1
|
Differential Uridyl-diphosphate-Glucuronosyl Transferase 1A enzymatic arsenal explains the specific cytotoxicity of resveratrol towards tumor colorectal cells. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
2
|
Hornedo-Ortega R, Jourdes M, Da Costa G, Courtois A, Gabaston J, Teissedre PL, Richard T, Krisa S. Oxyresveratrol and Gnetol Glucuronide Metabolites: Chemical Production, Structural Identification, Metabolism by Human and Rat Liver Fractions, and In Vitro Anti-inflammatory Properties. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:13082-13092. [PMID: 35195403 PMCID: PMC9585577 DOI: 10.1021/acs.jafc.1c07831] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Stilbene metabolites are attracting great interest because many of them exhibit similar or even stronger biological effects than their parent compounds. Furthermore, the metabolized forms are predominant in biological fluids; therefore, their study is highly relevant. After hemisynthesis production, isolation, and structural elucidation, three glucuronide metabolites for oxyresveratrol (ORV) were formed: trans-ORV-4'-O-glucuronide, trans-ORV-3-O-glucuronide, and trans-ORV-2'-O-glucuronide. In addition, two glucuronide metabolites were obtained for gnetol (GN): trans-GN-2'-O-glucuronide and trans-GN-3-O-glucuronide. When the metabolism of ORV and GN is studied in vitro by human and rat hepatic enzymes, four of the five hemisynthesized compounds were identified and quantified. Human enzymes glucuronidated preferably at the C-2' position, whereas rat enzymes do so at the C-3 position. In view of these kinetic findings, rat enzymes have a stronger metabolic capacity than human enzymes. Finally, ORV, GN, and their glucuronide metabolites (mainly at the C-3 position) decreased nitric oxide, reactive oxygen species, interleukin 1β, and tumor necrosis factor α production in lipopolysaccharide-stimulated macrophages.
Collapse
|
3
|
Zhou J, Argikar UA, Miners JO. Enzyme Kinetics of Uridine Diphosphate Glucuronosyltransferases (UGTs). Methods Mol Biol 2021; 2342:301-338. [PMID: 34272700 DOI: 10.1007/978-1-0716-1554-6_12] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Glucuronidation, catalyzed by uridine diphosphate glucuronosyltransferases (UGTs), is an important process for the metabolism and clearance of many lipophilic chemicals, including drugs, environmental chemicals, and endogenous compounds. Glucuronidation is a bisubstrate reaction that requires the aglycone and the cofactor, UDP-GlcUA. Accumulating evidence suggests that the bisubstrate reaction follows a compulsory-order ternary mechanism. To simplify the kinetic modeling of glucuronidation reactions in vitro, UDP-GlcUA is usually added to incubations in large excess. Many factors have been shown to influence UGT activity and kinetics in vitro, and these must be accounted for during experimental design and data interpretation. While the assessment of drug-drug interactions resulting from UGT inhibition has been challenging in the past, the increasing availability of UGT enzyme-selective substrate and inhibitor "probes" provides the prospect for more reliable reaction phenotyping and assessment of drug-drug interaction potential. Although extrapolation of the in vitro intrinsic clearance of a glucuronidated drug often underpredicts in vivo clearance, careful selection of in vitro experimental conditions and inclusion of extrahepatic glucuronidation may improve the predictivity of in vitro-in vivo extrapolation. Physiologically based pharmacokinetic (PBPK) modeling has also shown to be of value for predicting PK of drugs eliminated by glucuronidation.
Collapse
Affiliation(s)
- Jin Zhou
- Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT, USA.
| | - Upendra A Argikar
- Translational Medicine, Novartis Institutes for BioMedical Research, Inc., Cambridge, MA, USA
| | - John O Miners
- Department of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| |
Collapse
|
4
|
Mutlu E, Gibbs ST, South N, Pierfelice J, Burback B, Germolec D, Waidyanatha S. Comparative toxicokinetics of Trans-resveratrol and its major metabolites in Harlan Sprague Dawley rats and B6C3F1/N mice following oral and intravenous administration. Toxicol Appl Pharmacol 2020; 394:114962. [PMID: 32205187 PMCID: PMC7398575 DOI: 10.1016/j.taap.2020.114962] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/15/2020] [Accepted: 03/19/2020] [Indexed: 12/15/2022]
Abstract
Trans-resveratrol (RES) is a naturally occurring stilbene found in numerous plants and foods. Due to its widespread human exposure and lack of toxicity and carcinogenicity data, RES was nominated to the National Toxicology Program for testing. To aid the toxicology studies, the dose, sex, and species differences in RES toxicokinetics was investigated in Harlan Sprague Dawley rats and B6C3F1/N mice following single intravenous (IV) (10 mg/kg) or oral gavage administration (312.5, 625, and 1250 mg/kg and 625, 1250, and 2500 mg/kg in rats and mice, respectively). Following IV and gavage administration, systemic exposure of RES based on AUC was trans-resveratrol-3-O-β-D-glucuronide (R3G)> > trans-resveratrol-3-sulfate (R3S) > RES in both species. Following gavage administration Tmax_predicted values were ≤ 263 min for both species and sexes. RES elimination half-life was longer in rats than mice, and shortest in male mice. Clearance was slower in mice with no apparent sex difference in both species. In both rats and mice, following gavage administration AUC increased proportionally to the dose. After gavage administration, enterohepatic recirculation of RES was observed in both rats and mice with secondary peaks occurring around 640 min in the concentration-time profiles. RES was rapidly metabolized to R3S and R3G in both species. Extensive first pass conjugation and metabolism resulted in low levels of the parent compound RES which was confirmed by the low estimates for bioavailability. The bioavailability of RES was low, ~12-31% and ~2-6% for rats and mice, respectively, with no apparent difference between sexes.
