Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters

Publication year range
1.
Am J Physiol Renal Physiol ; 327(1): F137-F145, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38779756

ABSTRACT

Polymyxins are a last-resort treatment option for multidrug-resistant gram-negative bacterial infections, but they are associated with nephrotoxicity. Gelofusine was previously shown to reduce polymyxin-associated kidney injury in an animal model. However, the mechanism(s) of renal protection has not been fully elucidated. Here, we report the use of a cell culture model to provide insights into the mechanisms of renal protection. Murine epithelial proximal tubular cells were exposed to polymyxin B. Cell viability, lactate dehydrogenase (LDH) release, polymyxin B uptake, mitochondrial superoxide production, nuclear morphology, and apoptosis activation were evaluated with or without concomitant gelofusine. A megalin knockout cell line was used as an uptake inhibition control. Methionine was included in selected experiments as an antioxidant control. A polymyxin B concentration-dependent reduction in cell viability was observed. Increased viability was observed in megalin knockout cells following comparable polymyxin B exposures. Compared with polymyxin B exposure alone, concomitant gelofusine significantly increased cell viability as well as reduced LDH release, polymyxin B uptake, mitochondrial superoxide, and apoptosis. Gelofusine and methionine were more effective at reducing renal cell injury in combination than either agent alone. In conclusion, the mechanisms of renal protection by gelofusine involve decreasing cellular drug uptake, reducing subsequent oxidative stress and apoptosis activation. These findings would be valuable for translational research into clinical strategies to attenuate drug-associated acute kidney injury.NEW & NOTEWORTHY Gelofusine is a gelatinous saline solution with the potential to attenuate polymyxin-associated nephrotoxicity. We demonstrated that the mechanisms of gelofusine renal protection involve reducing polymyxin B uptake by proximal tubule cells, limiting subsequent oxidative stress and apoptosis activation. In addition, gelofusine was more effective at reducing cellular injury than a known antioxidant control, methionine, and a megalin knockout cell line, indicating that gelofusine likely has additional pharmacological properties besides only megalin inhibition.


Subject(s)
Anti-Bacterial Agents , Apoptosis , Polymyxin B , Animals , Polymyxin B/pharmacology , Mice , Apoptosis/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/toxicity , Cell Survival/drug effects , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/pathology , Cell Line , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Acute Kidney Injury/prevention & control , Acute Kidney Injury/chemically induced , Oxidative Stress/drug effects , L-Lactate Dehydrogenase/metabolism
2.
Toxicol Mech Methods ; 34(5): 572-583, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38390772

ABSTRACT

Irinotecan-induced severe diarrhea (IISD) not only limits irinotecan's application but also significantly affects patients' quality of life. However, existing animal models often inadequately represent the dynamics of IISD development, progression, and resolution across multiple chemotherapy cycles, yielding non-reproducible and highly variable response with limited clinical translation. Our studies aim to establish a reproducible and validated IISD model that better mimics the pathophysiology progression observed in patients, enhancing translational potential. We investigated the impact of dosing regimens (including different dose, infusion time, and two cycles of irinotecan administration), sex, age, tumor-bearing conditions, and irinotecan formulation on the IISD incidence and severity in mice and rats. Lastly, we investigated above factors' impact on pharmacokinetics of irinotecan, intestinal injury, and carboxylesterase activities. In summary, we successfully established a standard model establishment procedure for an optimized IISD model with highly reproducible severe diarrhea incidence rate (100%) and a low mortality rate (11%) in F344 rats. Additionally, the rats tolerated at least two cycles of irinotecan chemotherapy treatment. In contrast, the mouse model exhibited suboptimal IISD incidence rates (60%) and an extremely high mortality rate (100%). Notably, dosing regimen, age and tumor-bearing conditions of animals emerged as critical factors in IISD model establishment. In conclusion, our rat IISD model proves superior in mimicking pathophysiology progression and characteristics of IISD in patients, which stands as an effective tool for mechanism and efficacy studies in future chemotherapy-induced gut toxicity research.


Subject(s)
Diarrhea , Disease Models, Animal , Irinotecan , Rats, Inbred F344 , Irinotecan/toxicity , Animals , Diarrhea/chemically induced , Male , Female , Mice , Rats , Severity of Illness Index , Dose-Response Relationship, Drug , Humans , Reproducibility of Results
3.
J Nat Prod ; 84(9): 2486-2495, 2021 09 24.
Article in English | MEDLINE | ID: mdl-34463097

ABSTRACT

This study aims to characterize the pharmacokinetic (PK) profiles and identify important bioavailability barriers and pharmacological pathways of the key active components (KACs) of Antitumor B (ATB), a chemopreventive agent. KACs (matrine, dictamine, fraxinellone, and maackiain) of ATB were confirmed using the antiproliferative assay and COX-2 inhibition activities in oral cancer cells. The observed in vitro activities of KACs were consistent with their cell signaling pathways predicted using the in silico network pharmacology approach. The pharmacokinetics of KACs were determined after i.v., i.p., and p.o. delivery using ATB extract and a mixture of four KACs in mice. Despite good solubilities and permeabilities, poor oral bioavailabilities were estimated for all KACs, mostly because of first-pass metabolism in the liver (for all KACs) and intestines (for matrine and fraxinellone). Multiple-dose PK studies showed 23.2-fold and 8.5-fold accumulation of dictamine and maackiain in the blood, respectively. Moreover, saliva levels of dictamine and matrine were found significantly higher than their blood levels. In conclusion, the systemic bioavailabilities of ATB-KACs were low, but significant levels of dictamine and matrine were found in saliva upon repeated oral administration. Significant salivary concentrations of matrine justified its possible use as a drug-monitoring tool to track patient compliance during chemoprevention trials.


