previous research studies, warfarin can be detected in human bile after oral administration. Ferulic acid (FA) is the main bioactive component of many Chinese herbs for the treatment of cardiovascular disease. To elucidate the effects of FA on the pharmacokinetics of warfarin in rats after biliary drainage is necessary. Twenty rats were randomly divided into four groups: Group 1 (WN): healthy rats after the administration of warfarin sodium, Group 2 (WO): a rat model of biliary drainage after the administration of warfarin sodium, Group 3 (WFN): healthy rats after the administration of warfarin sodium and FA, and Group 4 (WFO): a rat model of biliary drainage after the administration of warfarin sodium and FA. Blood samples were collected at different time points after administration. The concentrations of blood samples were determined by ultraperformance liquid chromatography-tandem mass spectrometry. Comparisons between groups were performed according to the main pharmacokinetic parameters calculated by the DAS 2.1.1 software. The pharmacokinetic parameters showed a significant difference between the WN and WO groups, the WO group showed a decrease of 51% and 41.6% in area under the curve from 0 to time (AUC0- t ) and peak plasma concentration (C max), respectively, whereas time to C max (T max) was delayed 3.27 folds. There were significant differences between the WFO and WFN groups, the WFO group showed a decrease of 63.8% and 70% in AUC0- t and C max, respectively; the delay in T max between the WN and WFN groups (mean, from 132-432 minutes) was significantly different; the mean retention time from 0 to time (MRT0- t ) between the WO and WFO groups (mean, from 718.31-606.13 minutes) also showed a significant difference. Enterohepatic circulation markedly influences the disposition of warfarin in rats, and FA significantly affected the warfarin disposition in rat plasma.


Subject(s)
Coumaric Acids/chemistry , Drugs, Chinese Herbal/analysis , Enterohepatic Circulation/drug effects , Tandem Mass Spectrometry/methods , Warfarin/analysis , Warfarin/pharmacokinetics , Administration, Oral , Animals , Coumaric Acids/pharmacology , Drainage , Drugs, Chinese Herbal/administration & dosage , Drugs, Chinese Herbal/chemistry , Humans , Rats , Warfarin/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL