Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Clin Pharmacokinet ; 54(2): 179-93, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25316573

ABSTRACT

BACKGROUND AND OBJECTIVE: Accumulating evidence has shown that diabetes mellitus may affect the pharmacokinetics of some drugs, leading to alteration of pharmacodynamics and/or toxic effects. The aim of this study was to develop a novel physiologically based pharmacokinetic (PBPK) model for predicting drug pharmacokinetics in patients with type 2 diabetes mellitus quantitatively. METHODS: Contributions of diabetes-induced alteration of physiological parameters including gastric emptying rates, intestinal transit time, drug metabolism in liver and kidney functions were incorporated into the model. Plasma concentration-time profiles and pharmacokinetic parameters of seven drugs (antipyrine, nisoldipine, repaglinide, glibenclamide, glimepiride, chlorzoxazone, and metformin) in non-diabetic and diabetic patients were predicted using the developed model. The PBPK model coupled with a Monte-Carlo simulation was also used to predict the means and variability of pharmacokinetic parameters. RESULTS: The predicted area under the plasma concentration-time curve (AUC) and maximum (peak) concentration (C max) were reasonably consistent (<2-fold errors) with the reported values. Sensitivity analysis showed that gut transit time, hepatic enzyme activity, and renal function affected the pharmacokinetic characteristics of these drugs. Shortened gut transit time only decreased the AUC of controlled-released drugs and drugs with low absorption rates. Impairment of renal function markedly altered pharmacokinetics of drugs mainly eliminated via the kidneys. CONCLUSION: All of these results indicate that the developed PBPK model can quantitatively predict pharmacokinetic alterations induced by diabetes.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Hypoglycemic Agents/pharmacokinetics , Area Under Curve , Computer Simulation , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/drug therapy , Humans , Hypoglycemic Agents/blood , Kidney/metabolism , Liver/metabolism , Male , Models, Biological , Monte Carlo Method
2.
Metabolism ; 63(10): 1342-51, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25060691

ABSTRACT

OBJECTIVE: Ginsenosides, major bioactive constituents in Panax ginseng, have been shown to exert anti-hyperlipidemia effects. However, the underlying mechanism was not well-elucidated due to the low bioavailability of ginsenosides. Glucagon-like peptide-1 (GLP-1) was considered to be a critical regulator of energy homeostasis. Our previous studies have showed that ginseng total saponins (GTS) exhibited antidiabetic effects partly via modulating GLP-1 release. The aim of this study was to investigate the potential role of GLP-1 in anti-hyperlipidemia effect of GTS in rats fed with high-fat diet. MATERIAL AND METHODS: Male Sprague-Dawley rats were fed with normal diet (CON) or high-fat diet (HFD) for 4 weeks. Then, the HFD rats orally received vehicle (HFD), 150 mg/kg/day (HFD-GL) and 300 mg/kg/day of GTS (HFD-GH) for another 4 weeks, respectively. RESULTS: Four-week GTS treatment significantly ameliorated hyperlipidemia, decreased body fat, liver weight and improved insulin resistance. It was found that high-dose GTS treatment increased portal GLP-1 level induced by glucose loading, accompanied by increased intestinal GLP-1 content, L-cell number and prohormone convertase 3 mRNA expression. Data from NCI-H716 cells showed that both GTS and ginsenoside Rb1 significantly increased GLP-1 secretion as well as proglucagon mRNA level in NCI-H716 cells supplemented with 10% HFD-rat serum. CONCLUSIONS: Hyperlipidemia and insulin resistance were attenuated effectively in response to GTS treatment. These improvements may be associated with the increased secretion of GLP-1.


Subject(s)
Diet, High-Fat/adverse effects , Ginsenosides/pharmacology , Glucagon-Like Peptide 1/metabolism , Hypoglycemic Agents/pharmacology , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Body Weight/drug effects , Cell Line , Energy Intake/drug effects , Glucose/metabolism , Humans , Hyperlipidemias/blood , Hyperlipidemias/drug therapy , Hyperlipidemias/metabolism , Insulin/blood , Insulin Resistance , Lipids/blood , Male , Rats , Rats, Sprague-Dawley
3.
J Endocrinol ; 217(2): 185-96, 2013 May.
Article in English | MEDLINE | ID: mdl-23444389