Collapse
Affiliation(s)
- Esra Mutlu
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States of America.
| | - Seth T Gibbs
- Battelle, Columbus, OH, United States of America
| | | | | | | | - Dori Germolec
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States of America
| | - Suramya Waidyanatha
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States of America
| |
Collapse
|
5
|
Resveratrol Plays Protective Roles on Kidney of Uremic Rats via Activating HSP70 Expression. BIOMED RESEARCH INTERNATIONAL 2020; 2020:2126748. [PMID: 32280682 PMCID: PMC7125444 DOI: 10.1155/2020/2126748] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 02/27/2020] [Indexed: 12/26/2022]
Abstract
Objective To investigate the protective effects of resveratrol on kidney of uremic rats and to explore whether the mechanism is associated with heat shock protein 70 (HSP70) expression. Methods Sixty male Sprague Dawley rats were randomly separated into 5 groups, including sham group, uremic model group, and different doses of resveratrol group (5 mg/kg, 10 mg/kg, and 20 mg/kg). The serum creatinine (Cr) and urea nitrogen (BUN) levels were detected by Automatic Biochemical Analyzer (ABA). The pathological changes of renal tissues and the renal interstitial fibrosis were analyzed by hematoxylin-eosin (HE) and Masson, respectively. The expression of HSP70 protein in renal tissues was detected by immunohistochemistry. The expression of HSP70 and NF-κB pathway-related proteins were detected by Western blot. To further validate the protective role of resveratrol through activating HSP70 in uremic rats, HSP70 activator (17-AAG) and HSP70 inhibitor group (MKT-077) were used. Results In the model group, the levels of Cr and BUN in serum were significantly increased, and the renal interstitial collagen deposition was also obviously increased (p < 0.05). Compared with the model group, the levels of Cr and BUN in different doses of resveratrol groups were remarkably declined, and the renal interstitial collagen deposition was declined (p < 0.05). Resveratrol also significantly improved the renal tissue lesions when compared with the model group. In renal tissues, different doses of resveratrol treatment remarkably raised HSP70 and p-IκBα expression and also remarkably declined the level of p-P65 protein (p < 0.05). Meanwhile, the effect of 17-AAG was similar to 20 mg/kg resveratrol on NF-κB pathway-related proteins expression. After the added MKT-077 in the resveratrol treatment group, the levels of HSP70 and p-IκBα in the renal tissue were remarkably declined; however, the levels of p-P65 protein was remarkably raised (p < 0.05). Conclusion Resveratrol played a protective role on the kidney of uremic rats through activating HSP70 expression.
Collapse
|
6
|
Wu H, Chen L, Zhu F, Han X, Sun L, Chen K. The Cytotoxicity Effect of Resveratrol: Cell Cycle Arrest and Induced Apoptosis of Breast Cancer 4T1 Cells. Toxins (Basel) 2019; 11:toxins11120731. [PMID: 31847250 PMCID: PMC6950385 DOI: 10.3390/toxins11120731] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 12/15/2022] Open
Abstract
Resveratrol, a natural polyterpenoid, can scavenge reactive oxygen species in vivo to carry out the functions of antioxidation and antiaging. Resveratrol’s anti-cancer capability has attracted widespread attention, but its molecular mechanism has not been systematically explained. In this study, by comparing the activity of normal cell lines and cancer cell lines after treating with resveratrol, it was found that resveratrol has more significant cytotoxicity in cancer cell lines. Resveratrol could play a toxic role through inducing apoptosis of the cancer cell in a time- and concentration-dependent manner. A total of 330 significantly differential genes were identified through large-scale transcriptome sequencing, among which 103 genes were upregulated and 227 genes were downregulated. Transcriptome and qRT-PCR data proved that a large number of genes related to cell cycle were differentially expressed after the treatment of resveratrol. The changes of cell cycle phases at different time points after treating with resveratrol were further detected, and it was found that the cells were arrested in the S phase because of the percentage of cells in S phase increased and cells in G1/G0 phase decreased. In conclusion, resveratrol can inhibit the proliferation of 4T1 cancer cells by inhibiting cell cycle and inducing apoptosis.
Collapse
Affiliation(s)
| | | | | | | | | | - Keping Chen
- Correspondence: ; Tel./Fax: +86-511-88791923
| |
Collapse
|
7
|
NMR-based metabonomic approach reveals changes in the urinary and fecal metabolome caused by resveratrol. J Pharm Biomed Anal 2019; 162:234-241. [DOI: 10.1016/j.jpba.2018.09.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 09/02/2018] [Accepted: 09/12/2018] [Indexed: 01/14/2023]
|
8
|
Matin B, Sherbini AA, Alam N, Harmatz JS, Greenblatt DJ. Resveratrol glucuronidation in vitro: potential implications of inhibition by probenecid. J Pharm Pharmacol 2018; 71:371-378. [PMID: 30417385 DOI: 10.1111/jphp.13037] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 10/19/2018] [Indexed: 12/15/2022]
Abstract
OBJECTIVES Resveratrol is a naturally occurring antioxidant with therapeutic potential in prevention and treatment of neoplastic disease and other human disorders. However, net clearance of resveratrol in humans is very high, mainly due to glucuronide conjugation. This leads to extensive presystemic extraction and low plasma concentrations after oral dosage. The present study evaluated the effect of probenecid, an inhibitor of glucuronide conjugation, on resveratrol metabolism in vitro. METHODS Biotransformation of resveratrol to its 3-O-glucuronide and 4'-O-glucuronide conjugates was studied in vitro using human liver microsomal preparations. The mechanism and inhibitory potency of probenecid were evaluated based on a mixed competitive-noncompetitive inhibition model. KEY FINDINGS Probenecid inhibition of resveratrol 3-O-glucuronidation was predominantly noncompetitive, with an inhibition constant (Ki ) averaging 3.1 mm. CONCLUSIONS The ratio of in vivo maximum concentration of probenecid [I] during usual clinical use to the in vitro Ki value ([I]/Ki ) exceeds the boundary value of 0.1, used by regulatory agencies to identify the possibility of clinical drug interactions. This finding, together with the known property of probenecid as an inhibitor of glucuronide conjugation in humans, suggests that probenecid could serve as a pharmacokinetic boosting agent to enhance systemic exposure to resveratrol in humans.
