Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 149
1.
Yakugaku Zasshi ; 143(2): 101-104, 2023.
Article Ja | MEDLINE | ID: mdl-36724921

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.


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
2.
Int J Oncol ; 61(4)2022 Oct.
Article En | MEDLINE | ID: mdl-35929515

Bile acids (BAs) are the major components of bile and products of cholesterol metabolism. Cholesterol is catalyzed by a variety of enzymes in the liver to form primary BAs, which are excreted into the intestine with bile, and secondary BAs are formed under the modification of the gut microbiota. Most of the BAs return to the liver via the portal vein, completing the process of enterohepatic circulation. BAs have an important role in the development of hepatocellular carcinoma (HCC), which may participate in the progression of HCC by recognizing receptors such as farnesoid X receptor (FXR) and mediating multiple downstream pathways. Certain BAs, such as ursodeoxycholic acid and obeticholic acid, were indicated to be able to delay liver injury and HCC progression. In the present review, the structure and function of BAs were introduced and the metabolism of BAs and the process of enterohepatic circulation were outlined. Furthermore, the mechanisms by which BAs participate in the development of HCC were summarized and possible strategies for targeting BAs and key sites of their metabolic processes to treat HCC were suggested.


Carcinoma, Hepatocellular , Liver Neoplasms , Bile Acids and Salts/metabolism , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Cholesterol/metabolism , Enterohepatic Circulation/physiology , Humans , Liver/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism
3.
Article En | MEDLINE | ID: mdl-34752368

OBJETIVE: To evaluate the enterohepatic circulation of 75-Selenium turoselecolic acid (75Se-SeHCAT) during the first 3 h and its correlation with the abdominal retention at the 7th day (AR7), as contribution to the clinical study of biliar acid malabsorption (BAM). MATERIALS AND METHODS: 38 patients with chronic diarrhea were retrospectively studied. Acquisition protocol included static abdominal images at 1st, 2nd and 3rd hour and the 7th day after oral administration of the radiopharmaceutical. Images of 1-3 h determined 5 patterns of enterohepatic circulation that, due to their location, were characterized as: 1) gallbladder 2-3 h, 2) gallbladder 3 h, 3) gallbladder-abdomen 2-3 h, 4) abdomen, 5) upper left abdomen. The association of these patterns with the AR7 (Fisher, STATA) were investigated. Patients were classified as Non BAM (AR7 > 15%), mild-BAM (AR7 15-10%), moderate-BAM (AR7 10-5%) or severe-BAM (AR7 < 5%). RESULTS: 19 patients had an AR7 diagnostic of BAM (7 mild-BAM, 5 moderate-BAM, 7 severe-BAM). The pattern "gallbladder at 2-3 h" was statistically associated with Non BAM (p 0,008), while "gallbladder-abdomen at 2-3 h" was correlated with having BAM (p 0,029). CONCLUSION: Variations detected at the abdominal level in images during the first 3 h were associated with changes in intestinal absorption and the incorporation of the radiopharmaceutical into the pool of bile acids, so visual interpretation of the images at 2nd and 3rd hour could be useful in the final assessment of the study.


Bile Acids and Salts/metabolism , Diarrhea/metabolism , Enterohepatic Circulation/physiology , Malabsorption Syndromes/diagnostic imaging , Taurocholic Acid/analogs & derivatives , Abdomen/diagnostic imaging , Adult , Aged , Aged, 80 and over , Chronic Disease , Diarrhea/etiology , Female , Gallbladder/diagnostic imaging , Gallbladder/metabolism , Humans , Intestinal Absorption , Malabsorption Syndromes/metabolism , Male , Middle Aged , Retrospective Studies , Taurocholic Acid/administration & dosage , Taurocholic Acid/pharmacokinetics , Time Factors , Young Adult
4.
Am J Physiol Gastrointest Liver Physiol ; 321(3): G270-G279, 2021 09 01.
Article En | MEDLINE | ID: mdl-34288725

The use of human tissue stem cell-derived organoids has advanced our knowledge of human physiological and pathophysiological processes that are unable to be studied using other model systems. Increased understanding of human epithelial tissues including intestine, stomach, liver, pancreas, lung, and brain have been achieved using organoids. However, it is not yet clear whether these cultures recapitulate in vivo organ-to-organ signaling or communication. In this work, we demonstrate that mature stem cell-derived intestinal and liver organoid cultures each express functional molecules that modulate bile acid uptake and recycling. These organoid cultures can be physically coupled in a Transwell system and display increased secretion of fibroblast growth factor 19 (FGF19) (intestine) and downregulation of P450 enzyme cholesterol 7 α-hydroxylase (CYP7A) (liver) in response to apical exposure of the intestine to bile acids. This work establishes that organoid cultures can be used to study and therapeutically modulate interorgan interactions and advance the development of personalized approaches to medical care.NEW & NOTEWORTHY Interorgan signaling is a critical feature of human biology and physiology, yet has remained difficult to study due to the lack of in vitro models. Here, we demonstrate that physical coupling of ex vivo human intestine and liver epithelial organoid cultures recapitulates in vivo interorgan bile acid signaling. These results suggest that coupling of multiple organoid systems provides new models to investigate interorgan communication and advances our knowledge of human physiological and pathophysiological processes.


Cell Differentiation/physiology , Intestines/cytology , Organoids/cytology , Stem Cells/cytology , Cells, Cultured , Enterohepatic Circulation/physiology , Humans , Liver/metabolism , Stomach/cytology
5.
J Clin Pharmacol ; 61(12): 1592-1605, 2021 12.
Article En | MEDLINE | ID: mdl-34169529

Mycophenolic acid exhibits significant interpatient pharmacokinetic variability attributed to factors including race, sex, concurrent medications, and enterohepatic circulation of the mycophenolic acid glucuronide metabolite to mycophenolic acid. This conversion by enterohepatic circulation is mediated by the multidrug resistance-associated protein 2, encoded by ABCC2. This study investigated ABCC2 haplotype associations with mycophenolic acid pharmacokinetics in 147 stable kidney transplant recipients receiving mycophenolic acid in combination with calcineurin inhibitors. The role of the ABCC2 genotypes -24C>T (rs717620), 1249C>T (rs2273697), and 3972C>T (rs3740066) were evaluated in prospective, cross-sectional pharmacokinetic studies of stable recipients receiving mycophenolic acid and either tacrolimus or cyclosporine. Haplotype phenotypic associations with mycophenolic acid pharmacokinetic parameters were computed using THESIAS (v. 3.1). Four ABCC2 haplotypes with estimated frequencies greater than 10% were identified (H1:CGC [wild type], H9:CGT, H2:CAC, H12:TGT). There were no differences in haplotype frequencies by either race or sex. There were significant associations of pharmacokinetic parameters with ABCC2 haplotypes for mycophenolic acid clearance (L/h), mycophenolic acid AUC0-12h (mg·h/L), and the ratio of mycophenolic acid glucuronide to mycophenolic acid AUC0-12h . The wild-type haplotype ABCC2 CGC had greater mycophenolic acid AUC0-12h (P = .017), slower clearance (P = .013), and lower mycophenolic acid glucuronide to mycophenolic acid AUC0-12h ratio (P = .047) compared with the reduced function ABCC2 haplotype CGT. These differences were most pronounced among patients receiving tacrolimus cotreatment. No phenotypic associations were found with the cyclosporine-mycophenolic acid regimen. Variation in ABCC2 haplotypes contributes to subtherapeutic mycophenolic acid exposure and influences interpatient variability in pharmacokinetic phenotypes based on concurrent calcineurin inhibitor treatment.


Immunosuppressive Agents/pharmacokinetics , Kidney Transplantation , Multidrug Resistance-Associated Protein 2/genetics , Adult , Area Under Curve , Calcineurin Inhibitors/administration & dosage , Calcineurin Inhibitors/pharmacology , Cross-Sectional Studies , Enterohepatic Circulation/physiology , Female , Haplotypes , Humans , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/pharmacology , Male , Middle Aged , Mycophenolic Acid/pharmacokinetics , Prospective Studies
6.
Am J Physiol Gastrointest Liver Physiol ; 321(1): G55-G66, 2021 07 01.
Article En | MEDLINE | ID: mdl-33978477

Regulation of bile acid metabolism is normally discussed as the regulation of bile acid synthesis, which serves to compensate for intestinal loss in order to maintain a constant pool size. After a meal, bile acids start cycling in the enterohepatic circulation. Farnesoid X receptor-dependent ileal and hepatic processes lead to negative feedback inhibition of bile acid synthesis. When the intestinal bile acid flux decreases, the inhibition of synthesis is released. The degree of inhibition of synthesis and the mechanism and degree of activation are still unknown. Moreover, in humans, a biphasic diurnal expression pattern of bile acid synthesis has been documented, indicating maximal synthesis around 3 PM and 9 PM. Quantitative data on the hourly synthesis schedule as compensation for intestinal loss are lacking. In this review, we describe the classical view on bile acid metabolism and present alternative concepts that are based on the overlooked feature that bile acids transit through the enterohepatic circulation very rapidly. A daily profile of the cycling and total bile acid pool sizes and potential controlled and uncontrolled mechanisms for synthesis are predicted. It remains to be elucidated by which mechanism clock genes interact with the Farnesoid X receptor-controlled regulation of bile acid synthesis. This mechanism could become an attractive target to enhance bile acid synthesis at night, when cholesterol synthesis is high, thus lowering serum LDL-cholesterol.


Bile Acids and Salts/metabolism , Enterohepatic Circulation/physiology , Intestines/physiology , Liver/metabolism , Animals , Feedback, Physiological/physiology , Humans , Ileum/metabolism
7.
ACS Appl Mater Interfaces ; 13(20): 23314-23327, 2021 May 26.
Article En | MEDLINE | ID: mdl-33587600

Intermittent subcutaneous (S.C.) injection of teriparatide [PTH (1-34)] is one of the effective therapies to cure osteoporosis. However, a long-term repeated administration of teriparatide by S.C. to the patients is highly challenging. Herein, a triple padlock nanocarrier prepared by a taurocholic acid-conjugated chondroitin sulfate A (TCSA) is designed to develop an oral dosage form of recombinant human teriparatide (rhPTH). Oral administration of TCSA/rhPTH to the bilateral ovariectomized (OVX) rats resulted in the recovery of the bone marrow density and healthy serum bone parameters from the severe osteoporotic conditions. Also, it enhanced new bone formation in the osteoporotic tibias. This triple padlock oral delivery platform overcame the current barriers associated with teriparatide administration and exhibited a promising therapeutic effect against osteoporosis.


Drug Carriers , Enterohepatic Circulation/physiology , Nanoparticles , Osteoporosis/metabolism , Teriparatide , Administration, Oral , Animals , Bone and Bones/drug effects , Bone and Bones/physiology , Chondroitin Sulfates/chemistry , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Female , Humans , Mice , Mice, Inbred ICR , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Nanoparticles/metabolism , Osteoblasts/drug effects , Ovariectomy , Rats , Rats, Sprague-Dawley , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacokinetics , Taurocholic Acid/chemistry , Teriparatide/administration & dosage , Teriparatide/pharmacokinetics , Teriparatide/pharmacology
8.
Dig Dis Sci ; 66(2): 568-576, 2021 02.
Article En | MEDLINE | ID: mdl-32198567

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.


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
9.
Hepatology ; 73(6): 2577-2585, 2021 06.
Article En | MEDLINE | ID: mdl-33222321

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.


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
10.
Cell Mol Gastroenterol Hepatol ; 10(1): 149-170, 2020.
Article En | MEDLINE | ID: mdl-32112828

BACKGROUND & AIMS: Disturbances of the enterohepatic circulation of bile acids (BAs) are seen in a number of clinically important conditions, including metabolic disorders, hepatic impairment, diarrhea, and gallstone disease. To facilitate the exploration of underlying pathogenic mechanisms, we developed a mathematical model built on quantitative physiological observations across different organs. METHODS: The model consists of a set of kinetic equations describing the syntheses of cholic, chenodeoxycholic, and deoxycholic acids, as well as time-related changes of their respective free and conjugated forms in the systemic circulation, the hepatoportal region, and the gastrointestinal tract. The core structure of the model was adapted from previous modeling research and updated based on recent mechanistic insights, including farnesoid X receptor-mediated autoregulation of BA synthesis and selective transport mechanisms. The model was calibrated against existing data on BA distribution and feedback regulation. RESULTS: According to model-based predictions, changes in intestinal motility, BA absorption, and biotransformation rates affected BA composition and distribution differently, as follows: (1) inhibition of transintestinal BA flux (eg, in patients with BA malabsorption) or acceleration of intestinal motility, followed by farnesoid X receptor down-regulation, was associated with colonic BA accumulation; (2) in contrast, modulation of the colonic absorption process was predicted to not affect the BA pool significantly; and (3) activation of ileal deconjugation (eg, in patents with small intestinal bacterial overgrowth) was associated with an increase in the BA pool, owing to higher ileal permeability of unconjugated BA species. CONCLUSIONS: This model will be useful in further studying how BA enterohepatic circulation modulation may be exploited for therapeutic benefits.


Bile Acids and Salts/metabolism , Liver/metabolism , Models, Biological , Diarrhea/metabolism , Diarrhea/pathology , Enterohepatic Circulation/physiology , Gallbladder/metabolism , Gallbladder/pathology , Gallstones/metabolism , Gallstones/pathology , Gastrointestinal Motility/physiology , Humans , Ileum/metabolism , Intestinal Absorption/physiology , Intestinal Mucosa/metabolism , Liver/pathology , Liver Diseases/metabolism , Liver Diseases/pathology , Metabolic Diseases/metabolism , Metabolic Diseases/pathology , Permeability
11.
Cells ; 8(11)2019 10 24.
Article En | MEDLINE | ID: mdl-31653030

Hepatic fibrosis is the consequence of an unresolved wound healing process in response to chronic liver injury and involves multiple cell types and molecular mechanisms. The hepatic endocannabinoid and apelin systems are two signalling pathways with a substantial role in the liver fibrosis pathophysiology-both are upregulated in patients with advanced liver disease. Endogenous cannabinoids are lipid-signalling molecules derived from arachidonic acid involved in the pathogenesis of cardiovascular dysfunction, portal hypertension, liver fibrosis, and other processes associated with hepatic disease through their interactions with the CB1 and CB2 receptors. Apelin is a peptide that participates in cardiovascular and renal functions, inflammation, angiogenesis, and hepatic fibrosis through its interaction with the APJ receptor. The endocannabinoid and apelin systems are two of the multiple cell-signalling pathways involved in the transformation of quiescent hepatic stellate cells into myofibroblast like cells, the main matrix-producing cells in liver fibrosis. The mechanisms underlying the control of hepatic stellate cell activity are coincident despite the marked dissimilarities between the endocannabinoid and apelin signalling pathways. This review discusses the current understanding of the molecular and cellular mechanisms by which the hepatic endocannabinoid and apelin systems play a significant role in the pathophysiology of liver fibrosis.


Apelin/metabolism , Endocannabinoids/metabolism , Liver Cirrhosis/pathology , Liver/metabolism , Carrier Proteins/metabolism , Enterohepatic Circulation/physiology , Fibrosis , Humans , Hypertension, Portal , Inflammation/pathology , Liver/pathology , Liver Circulation/physiology , Portal System/physiology
12.
Obes Surg ; 29(6): 1901-1910, 2019 06.
Article En | MEDLINE | ID: mdl-30805859

BACKGROUND AND AIMS: Duodenal-jejunal bypass (DJB) shows great effects on weight loss and diabetes improvement. Previously, we reported that the bilio-pancreatic (BP) limb plays an important role in glycemic improvement and in serum bile acid (BA) level increase as reported by Miyachi et al. (Surgery 159(5):1360-71, 2016). This study aimed to investigate the mechanism of BA elevation after DJB and the relationship between these effects and BP-limb length. METHODS: Otsuka Long-Evans Tokushima Fatty rats with diabetes were randomly assigned into four groups: one sham group and three DJB groups. Three DJB groups were defined according to the BP-limb length: 0 cm, 15 cm, and 30 cm. The lengths of the alimentary limb and common channel were set equally in each DJB groups. Body weight, glucose tolerance, and BA levels in the liver, bile juice, portal vein, and intestinal contents were assessed postoperatively. Changes in enterohepatic circulation of BAs were assessed using labeled BA. RESULTS: BA elevation after DJB was higher with longer BP-limb. In the 30-cm group, the serum total BA level and BA levels in the portal vein, liver, and bile juice were greater than those in other groups. The enterohepatic circulation was shortened in the 15-cm and 30-cm groups. CONCLUSIONS: Shortening of the "enterohepatic circulation" by early reabsorption of BAs in the BP-limb, not by the early influx of bile juice into the ileum, was the main cause of BA elevation after DJB. Thus, glycemic improvement and elevation of BA concentration after DJB depend on the BP-limb length.


Bile Acids and Salts/metabolism , Biliary Tract/metabolism , Diabetes Mellitus, Experimental/metabolism , Enterohepatic Circulation/physiology , Pancreas/metabolism , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/surgery , Duodenum/surgery , Jejunum/surgery , Male , Obesity, Morbid/metabolism , Obesity, Morbid/surgery , Rats , Rats, Sprague-Dawley , Weight Loss
13.
Eur J Drug Metab Pharmacokinet ; 44(4): 493-504, 2019 Aug.
Article En | MEDLINE | ID: mdl-30488336

BACKGROUND AND OBJECTIVES: Strategies for modeling the enterohepatic circulation (EHC) process reported in the literature vary; however, gallbladder-based models currently provide the best physiological representation of the process. Regardless, the addition of a gallbladder to the model does not fully depict the physiology of EHC. A more physiological gallbladder-based EHC model is needed. This model should take into account a physiological representation of the bile secretion, gallbladder filling and emptying, the duration of gallbladder emptying, and irregular mealtimes. Considering all of these factors, the objectives of the present analysis were to propose a gallbladder-based EHC model and then to use that model to perform sensitivity analyses evaluating the effect of the extent of EHC on the pharmacokinetic profile and noncompartmental analysis (NCA) calculations. METHODS: A gallbladder-based model that describes the EHC process was developed and used to perform determinant simulations assuming various degrees of EHC. Next, these simulations were compared to evaluate the effect of the EHC on the pharmacokinetic profiles of orally administered drugs. The influence of the EHC process on the NCA calculations was determined while assuming two sampling schemes that differed in the times at which sampling was performed in relation to meal times. RESULTS: The presence of EHC results in nonlinearity in the system and changes the pharmacokinetic profile, affecting the maximum concentration (Cmax), time to Cmax (Tmax), and half-life estimates. Comparison of the results obtained using the two sampling schemes for a drug undergoing various degrees of EHC demonstrated a significant influence of the selected sampling times on the NCA estimations. Bias in the NCA calculations was also dependent on the sampling times used. CONCLUSION: Caution should be taken when designing clinical studies for drugs that undergo EHC. It may be essential to consider the timing of meals when planning pharmacokinetic studies and defining sampling times. The period over which samples are taken needs to be extended as compared to that traditionally used with other drugs. Future studies that attempt to identify the best sampling strategies in the presence of EHC are needed.


Enterohepatic Circulation/physiology , Gallbladder/metabolism , Half-Life , Humans , Models, Biological
14.
J Clin Pharmacol ; 58(5): 628-639, 2018 05.
Article En | MEDLINE | ID: mdl-29329489

Mycophenolic acid (MPA) is an approved immunosuppressive agent widely prescribed to prevent rejection after kidney transplantation. Wide between-subject variability (BSV) in MPA exposure exists which in part may be due to variability in enterohepatic recirculation (EHC). Several modeling strategies were developed to evaluate EHC as part of MPA pharmacokinetics, however mechanistic representation of EHC is limited. These models have not provided a satisfactory representation of the physiology of EHC in their modeling assumptions. The aim of this study was i) to develop an integrated model of MPA (total and unbound) and its metabolites (MPAG and acyl-MPAG) in kidney recipients, where this model provides a more physiological representation of EHC process, and ii) to evaluate the effect of donor and recipient clinical covariates and genotypes on MPA disposition. A five-compartment model with first-order input into an unbound MPA compartment connected to the MPAG, acyl-MPAG, and gallbladder compartment best fit the data. To represent the EHC process, the model was built based on the physiological concepts related to the hepatobiliary system and the gallbladder filling and emptying processes. The effect of cyclosporine versus tacrolimus on clearance of unbound MPA was included in the base model. Covariate analysis showed creatinine clearance to be significant on oral clearance of unbound MPA. The hepatic nuclear factor 1 alpha (HNF1A) genetic single nucleotide polymorphism (SNP) (rs2393791) in the recipient significantly affected the fraction of enterohepatically-circulated drug. Oral clearance of MPAG was affected by recipient IMPDH1 SNP (rs2288553), diabetes at the time of transplant, and donor sex.


Immunosuppressive Agents/pharmacokinetics , Kidney Transplantation , Models, Biological , Mycophenolic Acid/pharmacokinetics , Adult , Cyclosporine/administration & dosage , Cyclosporine/pharmacology , Drug Interactions , Enterohepatic Circulation/physiology , Female , Humans , Immunosuppressive Agents/administration & dosage , Male , Middle Aged , Mycophenolic Acid/administration & dosage , Tacrolimus/administration & dosage , Tacrolimus/pharmacology
15.
Am J Physiol Gastrointest Liver Physiol ; 314(5): G537-G546, 2018 05 01.
Article En | MEDLINE | ID: mdl-29351394

Bile acids (BAs), which are synthesized in the liver and cycled in the enterohepatic circulation, have been recognized as signaling molecules by activating their receptors in the intestine and liver. Serum taurine-conjugated BAs have been shown to be elevated after bariatric surgeries although the postoperative BA profiles within the enterohepatic circulation have not been investigated. Clarification of these profiles could help explain the mechanisms by which bariatric surgery leads to BA profile alterations and subsequent metabolic effects. We performed duodenal-jejunal bypass (DJB), sleeve gastrectomy (SG), and sham procedures in an obese diabetic rat model induced by high-fat diet and streptozotocin. The weight loss and antidiabetic effects were evaluated postsurgery. BA profiles in the systemic serum and within the enterohepatic circulation were analyzed, together with the expression of related BA transporters and enzymes at week 12 after surgery. Compared with sham, SG induced sustained weight loss, and both DJB and SG significantly improved glucose tolerance and insulin sensitivity with enhanced glucagon-like peptide 1 secretion. Similar to changes in the serum, BAs, especially taurine-conjugated species, were also elevated in the enterohepatic circulation (bile and portal vein) after DJB and SG. In addition, the expression of key BA transporters and conjugational enzymes was elevated postoperatively, whereas the enzymes responsible for BA synthesis were decreased. In conclusion, DJB and SG elevated BA levels in the systemic serum and enterohepatic circulation, especially taurine-conjugated species, which likely indicates increased ileal reabsorption and hepatic conjugation rather than synthesis. NEW & NOTEWORTHY Bile acids (BAs) have been implicated as potential mediators of the weight-independent effects of bariatric surgery. For the first time, we discovered that duodenal-jejunal bypass and sleeve gastrectomy elevated BAs, particularly the taurine-conjugated species in the enterohepatic circulation, likely through the promotion of ileal reabsorption and hepatic conjugation rather than BA synthesis. These findings will improve our understanding of BA metabolism after bariatric surgery and their subsequent metabolic effects.


