Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 123
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Appl Toxicol ; 44(6): 863-873, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38311468

RESUMEN

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.


Asunto(s)
Ácidos y Sales Biliares , Colchicina , Circulación Enterohepática , Homeostasis , Hígado , Animales , Ácidos y Sales Biliares/metabolismo , Circulación Enterohepática/efectos de los fármacos , Colchicina/toxicidad , Homeostasis/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones , Masculino , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores Citoplasmáticos y Nucleares/efectos de los fármacos , Receptores Citoplasmáticos y Nucleares/genética , Íleon/efectos de los fármacos , Íleon/metabolismo , Diarrea/inducido químicamente
2.
Hepatology ; 73(6): 2577-2585, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33222321

RESUMEN

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.


Asunto(s)
Miembro 11 de la Subfamilia B de Transportador de Casetes de Unión al ATP , Ácidos y Sales Biliares/metabolismo , Circulación Enterohepática/fisiología , Proteínas de Transporte de Membrana , Transportadores de Anión Orgánico Sodio-Dependiente , Receptores Acoplados a Proteínas G , Simportadores , Miembro 11 de la Subfamilia B de Transportador de Casetes de Unión al ATP/genética , Miembro 11 de la Subfamilia B de Transportador de Casetes de Unión al ATP/metabolismo , Animales , Transporte Biológico Activo/efectos de los fármacos , Transporte Biológico Activo/fisiología , Desarrollo de Medicamentos , Circulación Enterohepática/efectos de los fármacos , Humanos , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Transportadores de Anión Orgánico Sodio-Dependiente/antagonistas & inhibidores , Transportadores de Anión Orgánico Sodio-Dependiente/genética , Transportadores de Anión Orgánico Sodio-Dependiente/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Simportadores/antagonistas & inhibidores , Simportadores/genética , Simportadores/metabolismo
3.
Dig Dis Sci ; 66(2): 568-576, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32198567

RESUMEN

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.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Ácido Desoxicólico/toxicidad , Dieta Occidental/efectos adversos , Disbiosis/metabolismo , Circulación Enterohepática/fisiología , Enfermedades Inflamatorias del Intestino/metabolismo , Animales , Ácido Desoxicólico/administración & dosificación , Disbiosis/inducido químicamente , Disbiosis/patología , Circulación Enterohepática/efectos de los fármacos , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/fisiología , Enfermedades Inflamatorias del Intestino/inducido químicamente , Enfermedades Inflamatorias del Intestino/patología , Ratones , Ratones Endogámicos C57BL
4.
Alcohol Alcohol ; 55(3): 264-277, 2020 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-32232424

RESUMEN

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.


Asunto(s)
Antineoplásicos/farmacocinética , Ácidos y Sales Biliares/metabolismo , Circulación Enterohepática/efectos de los fármacos , Irinotecán/farmacocinética , Consumo de Bebidas Alcohólicas , Animales , Ácidos y Sales Biliares/sangre , Western Blotting , Colon/efectos de los fármacos , Colon/patología , Modelos Animales de Enfermedad , Vesícula Biliar/efectos de los fármacos , Vesícula Biliar/patología , Irinotecán/farmacología , Hígado/efectos de los fármacos , Hígado/patología , Masculino , Ratones
5.
Xenobiotica ; 49(4): 446-456, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29629619

RESUMEN

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.


Asunto(s)
Circulación Enterohepática , Ezetimiba/farmacocinética , Administración Oral , Adulto , Relación Dosis-Respuesta a Droga , Circulación Enterohepática/efectos de los fármacos , Ezetimiba/administración & dosificación , Ezetimiba/farmacología , Femenino , Humanos , Masculino
6.
Am J Emerg Med ; 37(9): 1809.e1-1809.e3, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31257124

RESUMEN

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.


Asunto(s)
Carbón Orgánico/uso terapéutico , Sobredosis de Droga/terapia , Hipnóticos y Sedantes/envenenamiento , Fenobarbital/envenenamiento , Circulación Enterohepática/efectos de los fármacos , Femenino , Hemoperfusión/métodos , Humanos , Hipnóticos y Sedantes/farmacocinética , Fenobarbital/farmacocinética , Adulto Joven
7.
Acta Pharmacol Sin ; 37(7): 1002-12, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27180979

RESUMEN

AIM: Diclofenac is a non-steroidal anti-inflammatory drug (NSAID), which may cause serious intestinal adverse reactions (enteropathy). In this study we investigated whether co-administration of ciprofloxacin affected the pharmacokinetics of diclofenac and diclofenac-induced enteropathy in rats. METHODS: The pharmacokinetics of diclofenac was assessed in rats after receiving diclofenac (10 mg/kg, ig, or 5 mg/kg, iv), with or without ciprofloxacin (20 mg/kg, ig) co-administered. After receiving 6 oral doses or 15 intravenous doses of diclofenac, the rats were sacrificed, and small intestine was removed to examine diclofenac-induced enteropathy. ß-Glucuronidase activity in intestinal content, bovine liver and E coli was evaluated. RESULTS: Following oral or intravenous administration, the pharmacokinetic profile of diclofenac displayed typical enterohepatic circulation, and co-administration of ciprofloxacin abolished the enterohepatic circulation, resulted in significant reduction in the plasma content of diclofenac. In control rats, ß-glucuronidase activity in small intestinal content was region-dependent: proximal intestine

Asunto(s)
Antiinflamatorios no Esteroideos/efectos adversos , Ciprofloxacina/farmacología , Diclofenaco/efectos adversos , Diclofenaco/farmacocinética , Circulación Enterohepática/efectos de los fármacos , Glucuronidasa/antagonistas & inhibidores , Enfermedades Intestinales/prevención & control , Intestino Delgado/enzimología , Animales , Bovinos , Diclofenaco/sangre , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Escherichia coli/metabolismo , Enfermedades Intestinales/inducido químicamente , Intestino Delgado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratas
8.
J Lipid Res ; 55(12): 2576-86, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25278499

RESUMEN

Statins are effective cholesterol-lowering drugs to treat CVDs. Bile acids (BAs), the end products of cholesterol metabolism in the liver, are important nutrient and energy regulators. The present study aims to investigate how statins affect BA homeostasis in the enterohepatic circulation. Male C57BL/6 mice were treated with atorvastatin (100 mg/kg/day po) for 1 week, followed by BA profiling by ultra-performance LC-MS/MS. Atorvastatin decreased BA pool size, mainly due to less BA in the intestine. Surprisingly, atorvastatin did not alter total BAs in the serum or liver. Atorvastatin increased the ratio of 12α-OH/non12α-OH BAs. Atorvastatin increased the mRNAs of the BA-synthetic enzymes cholesterol 7α-hydroxylase (Cyp7a1) (over 10-fold) and cytochrome P450 27a1, the BA uptake transporters Na⁺/taurocholate cotransporting polypeptide and organic anion transporting polypeptide 1b2, and the efflux transporter multidrug resistance-associated protein 2 in the liver. Noticeably, atorvastatin suppressed the expression of BA nuclear receptor farnesoid X receptor (FXR) target genes, namely small heterodimer partner (liver) and fibroblast growth factor 15 (ileum). Furthermore, atorvastatin increased the mRNAs of the organic cation uptake transporter 1 and cholesterol efflux transporters Abcg5 and Abcg8 in the liver. The increased expression of BA-synthetic enzymes and BA transporters appear to be a compensatory response to maintain BA homeostasis after atorvastatin treatment. The Cyp7a1 induction by atorvastatin appears to be due to suppressed FXR signaling in both the liver and intestine.


Asunto(s)
Colesterol 7-alfa-Hidroxilasa/metabolismo , Inductores de las Enzimas del Citocromo P-450/farmacología , Ácidos Heptanoicos/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Mucosa Intestinal/efectos de los fármacos , Hígado/efectos de los fármacos , Pirroles/farmacología , Proteínas de Unión al ARN/antagonistas & inhibidores , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 5 , Transportador de Casete de Unión a ATP, Subfamilia G, Miembro 8 , Transportadoras de Casetes de Unión a ATP/agonistas , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Atorvastatina , Ácidos y Sales Biliares/sangre , Ácidos y Sales Biliares/metabolismo , Colestanotriol 26-Monooxigenasa/química , Colestanotriol 26-Monooxigenasa/genética , Colestanotriol 26-Monooxigenasa/metabolismo , Colesterol 7-alfa-Hidroxilasa/química , Colesterol 7-alfa-Hidroxilasa/genética , Circulación Enterohepática/efectos de los fármacos , Humanos , Íleon/efectos de los fármacos , Íleon/enzimología , Íleon/metabolismo , Mucosa Intestinal/enzimología , Mucosa Intestinal/metabolismo , Lipoproteínas/agonistas , Lipoproteínas/genética , Lipoproteínas/metabolismo , Hígado/enzimología , Hígado/metabolismo , Transportador 1 de Anión Orgánico Específico del Hígado , Masculino , Ratones Endogámicos C57BL , Transportadores de Anión Orgánico Sodio-Dependiente/agonistas , Transportadores de Anión Orgánico Sodio-Dependiente/química , Transportadores de Anión Orgánico Sodio-Dependiente/genética , Transportadores de Anión Orgánico Sodio-Independiente/agonistas , Transportadores de Anión Orgánico Sodio-Independiente/genética , Transportadores de Anión Orgánico Sodio-Independiente/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Transducción de Señal/efectos de los fármacos , Simportadores/agonistas , Simportadores/química , Simportadores/genética
9.
Biopharm Drug Dispos ; 35(5): 275-83, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24615849

RESUMEN

The interaction between mycophenolate (MPA) and quinolone antibiotics such as ciprofloxacin is considered to reduce the enterohepatic recycling of MPA, which is biotransformed in the intestine from MPA glucuronide (MPAG) conjugate excreted via the biliary system; however, the molecular mechanism underlying this biotransformation of MPA is still unclear. In this study, an in vitro system was established to evaluate ß-glucuronidase-mediated deconjugation and to examine the influence of ciprofloxacin on the enzymatic deconjugation of MPAG and MPA resynthesis. Resynthesis of MPA via deconjugation of MPAG increased in a time-dependent manner from 5 to 60 min in the presence of ß-glucuronidase. Ciprofloxacin and phenolphthalein-ß-d-glucuronide (PhePG), a typical ß-glucuronidase substrate, significantly decreased the production of MPA from MPAG in the ß-glucuronidase-mediated deconjugation system. In addition, enoxacin significantly inhibited the production of MPA from MPAG, while levofloxacin and ofloxacin had no inhibitory effect on MPA synthesis. Pharmacokinetic analysis revealed that ciprofloxacin showed a dose-dependent inhibitory effect on MPA production from MPAG via ß-glucuronidase with a half-maximal inhibitory concentration (IC50 ) value of 30.4 µm. While PhePG inhibited the ß-glucuronidase-mediated production of MPA from MPAG in a competitive manner, ciprofloxacin inhibited MPA synthesis via noncompetitive inhibition. These findings suggest that the reduction in the serum MPA concentration during the co-administration of ciprofloxacin is at least in part due to the decreased enterohepatic circulation of MPA because of noncompetitive inhibition of deconjugation of MPAG by intestinal ß-glucuronidase.


Asunto(s)
Ciprofloxacina/farmacología , Glucuronidasa/metabolismo , Glucurónidos/farmacocinética , Ácido Micofenólico/análogos & derivados , Ácido Micofenólico/farmacocinética , Antibacterianos/administración & dosificación , Antibacterianos/farmacología , Ciprofloxacina/administración & dosificación , Relación Dosis-Respuesta a Droga , Enoxacino/farmacología , Circulación Enterohepática/efectos de los fármacos , Inmunosupresores/administración & dosificación , Inmunosupresores/farmacocinética , Técnicas In Vitro , Concentración 50 Inhibidora , Levofloxacino/farmacología , Ofloxacino/farmacología , Fenolftaleínas/farmacología , Factores de Tiempo
10.
Toxicol Appl Pharmacol ; 273(3): 680-90, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-24183703

RESUMEN

Previous studies showed glucose and insulin signaling can regulate bile acid (BA) metabolism during fasting or feeding. However, limited knowledge is available on the effect of calorie restriction (CR), a well-known anti-aging intervention, on BA homeostasis. To address this, the present study utilized a "dose-response" model of CR, where male C57BL/6 mice were fed 0, 15, 30, or 40% CR diets for one month, followed by BA profiling in various compartments of the enterohepatic circulation by UPLC-MS/MS technique. This study showed that 40% CR increased the BA pool size (162%) as well as total BAs in serum, gallbladder, and small intestinal contents. In addition, CR "dose-dependently" increased the concentrations of tauro-cholic acid (TCA) and many secondary BAs (produced by intestinal bacteria) in serum, such as tauro-deoxycholic acid (TDCA), DCA, lithocholic acid, ω-muricholic acid (ωMCA), and hyodeoxycholic acid. Notably, 40% CR increased TDCA by over 1000% (serum, liver, and gallbladder). Interestingly, 40% CR increased the proportion of 12α-hydroxylated BAs (CA and DCA), which correlated with improved glucose tolerance and lipid parameters. The CR-induced increase in BAs correlated with increased expression of BA-synthetic (Cyp7a1) and conjugating enzymes (BAL), and the ileal BA-binding protein (Ibabp). These results suggest that CR increases BAs in male mice possibly through orchestrated increases in BA synthesis and conjugation in liver as well as intracellular transport in ileum.


Asunto(s)
Ácidos y Sales Biliares/sangre , Restricción Calórica , Circulación Enterohepática/efectos de los fármacos , Animales , Ácidos y Sales Biliares/biosíntesis , Glucemia/metabolismo , Peso Corporal , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Colesterol 7-alfa-Hidroxilasa/genética , Colesterol 7-alfa-Hidroxilasa/metabolismo , Ácidos Cólicos/sangre , Ácido Desoxicólico/sangre , Relación Dosis-Respuesta a Droga , Homeostasis , Intestinos/efectos de los fármacos , Intestinos/enzimología , Lípidos/sangre , Hígado/efectos de los fármacos , Hígado/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/genética , ARN Mensajero/metabolismo , Espectrometría de Masas en Tándem
11.
Yakugaku Zasshi ; 143(2): 101-104, 2023.
Artículo en Japonés | MEDLINE | ID: mdl-36724921

RESUMEN

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.


Asunto(s)
Colestasis , Circulación Enterohepática , Hígado , Animales , Humanos , Ratones , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Colestasis/metabolismo , Intestinos , Hígado/efectos de los fármacos , Hígado/metabolismo , Proteínas de Neoplasias/metabolismo , Farmacocinética , Circulación Enterohepática/efectos de los fármacos , Circulación Enterohepática/fisiología
12.
Drug Metab Dispos ; 40(9): 1846-53, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22715475

RESUMEN

A widely used metric of substrate exposure in brain is the brain-to-serum partition coefficient (K(p,brain); C(brain)/C(serum)), most appropriately determined at distribution equilibrium between brain tissue and serum. In some cases, C(brain)/C(serum) can peak and then decrease, as opposed to monotonically increasing to a plateau, precluding accurate estimation of partitioning. This "overshoot" has been observed with compounds that undergo enterohepatic recycling (ER), such as valproic acid (VPA). Previous simulation experiments identified a relationship between overshoot in the C(brain)/C(serum) versus time profile and distribution into a peripheral "compartment" (e.g., the ER loop). This study was conducted to evaluate model predictions of that relationship. Initial experiments tested the ability of activated charcoal, antibiotics, or Mrp2 deficiency to impair VPA ER in rats, thereby limiting the apparent volume of distribution associated with ER. Mrp2 deficiency (significantly) and antibiotics (moderately) interrupted VPA ER. Subsequently, brain partitioning was evaluated in the presence versus absence of ER modulation. Although overshoot was not eliminated completely, deconvolution revealed that overshoot was reduced in Mrp2-deficient and antibiotic-treated rats. Consistent with model predictions, overshoot was higher after antibiotic treatment (moderate ER interruption) than in Mrp2 deficiency (substantial ER interruption). Steady-state K(p,brain) was unaffected by experimental manipulation, also consistent with model predictions. These data support the hypothesis that C(brain)/C(serum) may overshoot K(p,brain) based on the extent of peripheral sequestration. Consideration of this information, particularly for compounds that undergo significant extravascular distribution, may be necessary to avoid erroneous estimation of K(p,brain).