Subject(s)
Biological Availability , Drugs, Chinese Herbal/pharmacokinetics , Mouth Neoplasms/prevention & control , Alkaloids/pharmacokinetics , Animals , Benzofurans/pharmacokinetics , Chemoprevention , Mice , Mice, Inbred C57BL , Molecular Structure , Network Pharmacology , Pterocarpans/pharmacokinetics , Quinolines/pharmacokinetics , Quinolizines/pharmacokinetics , Matrines
4.
Inflamm Res ; 69(1): 131-137, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31797003

ABSTRACT

OBJECTIVE: This study sought to evaluate short-term treatment with COX-2 inhibitors and acute changes in colonic PGE2 levels as predictors of long-term efficacy in a genetic model of colorectal cancer. METHODS: Celecoxib oral suspension (40 mg/kg BID) was dosed to Apc-mutant Pirc (F344/NTac-Apcam1137) rats for 4 days (short-term group), or the equivalent dose of 1500 ppm celecoxib was administered in the diet for 4 months (long-term group). Percent inhibition of colonic PGE2 was calculated, and the reduction in colonic PGE2 was assessed in relation to suppression of adenomatous colon polyps. RESULTS: Colonic mucosa PGE2 was fourfold higher in Pirc than in F344 wild-type rats (21 vs. 5.6 pg/mg epithelial tissue), due at least in part to higher COX-2 expression, and this was confirmed by elevated PGE2-d11 levels in Pirc colonic S9 incubations. In the 4-day study, dose-dependent reductions in PGE2 were observed in colonic epithelium (-33% (P>0.05) and -57% (P=0.0012)), after low- and high-dose celecoxib treatments of 4 mg/kg and 40 mg/kg (bid), respectively. In the 4-month study, 1500 ppm celecoxib suppressed colonic epithelium PGE2 by 43.5%, and tumor multiplicity by 80% (P<0.0015). Suppression of plasma 6-keto PGF1α also was corroborated following long-term treatment with 1500 ppm celecoxib (P<0.05). CONCLUSIONS: Acute changes in colonic mucosa PGE2 provided a rapid means of predicting long-term chemopreventive effects from celecoxib, and might be useful for screening of new COX-2 inhibitor compounds.


Subject(s)
Celecoxib/pharmacology , Colon/drug effects , Colorectal Neoplasms/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Dinoprostone/metabolism , Animals , Biomarkers/metabolism , Celecoxib/therapeutic use , Colon/metabolism , Colorectal Neoplasms/drug therapy , Cyclooxygenase 2 Inhibitors/therapeutic use , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Male , Rats, Inbred F344 , Treatment Outcome
5.
J Sep Sci ; 43(24): 4414-4423, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33119204

ABSTRACT

The aim of this study is to establish a reliable liquid chromatography-mass spectrometry method to simultaneously quantitate raloxifene, and its major metabolites, raloxifene-6-glucuronide, raloxifene-4'-glucuronide, and raloxifene-6-sulfate in rat plasma samples for pharmacokinetic studies. The separation of the analytes was achieved on a Waters BEH C18 column. Water (0.1% formic acid) and acetonitrile were used as the mobile phases for elution. A one-step protein precipitation using a mixture solvent was applied for plasma sample preparation. The method was validated following the FDA guidance. The results showed that the linear range were 1.95-1000 nM for raloxifene-6-glucuronide, and raloxifene-4'-glucuronide, 0.195-100 nM for raloxifene-6-sulfate, and 0.195-200 nM for raloxifene, respectively. The lower limit of quantification was 1.95, 1.95, 0.195, and 0.195 nM for raloxifene-6-glucuronide, raloxifene-4'-glucuronide, raloxifene-6-sulfate, and raloxifene, respectively. Only 20 µl of plasma sample was required since the method is sensitive. The intra- and interday variance is <15% and the accuracy is within 85-115%. The variance of matrix effect and recovery were <15%. The method was successfully applied in a pharmacokinetic study in rats with oral administration of raloxifene.