Bariatric Surgery , Bile Acids and Salts , Enterohepatic Circulation/physiology , Obesity , Postoperative Complications/metabolism , Taurine/metabolism , Animals , Bariatric Surgery/adverse effects , Bariatric Surgery/classification , Bariatric Surgery/methods , Bile Acids and Salts/blood , Bile Acids and Salts/metabolism , Blood Glucose/metabolism , Body Weight/physiology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2 , Insulin Resistance , Intestinal Reabsorption/physiology , Obesity/metabolism , Obesity/physiopathology , Obesity/surgery , Rats
16.
Mol Aspects Med ; 56: 90-100, 2017 08.
Article En | MEDLINE | ID: mdl-28506676

There are numerous profound maternal physiological changes that occur from conception onwards and adapt throughout gestation in order to support a healthy pregnancy. By the time of late gestation, when circulating pregnancy hormones are at their highest concentrations, maternal adaptations include relative hyperlipidemia, hypercholanemia and insulin resistance. Bile acids have now been established as key regulators of metabolism, and their role in gestational changes in metabolism is becoming apparent. Bile acid homeostasis is tightly regulated by the nuclear receptor FXR, which has been shown to have reduced activity during pregnancy. This review focuses on the gestational alterations in bile acid homeostasis that occur in normal pregnancy, which in some women can become pathological, leading to the development of intrahepatic cholestasis of pregnancy. As well as their important role in maternal metabolic health, we will review bile acid metabolism in the feto-placental unit.


Bile Acids and Salts/metabolism , Cholestasis, Intrahepatic/metabolism , Diabetes, Gestational/metabolism , Homeostasis/physiology , Receptors, Cytoplasmic and Nuclear/genetics , Cholestasis, Intrahepatic/genetics , Cholestasis, Intrahepatic/pathology , Diabetes, Gestational/genetics , Diabetes, Gestational/pathology , Enterohepatic Circulation/physiology , Female , Fetus , Gene Expression Regulation , Gestational Age , Humans , Liver/metabolism , Placenta/metabolism , Pregnancy , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction
17.
Clin Res Hepatol Gastroenterol ; 41(5): 509-515, 2017 Oct.
Article En | MEDLINE | ID: mdl-28336180

The enterohepatic circulation of bile acids (BAs) is governed by specific transporters expressed in the liver and the intestine and plays a critical role in the digestion of fats and oils. During this process, the majority of the BAs secreted from the liver is reabsorbed in intestinal epithelial cells via the apical sodium-dependent bile acid transporter (ASBT/SLC10A2) and then transported into the portal vein. Previous studies revealed that regulation of the ASBT involves BAs and cholesterol. In addition, abnormal ASBT expression and function might lead to some diseases associated with disorders in the enterohepatic circulation of BAs and cholesterol homeostasis, such as diarrhoea and gallstones. However, decreasing cholesterol or BAs by partly inhibiting ASBT-mediated transport might be used for treatments of hypercholesterolemia, cholestasis and diabetes. This review mainly discusses the regulation of the ASBT by BAs and cholesterol and its relevance to diseases and treatment.


Bile Acids and Salts/physiology , Cholesterol/physiology , Enterohepatic Circulation/physiology , Homeostasis , Organic Anion Transporters, Sodium-Dependent/physiology , Symporters/physiology , Humans
18.
Curr Opin Gastroenterol ; 33(3): 189-195, 2017 May.
Article En | MEDLINE | ID: mdl-28169840

PURPOSE OF REVIEW: Our objective was to review advances in bile acids in health and disease published in the last 2 years. Bile acid diarrhea (BAD) is recognized as a common cause of chronic diarrhea, and its recognition has been facilitated by development of new screening tests. RECENT FINDINGS: Primary BAD can account for 30% of cases of chronic diarrhea. The mechanisms leading to BAD include inadequate feedback regulation by fibroblast growth factor 19 (FGF-19) from ileal enterocytes, abnormalities in synthesis or degradation of proteins involved in FGF-19 regulation in hepatocytes and variations as a function of the bile acid receptor, TGR5 (GPBAR1). SeHCAT is the most widely used test for diagnosis of BAD. There has been significant validation of fasting serum FGF-19 and 7 α-hydroxy-cholesten-3-one (C4), a surrogate measure of bile acid synthesis. Bile acid sequestrants are the primary treatments for BAD; the farnesoid X-receptor-FGF-19 pathway provides alternative therapeutic targets for BAD. Bile acid-stimulated intestinal mechanisms contribute to the beneficial effects of bariatric surgery on obesity, glycemic control and the treatment of recurrent Clostridium difficile infection. SUMMARY: Renewed interest in the role of bile acids is leading to novel management of diverse diseases besides BAD.


Bile Acids and Salts/physiology , Diarrhea/metabolism , Antidiarrheals/therapeutic use , Chronic Disease , Diarrhea/diagnosis , Diarrhea/epidemiology , Diarrhea/therapy , Enterohepatic Circulation/physiology , Fibroblast Growth Factors/metabolism , Gastric Bypass , Humans , Obesity, Morbid/metabolism , Obesity, Morbid/surgery
19.
Obes Surg ; 26(10): 2492-502, 2016 10.
Article En | MEDLINE | ID: mdl-27475800

Obesity and metabolic surgery (OMS) leads to several metabolic improvements, which often occur prior to substantial weight loss. Therefore, other factors in addition to weight loss contribute to the metabolic benefits. This literature review offers an overview of studies investigating bile acids (BAs) and their metabolic effects after OMS. Rearrangement of enterohepatic circulation, changes in BA synthesis, BA conjugation, intestinal reabsorption, and alterations in the gut microbiota are potential mechanisms for altered BA profiles after surgery. Increased BA levels are associated with improved glucose homeostasis and lipid profiles, which are mediated by two major receptors: the Transmembrane G-protein Coupled Receptor and the Farnesoid X Receptor. Therefore, pharmacological manipulation of BAs and their receptors may be viable targets for less invasive obesity treatment.


Bariatric Surgery , Bile Acids and Salts/metabolism , Enterohepatic Circulation/physiology , Gastrointestinal Microbiome/physiology , Obesity/physiopathology , Obesity/surgery , Adaptation, Physiological/physiology , Bile Acids and Salts/biosynthesis , Homeostasis , Humans , Intestines/physiopathology , Obesity/therapy
20.
Drug Metab Rev ; 48(2): 281-327, 2016 05.
Article En | MEDLINE | ID: mdl-26987379

Enterohepatic recirculation (EHC) concerns many physiological processes and notably affects pharmacokinetic parameters such as plasma half-life and AUC as well as estimates of bioavailability of drugs. Also, EHC plays a detrimental role as the compounds/drugs are allowed to recycle. An in-depth comprehension of this phenomenon and its consequences on the pharmacological effects of affected drugs is important and decisive in the design and development of new candidate drugs. EHC of a compound/drug occurs by biliary excretion and intestinal reabsorption, sometimes with hepatic conjugation and intestinal deconjugation. EHC leads to prolonged elimination half-life of the drugs, altered pharmacokinetics and pharmacodynamics. Study of the EHC of any drug is complicated due to unavailability of the apposite model, sophisticated procedures and ethical concerns. Different in vitro and in vivo methods for studies in experimental animals and humans have been devised, each having its own merits and demerits. Involvement of the different transporters in biliary excretion, intra- and inter-species, pathological and biochemical variabilities obscure the study of the phenomenon. Modeling of drugs undergoing EHC has always been intricate and exigent models have been exploited to interpret the pharmacokinetic profiles of drugs witnessing multiple peaks due to EHC. Here, we critically appraise the mechanisms of bile formation, factors affecting biliary drug elimination, methods to estimate biliary excretion of drugs, EHC, multiple peak phenomenon and its modeling.


Bile/metabolism , Biological Availability , Enterohepatic Circulation/physiology , Pharmaceutical Preparations/metabolism , Animals , Humans , Models, Biological
...