Asunto(s)
Sangre/metabolismo , Circulación Enterohepática , Ácido Valproico/farmacocinética , Transportadoras de Casetes de Unión a ATP/deficiencia , Transportadoras de Casetes de Unión a ATP/genética , Animales , Antibacterianos/farmacología , Área Bajo la Curva , Sangre/efectos de los fármacos , Carbón Orgánico/farmacología , Interacciones Farmacológicas , Circulación Enterohepática/efectos de los fármacos , Inyecciones Intraperitoneales , Masculino , Modelos Biológicos , Permeabilidad , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Ácido Valproico/administración & dosificación , Ácido Valproico/sangre
13.
Ther Drug Monit ; 33(5): 658-62, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21860343

RESUMEN

BACKGROUND: The large interindividual differences observed in mycophenolic acid (MPA) pharmacokinetics (MPA-PK) are in part attributed to the large variability in enterohepatic recirculation (EHC) of the drug. The main metabolite of MPA, MPA glucuronide is actively secreted into the bile via the multidrug resistance-associated protein 2 (MRP2) transporter. Nonsteroidal anti-inflammatory drugs (NSAIDs) have been shown to inhibit the MRP2 transporter, which can alter EHC and drug exposure. Here, we evaluated the effects of this potential drug-transporter interaction on MPA-PK in a cohort of patients with childhood-onset systemic lupus erythematosus on mycophenolate mofetil therapy. MATERIALS AND METHODS: Full MPA concentration-time profiles and demographics including comedications were available for 19 patients with childhood-onset systemic lupus erythematosus. Concentrations at predose (C(trough)), 9 hour (C9), and nadir (C(nadir); defined as the lowest concentration between C(max) and C9), and area under the curve (AUC0₋12 and AUC6₋12) were assessed using standard methods (WinNonlin5.1). AUC6-12/AUC0₋12 and C9/C(nadir) ratios were used to evaluate the effects of NSAID treatment on MPA-PK. RESULTS: Eleven out of 19 patients were on NSAID treatment and did not show visual evidence of EHC in their PK profile. In contrast, patients not on NSAID therapy showed evidence of EHC-related MPA concentration increase in the later part of their PK profiles, typically after 6 hours. This phenomenon could be well characterized by the C9/C(nadir) ratio, which was significantly lower in the NSAID-treated cohort (P < 0.01). CONCLUSIONS: These preliminary data suggest that the concomitant intake of NSAIDs may lower EHC of MPA possibly through the inhibition of MRP2 transport of MPA-G. Further mechanism-based studies are needed to further elucidate this potential transporter interaction.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Lupus Eritematoso Sistémico/clasificación , Lupus Eritematoso Sistémico/tratamiento farmacológico , Ácido Micofenólico/metabolismo , Ácido Micofenólico/uso terapéutico , Adolescente , Circulación Enterohepática/efectos de los fármacos , Femenino , Regulación de la Expresión Génica/fisiología , Humanos , Masculino , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Ácido Micofenólico/farmacocinética
15.
Gut Microbes ; 13(1): 1949095, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34313539

RESUMEN

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.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Ácidos y Sales Biliares/farmacocinética , Ácidos y Sales Biliares/uso terapéutico , Circulación Enterohepática/efectos de los fármacos , Microbioma Gastrointestinal/efectos de los fármacos , Enfermedades Intestinales/tratamiento farmacológico , Intestinos/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Humanos
16.
Cell Metab ; 2(4): 217-25, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16213224

RESUMEN

The liver and intestine play crucial roles in maintaining bile acid homeostasis. Here, we demonstrate that fibroblast growth factor 15 (FGF15) signals from intestine to liver to repress the gene encoding cholesterol 7alpha-hydroxylase (CYP7A1), which catalyzes the first and rate-limiting step in the classical bile acid synthetic pathway. FGF15 expression is stimulated in the small intestine by the nuclear bile acid receptor FXR and represses Cyp7a1 in liver through a mechanism that involves FGF receptor 4 (FGFR4) and the orphan nuclear receptor SHP. Mice lacking FGF15 have increased hepatic CYP7A1 mRNA and protein levels and corresponding increases in CYP7A1 enzyme activity and fecal bile acid excretion. These studies define FGF15 and FGFR4 as components of a gut-liver signaling pathway that synergizes with SHP to regulate bile acid synthesis.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Circulación Enterohepática/fisiología , Factores de Crecimiento de Fibroblastos/metabolismo , Homeostasis , Transducción de Señal , Animales , Células CACO-2 , Colesterol 7-alfa-Hidroxilasa/biosíntesis , Clonación Molecular , Proteínas de Unión al ADN/metabolismo , Circulación Enterohepática/efectos de los fármacos , Epitelio/metabolismo , Factores de Crecimiento de Fibroblastos/deficiencia , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/farmacología , Perfilación de la Expresión Génica , Homeostasis/efectos de los fármacos , Humanos , Mucosa Intestinal/metabolismo , Intestinos/citología , Hígado/enzimología , Masculino , Ratones , Datos de Secuencia Molecular , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptores Citoplasmáticos y Nucleares/deficiencia , Receptores Citoplasmáticos y Nucleares/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/metabolismo
17.
Nutrients ; 12(5)2020 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-32380648

RESUMEN

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.