Collapse
Affiliation(s)
- Bahar Matin
- Graduate Program in Pharmacology and Drug Development, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
| | - Ahmad A Sherbini
- Graduate Program in Pharmacology and Drug Development, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
| | - Novera Alam
- Graduate Program in Pharmacology and Drug Development, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
| | - Jerold S Harmatz
- Graduate Program in Pharmacology and Drug Development, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
| | - David J Greenblatt
- Graduate Program in Pharmacology and Drug Development, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
| |
Collapse
|
9
|
Protective effect of resveratrol against light-induced retinal degeneration in aged SAMP8 mice. Oncotarget 2017; 8:65778-65788. [PMID: 29029471 PMCID: PMC5630371 DOI: 10.18632/oncotarget.19473] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 06/24/2017] [Indexed: 12/17/2022] Open
Abstract
Purpose The purpose of this study was to determine the protective effects of Resveratrol (RESV) on acute bright light-induced retinal degeneration in aged senescence accelerated mouse strain. Methods Ten three-month-old male SAMP8 mice (prone to aging) were randomly assigned to two experimental dietary groups: one untreated group and one RESV treatment group (n=20 eyes for each group). After 30 days of treatment, mice were exposed to intense bright light. Ten male SAMR1 mice (resistant to aging) served as control (n=20 eyes). The protective effects of RESV administration on light-induced retinal degeneration in SAMP8 strain as well as the effect of bright light damage in the retinas of SAMP8 mice were analyzed by electroretinography (ERG), retinal histology, mRNA, protein and lipid profile. Results 68%-85% of a-wave amplitude and 72%-92% of b-wave amplitude were persevered by RESV in SAMP8 mice that were exposed to light damage. Also, RESV preserved their photoreceptor nuclei. mRNA expression of neuroprotective factors leukemia inhibitory factor (LIF), brain derived neurotrophic factor (BDNF), oncostatin M (OSM), cardiotrophin 1(CT-1) and cardiotrophin-like cytokine (CLC) were up-regulated 28, 8, 7, 5 and 9-fold in SAMP8 mice after RESV treatment. In addition, RESV could suppress the NF-κB pathway by down-regulating the expression of pIκB. Light damage led to increase of saturated FA, monoenoic FA, n6 PUFA and n6/n3 ratio and decrease of Docosahexaenoic acid (DHA). There was no significant difference on DHA and the ratio of n6/n3-FA between the untreated and RESV treated SAMP8 mice. Conclusions Collectively, our study provides evidence that RESV prevents light-induced retinal damage associated with aging.
Collapse
|
10
|
Antitumor effect of thymoquinone combined with resveratrol on mice transplanted with breast cancer. ASIAN PAC J TROP MED 2017; 10:400-408. [PMID: 28552110 DOI: 10.1016/j.apjtm.2017.03.026] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 02/14/2017] [Accepted: 03/15/2017] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE To test the anticancer potential activity of the combination of thymoquinone (TQ) and resveratrol (RES) against breast cancer in mice. METHODS The antiproliferative activity of TQ, RES and their combination was assessed against three breast cancer cell lines and one normal cells using MTT assay. The combination index was calculated using isobolographic method. Balb/C mice were inoculated with EMT6/P cells and in vivo antitumor activity was evaluated. RESULTS The combination therapy also caused significant decrease in tumor size with a percentage cure of 60%. The combination therapy induced geographic necrosis, enhanced apoptosis, and decreased VEGF expression. Serum levels of IFN-γ were elevated in mice treated with combination therapy with no liver or kidney toxicity. CONCLUSIONS The combination of TQ and RES against breast cancer in mice can work synergistically. The anticancer effect of this combination is mediated by apoptosis induction, angiogenesis inhibition and immune modulation.
Collapse
|
11
|
Combination Therapy using Co-encapsulated Resveratrol and Paclitaxel in Liposomes for Drug Resistance Reversal in Breast Cancer Cells in vivo. Sci Rep 2016; 6:22390. [PMID: 26947928 PMCID: PMC4780086 DOI: 10.1038/srep22390] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 02/11/2016] [Indexed: 12/19/2022] Open
Abstract
Multidrug resistance (MDR) is a major impediment to cancer treatment. A promising strategy for treating MDR is the joint delivery of combined anticancer agents to tumor cells in a single nanocarrier. Here, for the first time, Resveratrol (Res) was co-encapsulated with paclitaxel (PTX) in a PEGylated liposome to construct a carrier-delivered form of combination therapy for drug-resistant tumors. The composite liposome had an average diameter of 50 nm with encapsulated efficiencies of above 50%. The studies demonstrated that the composite liposome could generate potent cytotoxicity against the drug-resistant MCF-7/Adr tumor cells in vitro and enhance the bioavailability and the tumor-retention of the drugs in vivo. Moreover, systemic therapy with the composite liposome effectively inhibited drug-resistant tumor in mice (p < 0.01), without any notable increase in the toxicity. These results suggested that the co-delivery of Res and a cytotoxic agent in a nanocarrier may potentially improve the treatment of drug-resistant tumors.
Collapse
|
12
|
Foti RS, Tyndale RF, Garcia KLP, Sweet DH, Nagar S, Sharan S, Rock DA. "Target-Site" Drug Metabolism and Transport. Drug Metab Dispos 2015; 43:1156-68. [PMID: 25986849 PMCID: PMC11024933 DOI: 10.1124/dmd.115.064576] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Accepted: 05/18/2015] [Indexed: 04/20/2024] Open
Abstract
The recent symposium on "Target-Site" Drug Metabolism and Transport that was sponsored by the American Society for Pharmacology and Experimental Therapeutics at the 2014 Experimental Biology meeting in San Diego is summarized in this report. Emerging evidence has demonstrated that drug-metabolizing enzyme and transporter activity at the site of therapeutic action can affect the efficacy, safety, and metabolic properties of a given drug, with potential outcomes including altered dosing regimens, stricter exclusion criteria, or even the failure of a new chemical entity in clinical trials. Drug metabolism within the brain, for example, can contribute to metabolic activation of therapeutic drugs such as codeine as well as the elimination of potential neurotoxins in the brain. Similarly, the activity of oxidative and conjugative drug-metabolizing enzymes in the lung can have an effect on the efficacy of compounds such as resveratrol. In addition to metabolism, the active transport of compounds into or away from the site of action can also influence the outcome of a given therapeutic regimen or disease progression. For example, organic anion transporter 3 is involved in the initiation of pancreatic β-cell dysfunction and may have a role in how uremic toxins enter pancreatic β-cells and ultimately contribute to the pathogenesis of gestational diabetes. Finally, it is likely that a combination of target-specific metabolism and cellular internalization may have a significant role in determining the pharmacokinetics and efficacy of antibody-drug conjugates, a finding which has resulted in the development of a host of new analytical methods that are now used for characterizing the metabolism and disposition of antibody-drug conjugates. Taken together, the research summarized herein can provide for an increased understanding of potential barriers to drug efficacy and allow for a more rational approach for developing safe and effective therapeutics.
Collapse
Affiliation(s)
- Robert S Foti
- Amgen Pharmacokinetics and Drug Metabolism, Seattle, Washington (R.S.F., D.A.R.); Departments of Pharmacology and Toxicology and Psychiatry, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, Ontario, Canada (R.F.T., K.L.P.G.); Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia (D.H.S.); School of Pharmacy, Temple University, Philadelphia, Pennsylvania (S.N.); and College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma (S.S.)
| | - Rachel F Tyndale
- Amgen Pharmacokinetics and Drug Metabolism, Seattle, Washington (R.S.F., D.A.R.); Departments of Pharmacology and Toxicology and Psychiatry, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, Ontario, Canada (R.F.T., K.L.P.G.); Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia (D.H.S.); School of Pharmacy, Temple University, Philadelphia, Pennsylvania (S.N.); and College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma (S.S.)
| | - Kristine L P Garcia
- Amgen Pharmacokinetics and Drug Metabolism, Seattle, Washington (R.S.F., D.A.R.); Departments of Pharmacology and Toxicology and Psychiatry, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, Ontario, Canada (R.F.T., K.L.P.G.); Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia (D.H.S.); School of Pharmacy, Temple University, Philadelphia, Pennsylvania (S.N.); and College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma (S.S.)
| | - Douglas H Sweet
- Amgen Pharmacokinetics and Drug Metabolism, Seattle, Washington (R.S.F., D.A.R.); Departments of Pharmacology and Toxicology and Psychiatry, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, Ontario, Canada (R.F.T., K.L.P.G.); Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia (D.H.S.); School of Pharmacy, Temple University, Philadelphia, Pennsylvania (S.N.); and College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma (S.S.)
| | - Swati Nagar
- Amgen Pharmacokinetics and Drug Metabolism, Seattle, Washington (R.S.F., D.A.R.); Departments of Pharmacology and Toxicology and Psychiatry, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, Ontario, Canada (R.F.T., K.L.P.G.); Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia (D.H.S.); School of Pharmacy, Temple University, Philadelphia, Pennsylvania (S.N.); and College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma (S.S.)
| | - Satish Sharan
- Amgen Pharmacokinetics and Drug Metabolism, Seattle, Washington (R.S.F., D.A.R.); Departments of Pharmacology and Toxicology and Psychiatry, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, Ontario, Canada (R.F.T., K.L.P.G.); Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia (D.H.S.); School of Pharmacy, Temple University, Philadelphia, Pennsylvania (S.N.); and College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma (S.S.)
| | - Dan A Rock
- Amgen Pharmacokinetics and Drug Metabolism, Seattle, Washington (R.S.F., D.A.R.); Departments of Pharmacology and Toxicology and Psychiatry, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, Ontario, Canada (R.F.T., K.L.P.G.); Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia (D.H.S.); School of Pharmacy, Temple University, Philadelphia, Pennsylvania (S.N.); and College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma (S.S.)
| |
Collapse
|
13
|
Lepak A, Gutmann A, Kulmer ST, Nidetzky B. Creating a Water-Soluble Resveratrol-Based Antioxidant by Site-Selective Enzymatic Glucosylation. Chembiochem 2015; 16:1870-1874. [DOI: 10.1002/cbic.201500284] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Indexed: 11/10/2022]
|
14
|
Zhang Q, Bian Y, Shi Y, Zheng S, Gu X, Zhang D, Zhu X, Wang X, Jiang D, Xiong Q. An economical and efficient technology for the extraction of resveratrol from peanut (Arachis hypogaea) sprouts by multi-stage countercurrent extraction. Food Chem 2015; 179:15-25. [DOI: 10.1016/j.foodchem.2015.01.113] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 12/10/2014] [Accepted: 01/20/2015] [Indexed: 11/25/2022]
|
15
|
Possible role of resveratrol targeting estradiol and neprilysin pathways in lipopolysaccharide model of Alzheimer disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 822:107-18. [PMID: 25416980 DOI: 10.1007/978-3-319-08927-0_12] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease (AD) is an irreversible, progressive neurodegenerative brain disease that slowly destroys memory and thinking skills. It is the most common cause of dementia among older people. One of the most important hallmarks of AD is the presence of amyloid beta (Aβ) peptide in the brain that suggests that it is the primary trigger for neuronal loss. Herbal extracts have been studied over the years for their potential therapeutic effect in AD. Resveratrol (RSV), one of the most important phytoestrogens, is considered to be useful as estrogen plays an important role in AD. One of the most important amyloid degrading enzymes is neprilysin (NEP), which plays a major role in degrading Aβ, and mainly affected by estrogen. So, the aim of the present study is investigating the possible role of resveratrol in lipopolysaccharide model of AD and the implication of its possible role in regulating the estradiol and neprilysin pathways. Mice were divided into four groups: Control group (0.9 % saline), LPS group (0.8 mg/kg i.p once), Treatment group with RSV (mice were once injected with LPS then after 30 min given a dose of {4 mg/kg} RSV for 7 days), and RSV group only (mice received 4 mg/kg i.p for 7 days only). After 7 days mice were subjected to different behavioral tests using Y-maze, object recognition test, and open field tests. Estradiol and NEP level were measured using ELISA kit. Results showed RSV was able to reverse the decline in different types of memory (working, nonspatial, and locomotor functions) caused by LPS induction in mice. Moreover RSV was able to significantly increase both the estradiol level and NEP level and that may have a great role to decrease Aβ deposition as it has been confirmed that there is a link between NEP and estradiol level; by upregulation of estradiol level this consequently leads to increase in the level of NEP level, and by increasing the NEP level in brain, this lead to decrease in Aβ deposition and enhancing its degradation by NEP.
Collapse
|
16
|
Abstract
Glucuronidation, catalyzed by uridine diphosphate glucuronosyltransferases (UGTs), is an important process for the metabolism and clearance of many lipophilic chemicals, including drugs, environmental chemicals, and endogenous compounds. Glucuronidation is a bi-substrate reaction that requires the aglycone and a cofactor, UDPGA. Accumulating evidence suggests that the bi-substrate reaction follows a compulsory-order ternary mechanism. To simplify the kinetic modelling of glucuronidation reactions in vitro, UDPGA is usually added to incubations in large excess. Many factors have been shown to influence UGT activity and kinetics in vitro, and these must be accounted for in experimental design and data interpretation. Assessing drug-drug interactions (DDIs) involving UGT inhibition remains challenging. However, the increasing availability of UGT enzyme-specific substrate and inhibitor "probes" provides the prospect for more reliable reaction phenotyping and assessment of DDI potential. Although extrapolation of the in vitro intrinsic clearance of a glucuronidated drug often under-predicts in vivo clearance, careful selection of in vitro experimental conditions and inclusion of extrahepatic glucuronidation may improve the predictivity of in vitro-in vivo extrapolation (IVIVE).
Collapse
|
17
|
Tian X, Liang S, Wang C, Wu B, Ge G, Deng S, Liu K, Yang L, Ma X. Regioselective glucuronidation of andrographolide and its major derivatives: metabolite identification, isozyme contribution, and species differences. AAPS JOURNAL 2014; 17:156-66. [PMID: 25204783 DOI: 10.1208/s12248-014-9658-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 08/21/2014] [Indexed: 01/16/2023]
Abstract
Andrographolide (AND) and two of its derivatives, deoxyandrographolide (DEO) and dehydroandrographolide (DEH), are widely used in clinical practice as anti-inflammatory agents. However, UDP-glucuronosyltransferase (UGT)-mediated phase II metabolism of these compounds is not fully understood. In this study, glucuronidation of AND, DEO, and DEH was characterized using liver microsomes and recombinant UGT enzymes. We isolated six glucuronides and identified them using 1D and 2D nuclear magnetic resonance (NMR) spectroscopy. We also systematically analyzed various kinetic parameters (K m, V max, and CLint) for glucuronidation of AND, DEO, and DEH. Among 12 commercially available UGT enzymes, UGT1A3, 1A4, 2B4, and 2B7 exhibited metabolic activities toward AND, DEO, and DEH. Further, UGT2B7 made the greatest contribution to glucuronidation of all three anti-inflammatory agents. Regioselective glucuronidation showed considerable species differences. 19-O-Glucuronides were present in liver microsomes from all species except rats. 3-O-Glucuronides were produced by pig and cynomolgus monkey liver microsomes for all compounds, and 3-O-glucuronide of DEH was detected in mouse and rat liver microsomes (RLM). Variations in K m values were 48.6-fold (1.93-93.6 μM) and 49.5-fold (2.01-99.1 μM) for 19-O-glucuronide and 3-O-glucuronide formation, respectively. Total intrinsic clearances (CLint) for 3-O- and 19-O-glucuronidation varied 4.8-fold (22.7-110 μL min(-1) mg(-1)), 10.6-fold (94.2-991 μL min(-1) mg(-1)), and 8.3-fold (122-1,010 μL min(-1) mg(-1)), for AND, DEH, and DEO, respectively. Our results indicate that UGT2B7 is the major UGT enzyme involved in the metabolism of AND, DEO, and DEH. Metabolic pathways in the glucuronidation of AND, DEO, and DEH showed considerable species differences.
Collapse
Affiliation(s)
- Xiangge Tian
- College of Pharmacy, Academy of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Brantley SJ, Argikar AA, Lin YS, Nagar S, Paine MF. Herb-drug interactions: challenges and opportunities for improved predictions. Drug Metab Dispos 2014; 42:301-17. [PMID: 24335390 PMCID: PMC3935140 DOI: 10.1124/dmd.113.055236] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 12/11/2013] [Indexed: 01/23/2023] Open
Abstract
Supported by a usage history that predates written records and the perception that "natural" ensures safety, herbal products have increasingly been incorporated into Western health care. Consumers often self-administer these products concomitantly with conventional medications without informing their health care provider(s). Such herb-drug combinations can produce untoward effects when the herbal product perturbs the activity of drug metabolizing enzymes and/or transporters. Despite increasing recognition of these types of herb-drug interactions, a standard system for interaction prediction and evaluation is nonexistent. Consequently, the mechanisms underlying herb-drug interactions remain an understudied area of pharmacotherapy. Evaluation of herbal product interaction liability is challenging due to variability in herbal product composition, uncertainty of the causative constituents, and often scant knowledge of causative constituent pharmacokinetics. These limitations are confounded further by the varying perspectives concerning herbal product regulation. Systematic evaluation of herbal product drug interaction liability, as is routine for new drugs under development, necessitates identifying individual constituents from herbal products and characterizing the interaction potential of such constituents. Integration of this information into in silico models that estimate the pharmacokinetics of individual constituents should facilitate prospective identification of herb-drug interactions. These concepts are highlighted with the exemplar herbal products milk thistle and resveratrol. Implementation of this methodology should help provide definitive information to both consumers and clinicians about the risk of adding herbal products to conventional pharmacotherapeutic regimens.
Collapse
Affiliation(s)
- Scott J Brantley
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (S.J.B.); Department of Pharmaceutical Sciences, Temple University School of Pharmacy, Philadelphia, Pennsylvania (A.A.A., S.N.); Department of Pharmaceutics, University of Washington, Seattle, Washington (Y.S.L.); and College of Pharmacy, Washington State University, Spokane, Washington (M.F.P.)
| | | | | | | | | |
Collapse
|
19
|
|
20
|
Hu N, Mei M, Ruan J, Wu W, Wang Y, Yan R. Regioselective glucuronidation of oxyresveratrol, a natural hydroxystilbene, by human liver and intestinal microsomes and recombinant UGTs. Drug Metab Pharmacokinet 2013; 29:229-36. [PMID: 24256624 DOI: 10.2133/dmpk.dmpk-13-rg-102] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Oxyresveratrol (OXY) is a natural hydroxystilbene that shows similar bioactivity but better water solubility than resveratrol. This study aims to characterize its glucuronidation kinetics in human liver (HLMs) and intestinal (HIMs) microsomes and identify the main UDP-glucuronosyltransferase (UGT) isoforms involved. Three and four mono-glucuronides of OXY were generated in HIMs and HLMs, respectively, with oxyresveratrol-2-O-β-D-glucuronosyl (G4) as the major metabolite in both organs. The kinetics of G4 formation fit a sigmoidal model in HLMs and biphasic kinetics in HIMs. Multiple UGT isoforms catalyzed G4 formation with the highest activity observed with UGT1A9 followed by UGT1A1. G4 formation by both isoforms followed substrate inhibition kinetics. Propofol (UGT1A9 inhibitor) effectively blocked G4 generation in HLMs (IC50 63.7 ± 11.6 µM), whereas the UGT1A1 inhibitor bilirubin only produced partial inhibition in HLMs and HIMs. These findings shed light on the metabolic mechanism of OXY and arouse awareness of drug interactions.
Collapse
Affiliation(s)
- Nan Hu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau
| | | | | | | | | | | |
Collapse
|
21
|
Sánchez-Tena S, Reyes-Zurita FJ, Díaz-Moralli S, Vinardell MP, Reed M, García-García F, Dopazo J, Lupiáñez JA, Günther U, Cascante M. Maslinic acid-enriched diet decreases intestinal tumorigenesis in Apc(Min/+) mice through transcriptomic and metabolomic reprogramming. PLoS One 2013; 8:e59392. [PMID: 23527181 PMCID: PMC3601079 DOI: 10.1371/journal.pone.0059392] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Accepted: 02/14/2013] [Indexed: 12/22/2022] Open
Abstract
Chemoprevention is a pragmatic approach to reduce the risk of colorectal cancer, one of the leading causes of cancer-related death in western countries. In this regard, maslinic acid (MA), a pentacyclic triterpene extracted from wax-like coatings of olives, is known to inhibit proliferation and induce apoptosis in colon cancer cell lines without affecting normal intestinal cells. The present study evaluated the chemopreventive efficacy and associated mechanisms of maslinic acid treatment on spontaneous intestinal tumorigenesis in ApcMin/+ mice. Twenty-two mice were randomized into 2 groups: control group and MA group, fed with a maslinic acid–supplemented diet for six weeks. MA treatment reduced total intestinal polyp formation by 45% (P<0.01). Putative molecular mechanisms associated with suppressing intestinal polyposis in ApcMin/+ mice were investigated by comparing microarray expression profiles of MA-treated and control mice and by analyzing the serum metabolic profile using NMR techniques. The different expression phenotype induced by MA suggested that it exerts its chemopreventive action mainly by inhibiting cell-survival signaling and inflammation. These changes eventually induce G1-phase cell cycle arrest and apoptosis. Moreover, the metabolic changes induced by MA treatment were associated with a protective profile against intestinal tumorigenesis. These results show the efficacy and underlying mechanisms of MA against intestinal tumor development in the ApcMin/+ mice model, suggesting its chemopreventive potential against colorectal cancer.
Collapse
Affiliation(s)
- Susana Sánchez-Tena
- Department of Biochemistry and Molecular Biology, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
- Institute of Biomedicine, Universitat de Barcelona and CSIC-Associated Unit, Barcelona, Spain
| | | | - Santiago Díaz-Moralli
- Department of Biochemistry and Molecular Biology, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
- Institute of Biomedicine, Universitat de Barcelona and CSIC-Associated Unit, Barcelona, Spain
| | - Maria Pilar Vinardell
- Department of Physiology, Faculty of Pharmacy, University of Barcelona, Barcelona, Spain
| | - Michelle Reed
- Henry Wellcome Building for Biomolecular NMR Spectroscopy, CR UK Institute for Cancer Studies, University of Birmingham, Birmingham, United Kingdom
| | - Francisco García-García
- Functional Genomics Node, National Institute of Bioinformatics, Centro de Investigación Pricipe Felipe, Valencia, Spain
- Department of Bioinformatics, Centro de Investigación Pricipe Felipe, Valencia, Spain
- Centro de Investigación Biomédica En Red de Enfermedades Raras, Valencia, Spain
| | - Joaquín Dopazo
- Functional Genomics Node, National Institute of Bioinformatics, Centro de Investigación Pricipe Felipe, Valencia, Spain
- Department of Bioinformatics, Centro de Investigación Pricipe Felipe, Valencia, Spain
- Centro de Investigación Biomédica En Red de Enfermedades Raras, Valencia, Spain
| | - José A. Lupiáñez
- Department of Biochemistry and Molecular Biology, University of Granada, Granada, Spain
| | - Ulrich Günther
- Henry Wellcome Building for Biomolecular NMR Spectroscopy, CR UK Institute for Cancer Studies, University of Birmingham, Birmingham, United Kingdom
| | - Marta Cascante
- Department of Biochemistry and Molecular Biology, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
- Institute of Biomedicine, Universitat de Barcelona and CSIC-Associated Unit, Barcelona, Spain
- * E-mail:
| |
Collapse
|
22
|
Sharan S, Nagar S. Pulmonary metabolism of resveratrol: in vitro and in vivo evidence. Drug Metab Dispos 2013; 41:1163-9. [PMID: 23474649 DOI: 10.1124/dmd.113.051326] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The role of pulmonary metabolism in trans-resveratrol (RES) pharmacokinetics was studied in a mouse model. Plasma concentrations of RES and its major metabolites trans-resveratrol-3-sulfate (R3S) and trans-resveratrol-3-glucuronide (R3G) were compared after administration of RES by intravenous (IV) and intra-arterial (IA) routes. Total area under the curve (AUC) of RES decreased by approximately 50% when RES was administered by the IV route compared with the IA route. The AUC of R3G was also significantly higher in mice administered RES by the IV route compared with the IA route. In vitro studies performed with mouse and human lung fractions confirmed pulmonary metabolism of RES. Interestingly, mouse-lung fractions gave rise to both R3S and R3G, whereas human lung fractions yielded R3S. This indicates marked interspecies variation in RES conjugation, especially in the context of extrapolating rodent data to humans. Taken together, the results presented here underline, for the first time, the impact of pulmonary metabolism on resveratrol pharmacokinetics and interspecies differences in RES pulmonary metabolism.
Collapse
Affiliation(s)
- Satish Sharan
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, Philadelphia, Pennsylvania, USA
| | | |
Collapse
|
23
|
Uchihashi S, Nishikawa M, Sakaki T, Ikushiro SI. The critical role of amino acid residue at position 117 of mouse UDP-glucuronosyltransfererase 1a6a and 1a6b in resveratrol glucuronidation. J Biochem 2012; 152:331-40. [DOI: 10.1093/jb/mvs078] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
24
|
Ma L, Sun J, Peng Y, Zhang R, Shao F, Hu X, Zhu J, Wang X, Cheng X, Zhu Y, Wan P, Feng D, Wu H, Wang G. Glucuronidation of Edaravone by Human Liver and Kidney Microsomes: Biphasic Kinetics and Identification of UGT1A9 as the Major UDP-Glucuronosyltransferase Isoform. Drug Metab Dispos 2012; 40:734-41. [DOI: 10.1124/dmd.111.043356] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
25
|
Abstract
Inhibition of enzyme activity at high substrate concentrations, so-called "substrate inhibition," is commonly observed and has been recognized in drug metabolism reactions since the last decade. Although the importance of such "atypical" kinetics in vivo remains poorly understood, a substrate with substrate inhibition kinetics has been shown to unconventionally alter the metabolism of other substrates. In recent years, it is becoming increasingly evident that the mechanisms for substrate inhibition are highly complex, which are possibly contributed by multiple (at least two) binding sites within the enzyme protein, the formation of a ternary dead-end enzyme complex, and/or the ligand-induced changes in enzyme conformation. This review primarily discusses the mechanisms for substrate inhibition displayed by the important drug-metabolizing enzymes, such as cytochrome p450s, UDP-glucuronyltransferases, and sulfotransferases. Kinetic modeling of substrate inhibition in the absence or presence of a modifier is another central issue in this review because of its importance in the determination of kinetic parameters and in vitro/in vivo predictions.
Collapse
Affiliation(s)
- Baojian Wu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Texas, USA.
| |
Collapse
|
26
|
Chimalakonda KC, Bratton SM, Le VH, Yiew KH, Dineva A, Moran CL, James LP, Moran JH, Radominska-Pandya A. Conjugation of synthetic cannabinoids JWH-018 and JWH-073, metabolites by human UDP-glucuronosyltransferases. Drug Metab Dispos 2011; 39:1967-76. [PMID: 21746969 DOI: 10.1124/dmd.111.040709] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
K2, a synthetic cannabinoid (SC), is an emerging drug of abuse touted as "legal marijuana" and marketed to young teens and first-time drug users. Symptoms associated with K2 use include extreme agitation, syncope, tachycardia, and visual and auditory hallucinations. One major challenge to clinicians is the lack of clinical, pharmacological, and metabolic information for the detection and characterization of K2 and its metabolites in human samples. Information on the metabolic pathway of SCs is very limited. However, previous reports have shown the metabolites of these compounds are excreted primarily as glucuronic acid conjugates. Based on this information, this study evaluates nine human recombinant uridine diphosphate-glucuronosyltransferase (UGT) isoforms and human liver and intestinal microsomes for their ability to glucuronidate hydroxylated metabolites of 1-naphthalenyl-1(1-pentyl-1H-indol-3-yl)-methanone (JWH-018) and (1-butyl-1H-indol-3-yl)-1-naphthalenyl-methanone (JWH-073), the two most common SCs found in K2 products. Conjugates were identified and characterized using liquid chromatography/tandem mass spectrometry, whereas kinetic parameters were quantified using high-performance liquid chromatography-UV-visible methods. UGT1A1, UGT1A3, UGT1A9, UGT1A10, and UGT2B7 were shown to be the major enzymes involved, showing relatively high affinity with K(m) ranging from 12 to 18 μM for some hydroxylated K2s. These UGTs also exhibited a high metabolic capacity for these compounds, which indicates that K2 metabolites may be rapidly glucuronidated and eliminated from the body. Studies of K2 metabolites will help future development and validation of a specific assay for K2 and its metabolites and will allow researchers to fully explore their pharmacological actions.
Collapse
Affiliation(s)
- Krishna C Chimalakonda
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Miksits M, Maier-Salamon A, Vo TPN, Sulyok M, Schuhmacher R, Szekeres T, Jäger W. Glucuronidation of piceatannol by human liver microsomes: major role of UGT1A1, UGT1A8 and UGT1A10. J Pharm Pharmacol 2010; 62:47-54. [PMID: 20722998 DOI: 10.1211/jpp.62.01.0004] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVES Piceatannol, a dietary polyphenol present in grapes and wine, is known for its promising anticancer and anti-inflammatory activity. The aim of this study was to analyse the concentration-dependent glucuronidation of piceatannol in vitro. METHODS To determine the glucuronidation of piceatannol, experiments were conducted with human liver microsomes as well as using a panel of 12 recombinant UDP-glucuronosyltransferase isoforms. Furthermore, the chemical structures of novel glucuronides were identified by liquid chromatography-tandem mass spectrometry (LC-MS/MS). KEY FINDINGS Along with piceatannol it was possible to identify three metabolites whose structures were identified by LC-MS/MS as piceatannol monoglucuronides (M1-M3). Formation of M1 and M3 exhibited a pattern of substrate inhibition, with apparent K(i) and V(max)/K(m) values of 103 +/- 26.6 microm and 3.8 +/- 1.3 microl/mg protein per min, respectively, for M1 and 233 +/- 61.4 microm and 19.8 +/- 9.5 microl/mg protein per min, respectively, for M3. In contrast, formation of metabolite M2 followed classical Michaelis-Menten kinetics, with a K(m) of 18.9 +/- 8.1 microm and a V(max) of 0.21 +/- 0.02 nmol/mg protein per min. Incubation in the presence of human recombinant UDP-glucuronosyltransferases (UGTs) demonstrated that M1 was formed nearly equally by UGT1A1 and UGT1A8. M2 was preferentially catalysed by UGT1A10 and to a lesser extent by UGT1A1 and UGT1A8. The formation of M3, however, was mainly catalysed by UGT1A1 and UGT1A8. CONCLUSIONS Our results elucidate the importance of piceatannol glucuronidation in the human liver, which must be taken into account in humans after dietary intake of piceatannol.
Collapse
Affiliation(s)
- Michaela Miksits
- Department of Clinical Pharmacy and Diagnostics, University of Vienna, Vienna, Austria
| | | | | | | | | | | | | |
Collapse
|
28
|
Argikar UA, Gomez J, Ung D, Parkman HP, Nagar S. Identification of novel metoclopramide metabolites in humans: in vitro and in vivo studies. Drug Metab Dispos 2010; 38:1295-307. [PMID: 20423954 DOI: 10.1124/dmd.110.033357] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Metoclopramide (MCP) is frequently used to treat gastroparesis. Previous studies have documented MCP metabolism, but systematic structural identification of metabolites has not been performed. The aim of this study was to better understand MCP metabolism in humans. For examination of in vivo metabolism, a single oral 20-mg MCP dose was administered to eight healthy male volunteers, followed by complete urine collection over 24 h. In vitro incubations were performed in human liver microsomes (HLM) to characterize metabolism via cytochromes P450 and UDP-glucuronosyltransferases and in human liver cytosol for metabolism via sulfotransferases. Urine and subcellular incubations were analyzed for MCP metabolites on a mass spectrometer with accurate mass measurement capability. Five MCP metabolites were detected in vivo, and five additional metabolites were detected in vitro. The five metabolites of MCP identified both in vitro and in vivo were an N-O-glucuronide (M1), an N-sulfate (M2), a des-ethyl metabolite (M3), a hydroxylated metabolite (M4), and an oxidative deaminated metabolite (M5). To our knowledge, metabolites M1 and M4 have not been reported previously. M2 urinary levels varied 22-fold and M3 levels varied 16-fold among eight subjects. In vitro studies in HLM revealed the following additional metabolites: two ether glucuronides (M6 and M8), possibly on the phenyl ring after oxidation, an N-glucuronide (M7), a carbamic acid (M9), and a nitro metabolite (M10). Metabolites M6 to M10 have not been reported previously. In conclusion, this study describes the identification of MCP metabolites in vivo and in vitro in humans.
Collapse
Affiliation(s)
- Upendra A Argikar
- Novartis Institutes for Biomedical Research, Inc., Cambridge, Massachusetts, USA
| | | | | | | | | |
Collapse
|
29
|
Bourcier K, Hyland R, Kempshall S, Jones R, Maximilien J, Irvine N, Jones B. Investigation into UDP-Glucuronosyltransferase (UGT) Enzyme Kinetics of Imidazole- and Triazole-Containing Antifungal Drugs in Human Liver Microsomes and Recombinant UGT Enzymes. Drug Metab Dispos 2010; 38:923-9. [DOI: 10.1124/dmd.109.030676] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
|
30
|
Iwuchukwu O, Nagar S. Cis-resveratrol glucuronidation kinetics in human and recombinant UGT1A sources. Xenobiotica 2009; 40:102-8. [DOI: 10.3109/00498250903406754] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
31
|
Zhou J, Tracy TS, Remmel RP. Glucuronidation of dihydrotestosterone and trans-androsterone by recombinant UDP-glucuronosyltransferase (UGT) 1A4: evidence for multiple UGT1A4 aglycone binding sites. Drug Metab Dispos 2009; 38:431-40. [PMID: 20007295 DOI: 10.1124/dmd.109.028712] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
UDP-glucuronosyltransferase (UGT) 1A4-catalyzed glucuronidation is an important drug elimination pathway. Although atypical kinetic profiles (nonhyperbolic, non-Michaelis-Menten) of UGT1A4-catalyzed glucuronidation have been reported occasionally, systematic kinetic studies to explore the existence of multiple aglycone binding sites in UGT1A4 have not been conducted. To this end, two positional isomers, dihydrotestosterone (DHT) and trans-androsterone (t-AND), were used as probe substrates, and their glucuronidation kinetics with HEK293-expressed UGT1A4 were evaluated both alone and in the presence of a UGT1A4 substrate [tamoxifen (TAM) or lamotrigine (LTG)]. Coincubation with TAM, a high-affinity UGT1A4 substrate, resulted in a concentration-dependent activation/inhibition effect on DHT and t-AND glucuronidation, whereas LTG, a low-affinity UGT1A4 substrate, noncompetitively inhibited both processes. The glucuronidation kinetics of TAM were then evaluated both alone and in the presence of different concentrations of DHT or t-AND. TAM displayed substrate inhibition kinetics, suggesting that TAM may have two binding sites in UGT1A4. However, the substrate inhibition kinetic profile of TAM became more hyperbolic as the DHT or t-AND concentration was increased. Various two-site kinetic models adequately explained the interactions between TAM and DHT or TAM and t-AND. In addition, the effect of TAM on LTG glucuronidation was evaluated. In contrast to the mixed effect of TAM on DHT and t-AND glucuronidation, TAM inhibited LTG glucuronidation. Our results suggest that multiple aglycone binding sites exist within UGT1A4, which may result in atypical kinetics (both homotropic and heterotropic) in a substrate-dependent fashion.
Collapse
Affiliation(s)
- Jin Zhou
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | | | | |
Collapse
|
32
|
Hyland R, Osborne T, Payne A, Kempshall S, Logan YR, Ezzeddine K, Jones B. In vitro and in vivo glucuronidation of midazolam in humans. Br J Clin Pharmacol 2009; 67:445-54. [PMID: 19371318 DOI: 10.1111/j.1365-2125.2009.03386.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
AIMS Midazolam (MDZ) is a benzodiazepine used as a CYP3A4 probe in clinical and in vitro studies. A glucuronide metabolite of MDZ has been identified in vitro in human liver microsome (HLM) incubations. The primary aim of this study was to understand the in vivo relevance of this pathway. METHODS An authentic standard of N-glucuronide was generated from microsomal incubations and isolated using solid-phase extraction. The structure was confirmed using proton nuclear magnetic resonance (NMR) and (1)H-(13)C long range correlation experiments. The metabolite was quantified in vivo in human urine samples. Enzyme kinetic behaviour of the pathway was investigated in HLM and recombinant UGT (rUGT) enzymes. Additionally, preliminary experiments were performed with 1'-OH midazolam (1'-OH MDZ) and 4-OH-midazolam (4-OH MDZ) to investigate N-glucuronidation. RESULTS NMR data confirmed conjugation of midazolam N-glucuronide (MDZG) standard to be on the alpha-nitrogen of the imidazole ring. In vivo, MDZG in the urine accounted for 1-2% of the administered dose. In vitro incubations confirmed UGT1A4 as the enzyme of interest. The pathway exhibited atypical kinetics and a substrate inhibitory cooperative binding model was applied to determine K(m) (46 microM, 64 microM), V(max) (445 pmol min(-1) mg(-1), 427 pmol min(-1) mg(-1)) and K(i) (58 microM, 79 microM) in HLM and rUGT1A4, respectively. From incubations with HLM and rUGT enzymes, N-glucuronidation of 1'-OH MDZ and 4-OH MDZ is also inferred. CONCLUSIONS A more complete picture of MDZ metabolism and the enzymes involved has been elucidated. Direct N-glucuronidation of MDZ occurs in vivo. Pharmacokinetic modelling using Simcyp illustrates an increased role for UGT1A4 under CYP3A inhibited conditions.
Collapse
Affiliation(s)
- Ruth Hyland
- Pharmacokinetics Dynamics and Metabolism, Pfizer Global R&D, Ramsgate Road, Sandwich, Kent, CT13 9NJ, UK.
| | | | | | | | | | | | | |
Collapse
|
33
|
Iwuchukwu OF, Ajetunmobi J, Ung D, Nagar S. Characterizing the Effects of Common UDP Glucuronosyltransferase (UGT) 1A6 and UGT1A1 Polymorphisms on cis- and trans-Resveratrol Glucuronidation. Drug Metab Dispos 2009; 37:1726-32. [DOI: 10.1124/dmd.109.027391] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
34
|
Ung D, Nagar S. Trans-resveratrol-mediated inhibition of β-oestradiol conjugation in MCF-7 cells stably expressing human sulfotransferases SULT1A1 or SULT1E1, and human liver microsomes. Xenobiotica 2009; 39:72-9. [DOI: 10.1080/00498250802604082] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|