ABSTRACT

Panax ginseng is one of the most popular herbal remedies. Ginsenosides, major bioactive constituents in P. ginseng, have shown good antidiabetic action, but the precise mechanism was not fully understood. Glucagon-like peptide-1 (GLP1) is considered to be an important incretin that can regulate glucose homeostasis in the gastrointestinal tract after meals. The aim of this study was to investigate whether ginseng total saponins (GTS) exerts its antidiabetic effects via modulating GLP1 release. Ginsenoside Rb1 (Rb1), the most abundant constituent in GTS, was selected to further explore the underlying mechanisms in cultured NCI-H716 cells. Diabetic rats were developed by a combination of high-fat diet and low-dose streptozotocin injection. The diabetic rats orally received GTS (150 or 300 mg/kg) daily for 4 weeks. It was found that GTS treatment significantly ameliorated hyperglycemia and dyslipidemia, accompanied by a significant increase in glucose-induced GLP1 secretion and upregulation of proglucagon gene expression. Data from NCI-H716 cells showed that both GTS and Rb1 promoted GLP1 secretion. It was observed that Rb1 increased the ratio of intracellular ATP to ADP concentration and intracellular Ca2+ concentration. The metabolic inhibitor azide (3 mM), the KATP channel opener diazoxide (340 µM), and the Ca2+ channel blocker nifedipine (20 µM) significantly reversed Rb1-mediated GLP1 secretion. All these results drew a conclusion that ginsenosides stimulated GLP1 secretion both in vivo and in vitro. The antidiabetic effects of ginsenosides may be a result of enhanced GLP1 secretion.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diet, High-Fat/adverse effects , Ginsenosides/pharmacology , Glucagon-Like Peptide 1/metabolism , Hypoglycemic Agents/pharmacology , Streptozocin/adverse effects , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , Calcium/metabolism , Cell Line , Cells, Cultured , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/etiology , Disease Models, Animal , Gastrointestinal Tract/cytology , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/metabolism , Glucose/metabolism , Homeostasis , Humans , In Vitro Techniques , Male , Rats , Rats, Sprague-Dawley , Saponins/pharmacology
4.
Liver Int ; 33(2): 274-82, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22925079

ABSTRACT

BACKGROUND: P-glycoprotein (P-GP) and multidrug resistance-associated protein 2 (MRP2) are involved in transport of many drugs across blood-brain barrier (BBB). The function and expression of P-GP and MRP2 may be modulated by different pathologies. Acute liver failure (ALF) was reported to impair BBB function, resulting in the increased BBB permeability. AIMS: We investigated whether ALF altered function and expression of P-GP and MRP2 in brain of thioacetamide-induced ALF rats. METHODS: ALF was induced by intraperitoneal injection of thioacetamide (300 mg/kg) for 2 days with a 24-h interval. The rats were used for experiments at 6, 12 and 24 h after the second administration. P-GP and MRP2 function in brain were determined using the brain-to-plasma ratios of corresponding substrates (rhodamine 123 and vincristine for P-GP; sulfobromophthalein and dinitrophenyl-S-glutathione for MRP2). Evans blue was used for examining the BBB integrity. Western blot was accomplished to determine P-GP and MRP2 protein expression. RESULTS: The brain-to-plasma ratios of rhodamine 123 and vincristine were significantly increased in ALF-6 h rats and almost returned to normal levels in ALF-24 h rats, whereas those of sulfobromophthalein and dinitrophenyl-S-glutathione were decreased in all ALF rats. Western blot results showed that ALF decreased brain P-GP levels at 6 and 12 h, whereas increased MRP2 levels at 6, 12 and 24 h. No significant difference of Evans blue concentrations in brain was found among the four groups. CONCLUSIONS: Function and expression of P-GP and MRP2 in brain of thioacetamide-induced ALF rats were oppositely altered.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Blood-Brain Barrier/metabolism , Brain/metabolism , Liver Failure, Acute/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Animals , Blotting, Western , Evans Blue , Injections, Intraperitoneal , Liver Failure, Acute/chemically induced , Multidrug Resistance-Associated Protein 2 , Permeability , Rats , Rhodamine 123/blood , Rhodamine 123/metabolism , Thioacetamide/administration & dosage , Thioacetamide/toxicity , Time Factors , Vincristine/blood , Vincristine/metabolism
5.
Acta Pharmacol Sin ; 33(11): 1372-8, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22842736

ABSTRACT

AIM: To characterize pharmacokinetic-pharmacodynamic modeling of diclofenac in Freund's complete adjuvant (FCA)-induced arthritic rats using prostaglandin E(2) (PGE(2)) as a biomarker. METHODS: The pharmacokinetics of diclofenac was investigated using 20-day-old arthritic rats. PGE(2) level in the rats was measured using an enzyme immunoassay. A pharmacokinetic-pharmacodynamic (PK-PD) model was developed to illustrate the relationship between the plasma concentration of diclofenac and the inhibition of PGE(2) production. The inhibition of diclofenac on lipopolysaccharide (LPS)-induced PGE(2) production in blood cells was investigated in vitro. RESULTS: Similar pharmacokinetic behavior of diclofenac was found both in normal and FCA-induced arthritic rats. Diclofenac significantly decreased the plasma levels of PGE(2) in both normal and arthritic rats. The inhibitory effect on PGE(2) levels in the plasma was in proportion to the plasma concentration of diclofenac. No delay in the onset of inhibition was observed, suggesting that the effect compartment was located in the central compartment. An inhibitory effect sigmoid I(max) model was selected to characterize the relationship between the plasma concentration of diclofenac and the inhibition of PGE(2) production in vivo. The I(max) model was also used to illustrate the inhibition of diclofenac on LPS-induced PGE(2) production in blood cells in vitro. CONCLUSION: Arthritis induced by FCA does not alter the pharmacokinetic behaviors of diclofenac in rats, but the pharmacodynamics of diclofenac is slightly affected. A PK-PD model characterizing an inhibitory effect sigmoid I(max) can be used to fit the relationship between the plasma PGE(2) and diclofenac levels in both normal rats and FCA-induced arthritic rats.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Arthritis, Experimental/drug therapy , Diclofenac/pharmacology , Models, Biological , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Arthritis, Experimental/physiopathology , Diclofenac/pharmacokinetics , Dinoprostone/blood , Disease Models, Animal , Freund's Adjuvant/toxicity , Immunoenzyme Techniques , Lipopolysaccharides/pharmacology , Male , Rats , Rats, Sprague-Dawley
6.
Acta Pharmacol Sin ; 33(6): 767-74, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22543702

ABSTRACT

AIM: To investigate the effects of nitric oxide (NO) donors on the function and expression of P-glycoprotein (P-gp) in Caco-2 cells. METHODS: Caco-2 cells were exposed to NO donors for designated times. P-gp function and expression were assessed using Rhodamine123 uptake assay and Western blotting, respectively. Intracellular reactive oxygen species (iROS) and intracellular reactive nitrogen species (iRNS) levels were measured using ROS and RNS assay kits, respectively. RESULTS: Exposure of Caco-2 cells to 0.1 or 2 mmol/L of sodium nitroprusside (SNP) affected the function and expression of P-gp in concentration- and time-dependent manners. A short-term (4 h) exposure reduced P-gp function and expression accompanied with significantly increased levels of iROS and iRNS. In contrast, a long-term (24 h) exposure stimulated the P-gp function and expression. The stimulatory effects of 2 mmol/L SNP was less profound as compared to those caused by 0.1 mmol/L SNP. The other NO donors SIN-1 and SNAP showed similar effects. Neither the NO scavenger PTIO (2 mmol/L) nor soluble guanylate cyclase inhibitor ODQ (50 µmol/L) reversed the SNP-induced alteration of P-gp function. On the other hand, free radical scavengers ascorbate, glutathione and uric acid (2 mmol/L for each), PKC inhibitor chelerythrine (5 µmol/L), PI3K/Akt inhibitor wortmannin (1 µmol/L) and p38 MAPK inhibitor SB203580 (10 µmol/L) reversed the upregulation of P-gp function by the long-term exposure to SNP, but these agents had no effect on the impaired P-gp function following the short-term exposure to SNP. CONCLUSION: NO donors time-dependently regulate P-gp function and expression in Caco-2 cells: short-term exposure impairs P-gp function and expression, whereas long-term exposure stimulates P-gp function and expression. The regulation occurs via a NO-independent mechanism.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Gene Expression Regulation/drug effects , Nitric Oxide Donors/pharmacology , Nitroprusside/pharmacology , Caco-2 Cells , Free Radical Scavengers/pharmacology , Humans , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...