Asunto(s)
Colesterol/análogos & derivados , Corioamnionitis/tratamiento farmacológico , Corioamnionitis/microbiología , Portadores de Fármacos , Enterocolitis Necrotizante/microbiología , Enterocolitis Necrotizante/prevención & control , Circulación Enterohepática/efectos de los fármacos , Feto/irrigación sanguínea , Hígado/irrigación sanguínea , Fitosteroles/administración & dosificación , Fitoterapia , Profilaxis Posexposición/métodos , Sitoesteroles/administración & dosificación , Infecciones por Ureaplasma , Ureaplasma , beta-Ciclodextrinas , Animales , Colesterol/administración & dosificación , Colesterol/farmacología , Modelos Animales de Enfermedad , Femenino , Inflamación , Inyecciones Intralesiones , Fitosteroles/farmacología , Embarazo , Ovinos , Sitoesteroles/farmacología
18.
Am J Physiol Gastrointest Liver Physiol ; 297(3): G520-31, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19608735

RESUMEN

Essential fatty acid (EFA) deficiency in mice has been associated with increased bile production, which is mainly determined by the enterohepatic circulation (EHC) of bile salts. To establish the mechanism underlying the increased bile production, we characterized in detail the EHC of bile salts in EFA-deficient mice using stable isotope technique, without interrupting the normal EHC. Farnesoid X receptor (FXR) has been proposed as an important regulator of bile salt synthesis and homeostasis. In Fxr(-/-) mice we additionally investigated to what extent alterations in bile production during EFA deficiency were FXR dependent. Furthermore, we tested in differentiating Caco-2 cells the effects of EFA deficiency on expression of FXR-target genes relevant for feedback regulation of bile salt synthesis. EFA deficiency-enhanced bile flow and biliary bile salt secretion were associated with elevated bile salt pool size and synthesis rate (+146 and +42%, respectively, P < 0.05), despite increased ileal bile salt reabsorption (+228%, P < 0.05). Cyp7a1 mRNA expression was unaffected in EFA-deficient mice. However, ileal mRNA expression of Fgf15 (inhibitor of bile salt synthesis) was significantly reduced, in agreement with absent inhibition of the hepatic bile salt synthesis. Bile flow and biliary secretion were enhanced to the same extent in EFA-deficient wild-type and Fxr(-/-) mice, indicating contribution of other factors besides FXR in regulation of EHC during EFA deficiency. In vitro experiments show reduced induction of mRNA expression of relevant genes upon chenodeoxycholic acid and a selective FXR agonist GW4064 stimulation in EFA-deficient Caco-2 cells. In conclusion, our data indicate that EFA deficiency is associated with interrupted negative feedback of bile salt synthesis, possibly because of reduced ileal Fgf15 expression.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Bilis/metabolismo , Circulación Enterohepática , Ácidos Grasos Esenciales/deficiencia , Intestino Delgado/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Células CACO-2 , Ácido Quenodesoxicólico/metabolismo , Colesterol 7-alfa-Hidroxilasa/metabolismo , Circulación Enterohepática/efectos de los fármacos , Circulación Enterohepática/genética , Retroalimentación Fisiológica , Factores de Crecimiento de Fibroblastos/metabolismo , Humanos , Absorción Intestinal , Intestino Delgado/efectos de los fármacos , Isoxazoles/farmacología , Cinética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/metabolismo , Receptores Citoplasmáticos y Nucleares/deficiencia , Receptores Citoplasmáticos y Nucleares/genética
19.
Drug Metab Dispos ; 37(12): 2349-58, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19773540