Subject(s)
Raloxifene Hydrochloride , Animals , Chromatography, High Pressure Liquid , Female , Male , Mass Spectrometry , Mice , Mice, Inbred C57BL , Molecular Structure , Raloxifene Hydrochloride/blood , Raloxifene Hydrochloride/metabolism , Raloxifene Hydrochloride/pharmacokinetics , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley
6.
Arch Toxicol ; 94(11): 3737-3749, 2020 11.
Article in English | MEDLINE | ID: mdl-32918091

ABSTRACT

Icaritin (ICT), a prenylflavonoid derivative extracted from the Epimedium genus, has exhibited antitumor effects in hepatocellular carcinoma (HCC) cells and safety and tolerance in clinical settings. However, ICT exhibits low blood concentration and the in vivo dominant plasma species of ICT is glucuronides [icaritin-3-glucuronide (G1), icaritin-7-glucuronide (G2) and icaritin-3, 7-diglucuronide (DIG)]. Therefore, how ICT reaches the liver and exerts its effect with low toxicity remains unknown. Therefore, pharmacokinetic experiments (p.o. 5 mg/kg with/out 50 mg/kg inhibitor combo), intestinal perfusion (2 µM ICT), portal vein infusion (1.6 µM ICT, 7.1 µM G1, 6.8 µM G2 and 4.4 µM DIG), and in vitro studies (the concentration range of substrates: 0.3-10 µM) were conducted in the present study. Ultimately, ICT was shown to undergo glucuronidation by the intestine and subsequent uptake by hepatocytes via organic anion transporting peptides (OATPs) as conjugates, followed by biliary excretion mainly as diglucuronide. In conclusion, we found for the first time that the intestine is considered as the major metabolic organ, liver as the main recycling organ for the enterohepatic recycling (EHR) of ICT. Moreover, DIG is the main species in the systemic circulation following oral administration of ICT which explains the low toxicity of ICT in clinical settings.


Subject(s)
Flavonoids/metabolism , Flavonoids/pharmacokinetics , Glucuronides/metabolism , Glucuronides/pharmacokinetics , Animals , Caco-2 Cells , Chromatography, High Pressure Liquid , Enterohepatic Circulation , HEK293 Cells , Hepatobiliary Elimination , Hepatocytes/metabolism , Humans , Intestines , Liver/metabolism , Male , Microsomes, Liver/metabolism , Organic Anion Transporters/metabolism , Perfusion/methods , Portal Vein/metabolism , Rats , Rats, Wistar , Tandem Mass Spectrometry
7.
Molecules ; 25(18)2020 Sep 04.
Article in English | MEDLINE | ID: mdl-32899627

ABSTRACT

Cyclooxygenase-2 (COX-2) imaging agents are potent tools for early cancer diagnosis. Almost all of the COX2 imaging agents using celecoxib as backbone were chemically modified in the position of N-atom in the sulfonamide group. Herein, a novel COX-2 probe (CCY-5) with high targeting ability and a near-infrared wavelength (achieved by attaching a CY-5 dye on the pyrazole ring of celecoxib using a migration strategy) was evaluated for its ability to probe COX-2 in human cancer cells. CCY-5 is expected to have high binding affinity for COX-2 based on molecular docking and enzyme inhibition assay. Meanwhile, CCY-5 caused stronger fluorescence imaging of COX-2 overexpressing cancer cells (Hela and SCC-9 cells) than that of normal cell lines (RAW 264.7 cells). Lipopolysaccharide (LPS) treated RAW264.7 cells revealed an enhanced fluorescence as LPS was known to induce COX-2 in these cells. In inhibitory studies, a markedly reduced fluorescence intensity was observed in cancer cells, when they were co-treated with a COX-2 inhibitor celecoxib. Therefore, CCY-5 may be a selective bioimaging agent for cancer cells overexpressing COX-2 and could be useful as a good monitoring candidate for effective diagnosis and therapy in cancer treatment.


Subject(s)
Celecoxib/pharmacology , Cyclooxygenase 2/metabolism , Fluorescent Dyes/chemical synthesis , Infrared Rays , Neoplasms/diagnosis , Animals , Celecoxib/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Cyclooxygenase 2 Inhibitors/pharmacology , Fluorescence , Humans , Lipopolysaccharides/pharmacology , Mice , Molecular Docking Simulation , Neoplasms/pathology , RAW 264.7 Cells
8.
Mol Pharm ; 14(9): 2884-2898, 2017 09 05.
Article in English | MEDLINE | ID: mdl-28221813

ABSTRACT

Glucuronide metabolites require the action of efflux transporters to exit cells due to their hydrophilic properties. In this study, we proposed a transport-glucuronidation classification system and developed a PBPK model to predict the impact of BCRP on systemic exposure of glucuronides. The clearance by UGTs in S9 fractions and the efflux clearance of glucuronides by BCRP in human UGT1A9-overexpressing HeLa cells were incorporated in the classification system and PBPK model. Based on simulations for glucuronide AUC for theoretical compounds in the classification system, it was indicated that BCRP was more important for compounds with greater efflux clearance of their glucuronides by BCRP regardless of differences in clearance by UGTs. Pharmacokinetic studies were performed in WT and Bcrp1 (-/-) mice for 8 compounds to verify our predictions. Among eight compounds, the glucuronide AUC of daidzein and genistein increased significantly in Bcrp1 (-/-) mice, while only slight increases in systemic exposure were observed for other glucuronides. The results from pharmacokinetic studies were in agreement with the predictions except for resveratrol, which was effluxed predominantly by transporters other than BCRP. Therefore, for glucuronides that were predominantly mediated by BCRP, this study provided a useful approach in predicting the impact of BCRP on its disposition and the potential DDIs involving BCRP.


Subject(s)
Glucuronides/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Animals , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , HeLa Cells , Humans , Kinetics , Liver/metabolism , Male , Mice , Mice, Knockout , Resveratrol , Stilbenes/metabolism
9.
Pharm Res ; 33(3): 590-602, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26502886

ABSTRACT

PURPOSE: To evaluate the impact of curcumin on the disposition of resveratrol phase II metabolites in vivo, and explain the observations by performing in vitro studies in transporter-overexpressed cells. METHODS: Pharmacokinetic studies of resveratrol with and without the co-administration of curcumin were performed in both FVB wild-type and Bcrp1 (-/-) mice. Human UGT1A9-overexpressing HeLa cells and human MRP2-overexpressing MDCK II-UGT1A1 cells were used as in vitro tools to further determine the impact of curcumin as a transporter inhibitor on resveratrol metabolites. RESULTS: We observed higher exposure of resveratrol conjugates in Bcrp1 (-/-) mice compared to wild-type mice. In wild-type mice, curcumin increased the AUC of resveratrol glucuronide by 4-fold compared to the mice treated without curcumin. The plasma levels of resveratrol and its sulfate conjugate also increased moderately. In Bcrp1 (-/-) mice, there was a further increase (6-fold increase) in AUC of resveratrol glucuronide observed when curcumin was co-administered compared to AUC values obtained in wild-type mice without curcumin treatment. In the presence of 50 nM curcumin, the clearance of resveratrol-3-O-glucuronide and resveratrol-3-O-sulfate reduced in both MRP2-overexpressing MDCKII-UGT1A1 cells and Human UGT1A9-overexpressing HeLa cells. CONCLUSIONS: These results suggest that curcumin alters the phase II distribution of resveratrol through inhibiting efflux transporters including MRP2 and BCRP.


Subject(s)
ATP-Binding Cassette Transporters/antagonists & inhibitors , Curcumin/pharmacology , Metabolic Detoxication, Phase II/physiology , Multidrug Resistance-Associated Proteins/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Stilbenes/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/metabolism , Animals , Cell Line , Cell Line, Transformed , Dogs , Glucuronides/metabolism , Glucuronosyltransferase/metabolism , HeLa Cells , Humans , Madin Darby Canine Kidney Cells , Male , Membrane Transport Proteins , Mice , Mice, Knockout , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/metabolism , Neoplasm Proteins/metabolism , Resveratrol , UDP-Glucuronosyltransferase 1A9
10.
Molecules ; 20(12): 21254-73, 2015 Nov 30.
Article in English | MEDLINE | ID: mdl-26633325

ABSTRACT

Mango seed kernel extract (MSKE) and its key components (gallic acid, GA; methyl gallate, MG; and pentagalloyl glucopyranose, PGG) have generated interest because of their pharmacological activities. To develop the potential use of the key components in MSKE as natural therapeutic agents, their pharmacokinetic data are necessary. Therefore, this study was performed to evaluate the factors affecting their oral bioavailability as pure compounds and as components in MSKE. The in vitro chemical stability, biological stability, and absorption were evaluated in Hanks' Balanced Salt Solution, Caco-2 cell and rat fecal lysates, and the Caco-2 cell model, respectively. The in vivo oral pharmacokinetic behavior was elucidated in Sprague-Dawley rats. The key components were unstable under alkaline conditions and in Caco-2 cell lysates or rat fecal lysates. The absorptive permeability coefficient followed the order MG > GA > PGG. The in vivo results exhibited similar pharmacokinetic trends to the in vitro studies. Additionally, the co-components in MSKE may affect the pharmacokinetic behaviors of the key components in MSKE. In conclusion, chemical degradation under alkaline conditions, biological degradation by intestinal cell and colonic microflora enzymes, and low absorptive permeability could be important factors underlying the oral bioavailability of these polyphenols.


Subject(s)
Gallic Acid/analogs & derivatives , Gallic Acid/metabolism , Hydrolyzable Tannins/metabolism , Mangifera/chemistry , Plant Extracts/administration & dosage , Seeds/chemistry , Animals , Caco-2 Cells , Feces/chemistry , Humans , Intestinal Absorption , Male , Molecular Structure , Rats , Rats, Sprague-Dawley
11.
Antimicrob Agents Chemother ; 58(7): 4200-2, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24733472

ABSTRACT

Polymyxin B is increasingly used as a treatment of last resort against multidrug-resistant Gram-negative infections. Using a mammalian kidney cell line, we demonstrated that polymyxin B uptake into proximal tubular epithelial cells was saturable and occurred primarily through the apical membrane, suggesting the involvement of transporters in the renal uptake of polymyxin B. Megalin might play a role in the uptake and accumulation of polymyxin B into renal cells.


Subject(s)
Anti-Bacterial Agents/metabolism , Epithelial Cells/metabolism , Kidney Tubules, Proximal/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Polymyxin B/metabolism , Animals , Anti-Bacterial Agents/therapeutic use , Biological Transport, Active , Cell Line , Drug Resistance, Multiple, Bacterial , Epithelial Cells/cytology , Gram-Negative Bacterial Infections/drug therapy , Gram-Negative Bacterial Infections/microbiology , Kidney Tubules, Proximal/cytology , Polymyxin B/therapeutic use , Swine
12.
Antimicrob Agents Chemother ; 58(5): 2740-6, 2014 May.
Article in English | MEDLINE | ID: mdl-24566187

ABSTRACT

Despite concerns of nephrotoxicity, polymyxin antibiotics often remain the only susceptible agents for multidrug-resistant (MDR) Gram-negative bacteria. Colistin has been more commonly used clinically due to a perceived safety benefit. We compared the nephrotoxicity of colistin to polymyxin B. The in vitro cytotoxicity of colistin was compared to polymyxin B in two mammalian renal cell lines. To validate the clinical relevance of the findings, we evaluated adult patients with normal renal function who received a minimum of 72 h of polymyxin therapy in a multicenter study. The primary outcome was the prevalence of nephrotoxicity, as defined by the RIFLE (risk, injury, failure, loss, end-stage kidney disease) criteria. Colistin exhibited an in vitro cytotoxicity profile similar to polymyxin B. A total of 225 patients (121 receiving colistimethate, 104 receiving polymyxin B) were evaluated. Independent risk factors for colistimethate-associated nephrotoxicity included age (odds ratio [OR], 1.04; 95% confidence interval [CI], 1.00 to 1.07; P = 0.03), duration of therapy (OR 1.08; 95% CI, 1.02 to 1.15; P = 0.02), and daily dose by ideal body weight (OR 1.40; 95% CI, 1.05 to 1.88; P = 0.02). In contrast, cystic fibrosis was found to be a protective factor in patients who received colistimethate (OR, 0.03; 95% CI, 0.001 to 0.79; P = 0.04). In a matched analysis based on the risk factors identified (n = 76), the prevalence of nephrotoxicity was higher with colistimethate than with polymyxin B (55.3% versus 21.1%; P = 0.004). Polymyxin B was not found to be more nephrotoxic than colistin and may be the preferred polymyxin for MDR infections. A prospective study comparing the two polymyxins directly is warranted.


Subject(s)
Anti-Bacterial Agents/adverse effects , Colistin/analogs & derivatives , Polymyxins/adverse effects , Adult , Anti-Bacterial Agents/administration & dosage , Cell Line , Cell Survival/drug effects , Colistin/administration & dosage , Colistin/adverse effects , Humans , Kidney/drug effects , Middle Aged , Polymyxins/administration & dosage , Retrospective Studies
13.
Mol Pharm ; 10(5): 1736-50, 2013 May 06.
Article in English | MEDLINE | ID: mdl-23402418

ABSTRACT

Cellular production of flavonoid glucuronides requires the action of both UDP-glucuronosyltransferases (UGT) and efflux transporters since glucuronides are too hydrophilic to diffuse across the cellular membrane. We determined the kinetics of efflux of 13 flavonoid glucuronides using the newly developed HeLa-UGT1A9 cells and correlated them with kinetic parameters derived using expressed UGT1A9. The results indicated that, among the seven monohydroxylflavones (HFs), there was moderately good correlation (r(2) ≥ 0.65) between the fraction metabolized (fmet) derived from HeLa-UGT1A9 cells and CLint derived from the UGT1A9-mediated metabolism. However, there was weak or no correlation between these two parameters for six dihydroxylflavones (DHFs). Furthermore, there was weak or no correlation between various kinetic parameters (Km, Vmax, or CLint) for the efflux and the metabolism regardless of whether we were using seven HFs, six DHFs, or a combination thereof. Instead, the cellular excretion of many flavonoid glucuronides appears to be controlled by the efflux transporter, and the poor affinity of glucuronide to the efflux transporter resulted in major intracellular accumulation of glucuronides to a level that is above the dosing concentration of its aglycone. Hence, the efflux transporters appear to act as the "Revolving Door" to control the cellular excretion of glucuronides. In conclusion, the determination of a flavonoid's susceptibility to glucuronidation must be based on both its susceptibility to glucuronidation by the enzyme and resulting glucuronide's affinity to the relevant efflux transporters, which act as the "Revolving Door(s)" to facilitate or control its removal from the cells.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Glucuronides/metabolism , Glucuronosyltransferase/metabolism , Neoplasm Proteins/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Biological Transport, Active , Flavones/chemistry , Flavones/metabolism , Glucuronides/chemistry , Glucuronosyltransferase/genetics , HeLa Cells , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Kinetics , Models, Biological , Molecular Structure , UDP-Glucuronosyltransferase 1A9
14.
Pharm Res ; 30(3): 836-46, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23254888

ABSTRACT

PURPOSE: To investigate the ability of bacteria in the intestinal microbiome to convert naturally occurring primary ginsenosides in red ginseng extract to active secondary ginsenosides. METHODS: Anti-proliferative ginsenoside activity was tested using mouse lung cancer LM1 cells. Permeabilities were evaluated in Caco-2 cell monolayers. Systemic exposure of secondary ginsenosides was determined in A/J mice. 16S rRNA gene pyrosequencing was used to determine membership and abundance of bacteria in intestinal microbiome. RESULTS: Secondary ginsenoside C-K exhibited higher anti-proliferative activity and permeability than primary ginsenosides. Significant amounts of secondary ginsenosides (F2 and C-K) were found in blood of A/J mice following oral administration of primary ginsenoside Rb1. Because mammalian cells did not hydrolyze ginsenoside, we determined the ability of bacteria to hydrolyze ginsenosides and found that Rb1 underwent stepwise hydrolysis to Rd, F2, and then C-K. Formation of F2 from Rd was the rate-limiting step in the biotransformation of Rb1 to C-K. CONCLUSION: Conversion to F2 is the rate-limiting step in bioactivation of primary ginsenosides by A/J mouse intestinal microbiome, whose characterization reveals the presence of certain bacterial families capable of enabling the formation of F2 and C-K in vivo.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/pharmacokinetics , Bacteria/enzymology , Ginsenosides/pharmacology , Ginsenosides/pharmacokinetics , Glycoside Hydrolases/metabolism , Intestines/microbiology , Animals , Antineoplastic Agents, Phytogenic/metabolism , Bacteria/genetics , Bacteria/isolation & purification , Biological Availability , Biotransformation , Caco-2 Cells , Cell Line, Tumor , Cell Proliferation/drug effects , Drugs, Chinese Herbal/metabolism , Drugs, Chinese Herbal/pharmacokinetics , Drugs, Chinese Herbal/pharmacology , Ginsenosides/metabolism , Humans , Hydrolysis , Intestinal Mucosa/metabolism , Male , Metagenome , Mice , Neoplasms/drug therapy , Panax/chemistry , RNA, Bacterial/genetics
15.
Antimicrob Agents Chemother ; 56(9): 4625-9, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22687519

ABSTRACT

The increasing prevalence of multidrug-resistant Gram-negative infections has led to renewed interest in the use of systemic polymyxin B. However, the nephrotoxic properties of polymyxin B are still poorly understood. The objective of this study was to characterize nephrotoxicity associated with polymyxin B, with an emphasis on examining the impact of dosing frequencies on the onset of nephrotoxicity. Sprague-Dawley rats were divided into two groups and administered the same total daily dose of polymyxin B subcutaneously but with different dosing frequencies (either 20 mg/kg of body weight every 24 h [q24h] or 5 mg/kg q6h). Drug concentrations in renal tissue were compared between the two groups at 24 h. Kidney tissues were harvested at 48 h and compared histologically. Serum creatinine was measured daily for up to 10 days, and nephrotoxicity was defined as a significant elevation in serum creatinine (≥2× baseline). Kaplan-Meier analysis was used to compare the onset of nephrotoxicity. Polymyxin B-induced nephrotoxicity manifested as elevation in serum creatinine and acute tubular necrosis. Extensive injury of the proximal tubules was observed. The lesions were more severe and higher drug concentrations were achieved in the kidneys of the q6h dosing group. The q24h dosing group experienced a more gradual onset of nephrotoxicity, which could be attributed to the lower kidney tissue drug concentrations (48.5 ± 17.4 µg/g versus 92.1 ± 18.1 µg/g of polymyxin B1, P = 0.04). Preferential accumulation of polymyxin B in the kidneys suggests that uptake to renal cells is a nonpassive process and q24h dosing was less nephrotoxic than q6h dosing.


Subject(s)
Anti-Bacterial Agents/adverse effects , Kidney Tubular Necrosis, Acute/pathology , Kidney Tubules, Proximal/drug effects , Polymyxin B/adverse effects , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacokinetics , Cell Line , Cell Survival/drug effects , Creatinine/blood , Dogs , Drug Dosage Calculations , Female , Humans , Inhibitory Concentration 50 , Injections, Subcutaneous , Kaplan-Meier Estimate , Kidney Tubular Necrosis, Acute/blood , Kidney Tubular Necrosis, Acute/etiology , Kidney Tubules, Proximal/pathology , Polymyxin B/administration & dosage , Polymyxin B/pharmacokinetics , Rats , Rats, Sprague-Dawley , Time Factors
16.
Drug Metab Dispos ; 40(10): 1883-93, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22736306

ABSTRACT

It was recently proposed that the improved oral bioavailability of genistein aglycone and conjugates in Bcrp1(-/-) mice is mainly due to increased intestinal absorption of aglycone and subsequent elevated exposure to conjugation enzymes. Here we tested this proposed mechanism and found that intestinal absorption of genistein aglycone did not increase in Bcrp1(-/-) mice compared with wild-type mice using an in situ mouse intestinal perfusion model and that inhibition of breast cancer resistance protein (BCRP) in Caco-2 cells also did not significantly increase permeability or intracellular concentration of aglycone. Separately, we showed that 5- to 10-fold increases in exposures of conjugates and somewhat lower fold increases (<2-fold) in exposures of aglycone were apparent after both oral and intraperitoneal administration in Bcrp1(-/-) mice. In contrast, the intestinal and biliary excretion of genistein conjugates significantly decreased in Bcrp1(-/-) mice without corresponding changes in aglycone excretion. Likewise, inhibition of BCRP functions in Caco-2 cells altered polarized excretion of genistein conjugates by increasing their basolateral excretion. We further found that genistein glucuronides could be hydrolyzed back to genistein, whereas sulfates were stable in blood. Because genistein glucuronidation rates were 110% (liver) and 50% (colon) higher and genistein sulfation rates were 40% (liver) and 42% (colon) lower in Bcrp1(-/-) mice, the changes in genistein exposures are not mainly due to changes in enzyme activities. In conclusion, improved bioavailability of genistein and increased plasma area under the curve of its conjugates in Bcrp1(-/-) mice is due to altered distribution of genistein conjugates to the systemic circulation.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Genistein/pharmacokinetics , Intestinal Mucosa/metabolism , Neoplasm Proteins/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/deficiency , ATP-Binding Cassette Transporters/genetics , Adenosine/analogs & derivatives , Adenosine/pharmacology , Administration, Oral , Animals , Area Under Curve , Bile/metabolism , Biological Availability , Caco-2 Cells , Diketopiperazines , Genistein/administration & dosage , Genistein/blood , Glucuronides/metabolism , Heterocyclic Compounds, 4 or More Rings , Humans , Hydrolysis , Injections, Intraperitoneal , Intestines/drug effects , Male , Metabolic Detoxication, Phase II , Methotrexate/metabolism , Mice , Mice, Knockout , Neoplasm Proteins/antagonists & inhibitors , Perfusion , Permeability , Sulfates/metabolism
17.
Drug Metab Dispos ; 40(8): 1538-44, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22584255

ABSTRACT

Ginsenosides are hydrolyzed extensively by gut microflora after oral administration, and their metabolites are pharmacologically active against lung cancer cells. In this study, we measured the metabolism of various ginsenosides by gut microflora and determined the mechanisms responsible for the observed pharmacokinetic behaviors of its active metabolite, Compound K (C-K). The results showed that biotransformation into C-K is the major metabolic pathway of ginsenosides after the oral administration of the red ginseng extract containing both protopanaxadiol and protopanaxatriol ginsenosides. Pharmacokinetic studies in normal mice showed that C-K exhibited low oral bioavailability. To define the mechanisms responsible for this low bioavailability, two P-glycoprotein (P-gp) inhibitors, verapamil and cyclosporine A, were used, and their presence substantially decreased C-K's efflux ratio in Caco-2 cells (from 26.6 to <3) and significantly increased intracellular concentrations (by as much as 40-fold). Similar results were obtained when transcellular transport of C-K was determined using multidrug resistance 1 (MDR1)-overexpressing Madin-Darby canine kidney II cells. In MDR1a/b(-/-) FVB mice, its plasma C(max) and AUC(0-24h) were increased substantially by 4.0- and 11.7-fold, respectively. These increases appear to be due to slower elimination and faster absorption of C-K in MDR1a/b(-/-) mice. In conclusion, C-K is the major active metabolite of ginsenosides after microflora hydrolysis of primary ginsenosides in the red ginseng extract, and inhibition/deficiency of P-gp can lead to large enhancement of its absorption and bioavailability.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Ginsenosides/pharmacokinetics , Intestines/microbiology , Panax/chemistry , Plant Extracts/pharmacology , Administration, Oral , Animals , Biological Availability , Caco-2 Cells , Ginsenosides/administration & dosage , Humans , Mice
18.
Mol Pharm ; 9(4): 862-73, 2012 Apr 02.
Article in English | MEDLINE | ID: mdl-22352375

ABSTRACT

Flavonoids are polyphenolic compounds with various claimed health benefits, but the extensive metabolism by uridine-5'-diphospho-glucuronosyltransferases (UGTs) and sulfotransferases (SULTs) in liver and intestine led to poor oral bioavailabilities. The effects of structural changes on the sulfonation of flavonoids have not been systemically determined, although relevant effects of structural changes on the glucuronidation of flavonoids had. We performed the regiospecific sulfonation of sixteen flavonoids from five different subclasses of flavonoids, which are represented by apigenin (flavone), genistein (isoflavone), naringenin (flavanone), kaempherol (flavonol), and phloretin (chalcone). Additional studies were performed using 4 monohydroxyl flavonoids with a -OH group at the 3, 4', 5 or 7 position, followed by 5 dihydroxyl flavonoids, and 2 trihydroxyl flavonoids by using expressed human SULT1A3 and Caco-2 cell lysates. We found that these compounds were exclusively sulfated at the 7-OH position by SULT1A3 and primarily sulfated at the 7-OH position in Caco-2 cell lysates with minor amounts of 4'-O-sulfates formed as well. Sulfonation rates measured using SULT1A3 and Caco-2 cell lysates were highly correlated at substrate concentrations of 2.5 and 10 µM. Molecular docking studies provided structural explanations as to why sulfonation only occurred at the 7-OH position of flavones, flavonols and flavanones. In conclusion, molecular docking studies explain why SULT1A3 exclusively mediates sulfonation at the 7-OH position of flavones/flavonols, and correlation studies indicate that SULT1A3 is the main isoform responsible for flavonoid sulfonation in the Caco-2 cells.


Subject(s)
Flavonoids/metabolism , Sulfotransferases/metabolism , Apigenin/chemistry , Apigenin/metabolism , Arylsulfotransferase , Binding Sites , Caco-2 Cells , Flavanones/chemistry , Flavanones/metabolism , Flavonoids/chemistry , Genistein/chemistry , Genistein/metabolism , Humans , Kaempferols/chemistry , Kaempferols/metabolism , Kinetics , Phloretin/chemistry , Phloretin/metabolism , Sulfotransferases/chemistry , Tandem Mass Spectrometry
19.
Pharm Res ; 29(11): 3199-208, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22752253

ABSTRACT

PURPOSE: The efflux transporter breast cancer resistance protein (BCRP/ABCG2) plays an important role in excretion of anionic drugs and metabolites including glucuronides in humans. METHODS: In this article, our recently published cell model (i.e., HeLa cells over-expressing UGT1A9 (HeLa1A9)) is used to determine the kinetic parameters of BCRP-mediated transport of glucuronides. RESULTS: After incubation of the aglycone with the cells, a steady-state (i.e., zero-order or near zero-order) excretion of its glucuronide is rapidly achieved and then maintained. Kinetic profiling with different (intracellular) glucuronide concentrations and their corresponding excretion rates is enabled by varying the concentration of the aglycone, which allows for the determination of kinetic parameters responsible for BCRP-mediated efflux of glucuronides. This approach was validated theoretically using a cellular pharmacokinetic model incorporating various enzymatic and transporter-mediated kinetic processes. It was also validated experimentally in that kinetic parameters of efflux of glucuronides of 6-hydroxyflavone and 4-methylumberiferone in the HeLa1A9 cell model were shown to be consistent with those derived with BCRP-overexpressing membrane vesicles. CONCLUSION: This study provides a new strategy for rapidly evaluating the kinetics of glucuronide efflux by BCRP.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Glucuronides/pharmacokinetics , Neoplasm Proteins/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Biological Transport , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Flavonoids/pharmacokinetics , HeLa Cells , Humans
20.
J Inflamm Res ; 15: 4435-4447, 2022.
Article in English | MEDLINE | ID: mdl-35958187

ABSTRACT

Objective: We aim to quantify the absolute protein expression of cyclooxygenase-1 (COX-1) and cyclooxygenase-2 (COX-2) in various cells and tissues to determine the relative contribution of COX-1 and COX-2 to PGE2 production. Methods: An LC-MS method was developed and validated, then used for quantifying the absolute amounts of COX-1 and COX-2 in recombinant human COX-1 and COX-2, lysates from different cells, tissue microsomes of rodents and humans, Pirc rat colonic polyps, and biopsy specimens from squamous cell carcinoma (SCC) patients. The COX-1 and COX-2 turnover numbers were subsequently calculated based on apparent formation rates of PGE2. Results: A robust LC-MS method for quantification of COX-1 and COX-2 was developed and validated and then used to calculate their apparent turnover numbers. The results showed that COX-1 expression levels were much higher than that of COX-2 in all the tested tissues including the colonic epithelium of F344 (28-fold) and Pirc rats (20-fold), colonic polyps of Pirc rats (8-fold), and biopsy specimens of SCC patients (11-17-fold). In addition, both COX-1 and COX-2 were higher in polyps when compared to adjacent mucosa of Pirc rats. The turnover number of recombinant human COX-2 was 14-fold higher than that of recombinant human COX-1. LPS stimulation increased COX-2 protein expression in three cell lines (Raw 264.7, SCC9 and EOMA) as expected but unexpectedly increased COX-1 protein expression (13.8-fold) in EOMA cells. Conclusion: In human oral cancer tissues and cells as well as Pirc rat colon, COX-1 plays an unexpectedly but more important role than COX-2 in abnormal PGE2 production since COX-1 expression is much higher than COX-2. In addition, COX-1 expression levels are inducible in cells, and higher in polyps than surrounding mucosa in Pirc rat colon. These results indicate that targeted suppression of local COX-1 should be considered to reduce colon-specific PGE2-mediated inflammation.

SELECTION OF CITATIONS
SEARCH DETAIL