RESUMEN

The hepatobiliary transport and local disposition of rosuvastatin in pig were investigated, along with the impact of concomitant dosing with two known multiple transport inhibitors; cyclosporine and gemfibrozil. Rosuvastatin (80 mg) was administered as an intrajejunal bolus dose in treatments I, II, and III (TI, TII, and TIII, respectively; n = 6 per treatment). Cyclosporine (300 mg) and gemfibrozil (600 mg) were administered in addition to the rosuvastatin dose in TII and TIII, respectively. Cyclosporine was administered as a 2-h intravenous infusion and gemfibrozil as an intrajejunal bolus dose. In treatment IV (TIV, n = 4) 5.9 mg of rosuvastatin was administered as an intravenous bolus dose. The study was conducted using a pig model, which enabled plasma sampling from the portal (VP), hepatic (VH), and femoral veins and bile from the common hepatic duct. The biliary recoveries of the administered rosuvastatin dose were 9.0 +/- 3.5 and 35.7 +/- 15.6% in TI and TIV, respectively. Rosuvastatin was highly transported into bile as shown by the median AUC(bile)/AUC(VH) ratio in TI of 1770 (1640-11,300). Gemfibrozil did not have an effect on the plasma pharmacokinetics of rosuvastatin, most likely because the unbound inhibitor concentrations did not exceed the reported IC(50) values. However, cyclosporine significantly reduced the hepatic extraction of rosuvastatin (TI, 0.89 +/- 0.06; TII, 0.46 +/- 0.13) and increased the AUC(VP) and AUC(VH) by 1.6- and 9.1-fold, respectively. In addition, the biliary exposure and f(e, bile) were reduced by approximately 50%. The strong effect of cyclosporine was in accordance with inhibition of sinusoidal uptake transporters, such as members of the organic anion-transporting polypeptide family, rather than canalicular transporters.


Asunto(s)
Bilis/metabolismo , Ciclosporina/farmacología , Circulación Enterohepática/efectos de los fármacos , Fluorobencenos/farmacocinética , Gemfibrozilo/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacocinética , Hígado/efectos de los fármacos , Pirimidinas/farmacocinética , Sulfonamidas/farmacocinética , Administración Oral , Animales , Área Bajo la Curva , Ciclosporina/administración & dosificación , Femenino , Fluorobencenos/administración & dosificación , Fluorobencenos/sangre , Gemfibrozilo/administración & dosificación , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Inhibidores de Hidroximetilglutaril-CoA Reductasas/sangre , Infusiones Intravenosas , Inyecciones Intravenosas , Absorción Intestinal/efectos de los fármacos , Yeyuno/efectos de los fármacos , Yeyuno/metabolismo , Hígado/metabolismo , Masculino , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Modelos Biológicos , Orquiectomía , Pirimidinas/administración & dosificación , Pirimidinas/sangre , Rosuvastatina Cálcica , Sulfonamidas/administración & dosificación , Sulfonamidas/sangre , Porcinos
20.
J Clin Pharmacol ; 49(6): 684-99, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19386625

RESUMEN

Mycophenolic acid (MPA) is mainly metabolized to MPA-glucuronide (MPAG), which may be reconverted to MPA following enterohepatic circulation (EHC). A physiologically realistic EHC model was proposed to estimate and assess the impact of cyclosporine (CsA) dose on the extent of EHC of MPA and MPAG. After the first oral dose of mycophenolate mofetil (MMF), the MPA and MPAG plasma concentration-time data of 14 adult renal transplant patients (12 receiving concomitant CsA and prednisolone and 2 receiving only concomitant prednisolone without CsA) were analyzed by individual pharmacokinetic modeling using a proposed 5-compartment drug and metabolite EHC model with a time-varying gallbladder emptying process. Simulations were performed to assess the influence of the time of bile release after dosing (T(bile)) and the gallbladder emptying interval (tau(gall)) on the EHC process. The extent of EHC for both MPA and MPAG tended to be lower in the group receiving CsA coadministration and decreased with increasing total body weight-adjusted CsA dose. Simulations revealed that T(bile) and tau(gall) influenced the time of occurrence and maximum concentration of the second peak, as well as the extent of EHC, for MPA and MPAG.


Asunto(s)
Ciclosporina/uso terapéutico , Circulación Enterohepática , Glucurónidos/farmacocinética , Inmunosupresores/farmacocinética , Ácido Micofenólico/análogos & derivados , Ácido Micofenólico/farmacocinética , Adulto , Simulación por Computador , Ciclosporina/administración & dosificación , Interacciones Farmacológicas , Quimioterapia Combinada , Circulación Enterohepática/efectos de los fármacos , Femenino , Vaciamiento Vesicular , Humanos , Inmunosupresores/uso terapéutico , Trasplante de Riñón , Masculino , Persona de Mediana Edad , Modelos Biológicos , Ácido Micofenólico/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA