Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Dong-Xue Sun is active.

Publication


Featured researches published by Dong-Xue Sun.


Phytotherapy Research | 2010

Identification of cytochrome P450 (CYP) isoforms involved in the metabolism of corynoline, and assessment of its herb-drug interactions.

Zhong-Ze Fang; Yan-Yan Zhang; Guang-Bo Ge; Si-Cheng Liang; Dong-Xue Sun; Liang-Liang Zhu; Pei-Pei Dong; Yun-Feng Cao; Ling Yang

Corynoline, an isoquinoline alkaloid isolated from the genus Corydalis, has been demonstrated to show multiple pharmacological effects including inhibition of acetylcholinesterase, inhibition of cell adhesion, fungitoxic and cytotoxic activity. The present study focused on its metabolism and metabolism‐based herb–drug interactions. After corynoline was incubated with human liver microsomes (HLMs) in the presence of NADPH, two metabolites (M‐1 and M‐2) were formed. Chemical inhibition experiments and assays with recombinant CYP isoforms showed that CYP2C9 was mainly involved in the formation of M‐1 and CYP3A4 mainly catalysed the production of M‐2. Among seven major CYP isoforms tested, corynoline showed strong inhibitory effects on the activities of CYP3A4 and CYP2C9, with an IC50 of 3.3 ± 0.9 µm and 31.5 ± 0.5 µm, respectively. Kinetic analysis showed that inhibition of CYP3A4 by corynoline was best fit to a noncompetitive manner with Ki of 3.2 µm, while inhibition of CYP2C9 by corynoline was best fit to a competitive manner with Ki of 6.3 µm. Additionally, corynoline exhibited time‐dependent inhibition (TDI) toward CYP3A4. The inactivation kinetic parameters (KI and kinact) were calculated to be 6.8 µm and 0.07 min‐1, respectively. These data are of significance for the application of corynoline and corynoline‐containing herbs. Copyright


Xenobiotica | 2010

CYP3A catalyses schizandrin biotransformation in human, minipig and rat liver microsomes

Yun-Feng Cao; Yihua Zhang; J. Li; Guang-Bo Ge; D. Hu; H.-X. Liu; T. Huang; Yingli Wang; Zhong-Ze Fang; Dong-Xue Sun; Hong Huo; Jun Yin; Ling Yang

Schizandrin is recognized as the major absorbed effective constituent of Fructus schisandrae, which is extensively applied in Chinese medicinal formula. The present study aimed to profile the phase I metabolites of schizandrin and identify the cytochrome P450 (CYP) isoforms involved. After schizandrin was incubated with human liver microsomes, three metabolites were isolated by high-performance liquid chromatography (HPLC) and their structures were identified to be 8(R)-hydroxyl-schizandrin, 2-demethyl-8(R)-hydroxyl-schizandrin, 3-demethyl-8(R)-hydroxyl-schizandrin, by liquid chromatography-mass spectrometry (LC-MS), 1H-nuclear magnetic resonance (NMR), and 13C-NMR, respectively. A combination of correlation analysis, chemical inhibition studies, assays with recombinant CYPs, and enzyme kinetics indicated that CYP3A4 was the main hepatic isoform that cleared schizandrin. Rat and minipig liver microsomes were included when evaluating species differences, and the results showed little difference among the species. In conclusion, CYP3A4 plays a major role in the biotransformation of schizandrin in human liver microsomes. Minipig and rat could be surrogate models for man in schizandrin pharmacokinetic studies. Better knowledge of schizandrin’s metabolic pathway could provide the vital information for understanding the pharmacokinetic behaviours of schizandrin contained in Chinese medicinal formula.


Diabetes | 2017

An Intestinal Farnesoid X Receptor–Ceramide Signaling Axis Modulates Hepatic Gluconeogenesis in Mice

Cen Xie; Changtao Jiang; Jingmin Shi; Xiaoxia Gao; Dong-Xue Sun; Lulu Sun; Ting Wang; Shogo Takahashi; Mallappa Anitha; Kristopher W. Krausz; Andrew D. Patterson; Frank J. Gonzalez

Increasing evidence supports the view that intestinal farnesoid X receptor (FXR) is involved in glucose tolerance and that FXR signaling can be profoundly impacted by the gut microbiota. Selective manipulation of the gut microbiota–FXR signaling axis was reported to significantly impact glucose intolerance, but the precise molecular mechanism remains largely unknown. Here, caffeic acid phenethyl ester (CAPE), an over-the-counter dietary supplement and an inhibitor of bacterial bile salt hydrolase, increased levels of intestinal tauro-β-muricholic acid, which selectively suppresses intestinal FXR signaling. Intestinal FXR inhibition decreased ceramide levels by suppressing expression of genes involved in ceramide synthesis specifically in the intestinal ileum epithelial cells. The lower serum ceramides mediated decreased hepatic mitochondrial acetyl-CoA levels and pyruvate carboxylase (PC) activities and attenuated hepatic gluconeogenesis, independent of body weight change and hepatic insulin signaling in vivo; this was reversed by treatment of mice with ceramides or the FXR agonist GW4064. Ceramides substantially attenuated mitochondrial citrate synthase activities primarily through the induction of endoplasmic reticulum stress, which triggers increased hepatic mitochondrial acetyl-CoA levels and PC activities. These results reveal a mechanism by which the dietary supplement CAPE and intestinal FXR regulates hepatic gluconeogenesis and suggest that inhibiting intestinal FXR is a strategy for treating hyperglycemia.


Phytotherapy Research | 2010

Reversible inhibition of three important human liver cytochrome p450 enzymes by tiliroside

Dong-Xue Sun; Jin-Cai Lu; Zhong-Ze Fang; Yan-Yan Zhang; Yun-Feng Cao; Yu-Xi Mao; Liang-Liang Zhu; Jun Yin; Ling Yang

Tiliroside, an active flavonoid extensively found in many medicinal plants including Helichrysum italicum, Geranium mexicanum and Helianthemum glomeratum, has been demonstrated to exert multiple biological effects including antiinflammatory, antimicrobial, antioxidant and antitumor activities. Cytochrome P450 (CYP) enzymes play an important role in the Phase I oxidation metabolism of a wide range of xenobiotics and inhibition of CYP isoforms might influence the elimination of drugs and induce serious adverse drug response. The inhibition of seven CYP isoforms (CYP3A4, CYP1A2, CYP2A6, CYP2D6, CYP2C9, CYP2C8 and CYP2E1) by tiliroside was investigated using in vitro human liver microsomal incubation assays. The results showed that tiliroside strongly inhibited the activity of CYP3A4 (IC50 = 9.0 ± 1.7 μm), CYP2C8 (IC50 = 12.1 ± 0.9 μm) and CYP2C9 (IC50 = 10.2 ± 0.9 μm) with other CYP isoforms negligibly influenced. Further kinetic analysis showed that inhibition of these three CYP isoforms by tiliroside is best fit to a competitive way. The Ki value was calculated to be 5.5 μm, 3.3 μm, 9.4 μm for CYP3A4, CYP2C9 and CYP2C8, respectively. The relatively low Ki values suggested that tiliroside might induce drug–drug interactions with many clinically used drugs which are mainly metabolized by these three CYP isoforms. Therefore, attention should be given to the probable drug–drug interaction between tiliroside‐containing herbs and substrates of CYP3A4, CYP2C9 and CYP2C8. Copyright


Phytotherapy Research | 2010

Inhibitory effects of curcumenol on human liver cytochrome P450 enzymes.

Dong-Xue Sun; Zhong-Ze Fang; Yan-Yan Zhang; Yun-Feng Cao; Ling Yang; Jun Yin

Curcumenol, one of the major components of Zedoary turmeric oil, has been widely used to treat cancer and inflammation. As an antibiotic or anticancer drug, curcumenol is highly likely to be used in combination with various synthetic drugs in most cases, thus it is necessary to evaluate potential pharmacokinetic drug‐drug interactions induced by curcumenol. In this study, the inhibitory effects of curcumenol on seven CYP isoforms were investigated, and the results demonstrated that only CYP3A4 was strongly inhibited (IC50 = 12.6 ± 1.3 μM). Kinetic analysis showed the inhibition type was competitive with Ki value of 10.8 μM. Time‐ and NADPH‐dependent inhibitions were also investigated to show curcumenol is not a mechanism‐based inhibitor. Employing these in vitro data and maximum plasma concentration of curcumenol in human predicted from beagle dogs in vivo pharmacokinetic data, the change in AUC of victim drugs was predicted to be 0.4%, which suggested that curcumenol may be safely used without inducing metabolic drug‐drug interaction through P450 inhibition. Nevertheless, due to the limited pharmacokinetic data available for curcumenol in humans, it is still not possible to evaluate its potential clinical effects on human patients from in vitro data. Thus, the magnitude of drug‐drug interaction (DDI) induced by curcumenol warrants further investigation. Copyright


Xenobiotica | 2016

Inhibition behavior of fructus psoraleae’s ingredients towards human carboxylesterase 1 (hCES1)

Dong-Xue Sun; Guang-Bo Ge; Pei-Pei Dong; Yun-Feng Cao; Zhi-Wei Fu; Rui-Xue Ran; Xue Wu; Yan-Yan Zhang; Hui-Ming Hua; Zhenying Zhao; Zhong-Ze Fang

Abstract 1. Fructus psoraleae (FP) is the dried ripe seeds of Psoralea corylifolia L. (Fabaceae) widely used in Asia, and has been reported to exert important biochemical and pharmacological activities. The adverse effects of FP remain unclear. The present study aims to determine the inhibition of human carboxylesterase 1 (CES1) by FP’s major ingredients, including neobavaisoflavone, corylifolinin, coryfolin, psoralidin, corylin and bavachinin. 2. The probe substrate of CES1 2-(2-benzoyl-3-methoxyphenyl) benzothiazole (BMBT) was derived from 2-(2-hydroxy-3-methoxyphenyl) benzothiazole (HMBT), and human liver microsomes (HLMs)-catalyzed BMBT metabolism was used to phenotype the activity of CES1. In silico docking method was employed to explain the inhibition mechanism. 3. All the tested compounds exerted strong inhibition towards the activity of CES1 in a concentration-dependent behavior. Furthermore, the inhibition kinetics was determined for the inhibition of neobavaisoflavone, corylifolinin, coryfolin, corylin and bavachinin towards CES1. Both Dixon and Lineweaver–Burk plots showed that neobavaisoflavone, corylifolinin, coryfolin and corylin noncompetitively inhibited the activity of CES1, and bavachinin competitively inhibited the activity of CES1. The inhibition kinetic parameters (Ki) were calculated to be 5.3, 9.4, 1.9, 0.7 and 0.5 μM for neobavaisoflavone, corylifolinin, coryfolin, corylin and bavachinin, respectively. In conclusion, the inhibition behavior of CES1 by the FP’s constituents was given in this article, indicating the possible adverse effects of FP through the disrupting CES1-catalyzed metabolism of endogenous substances and xenobiotics.


Toxicology and Applied Pharmacology | 2016

Irinotecan (CPT-11)-induced elevation of bile acids potentiates suppression of IL-10 expression.

Zhong-Ze Fang; Dunfang Zhang; Yun-Feng Cao; Cen Xie; Dan Lu; Dong-Xue Sun; Naoki Tanaka; Changtao Jiang; Qianming Chen; Yu Chen; Haina Wang; Frank J. Gonzalez

Irinotecan (CPT-11) is a first-line anti-colon cancer drug, however; CPT-11-induced toxicity remains a key factor limiting its clinical application. To search for clues to the mechanism of CPT-11-induced toxicity, metabolomics was applied using ultra-performance liquid chromatography coupled with electrospray ionization quadrupole time-of-flight mass spectrometry. Intraperitoneal injection of 50 mg/kg of CPT-11 induced loss of body weight, and intestine toxicity. Changes in gallbladder morphology suggested alterations in bile acid metabolism, as revealed at the molecular level by analysis of the liver, bile, and ileum metabolomes between the vehicle-treated control group and the CPT-11-treated group. Analysis of immune cell populations further showed that CPT-11 treatment significantly decreased the IL-10-producing CD4 T cell frequency in intestinal lamina propria lymphocytes, but not in spleen or mesenteric lymph nodes. In vitro cell culture studies showed that the addition of bile acids deoxycholic acid and taurodeoxycholic acid accelerated the CPT-11-induced suppression of IL-10 secretion by activated CD4(+) naive T cells isolated from mouse splenocytes. These results showed that CPT-11 treatment caused metabolic changes in the composition of bile acids that altered CPT-11-induced suppression of IL-10 expression.


Nature Medicine | 2017

Activation of intestinal hypoxia-inducible factor 2α during obesity contributes to hepatic steatosis

Cen Xie; Tomoki Yagai; Yuhong Luo; Xianyi Liang; Tao Chen; Qiong Wang; Dong-Xue Sun; Jie Zhao; Sadeesh K. Ramakrishnan; Lulu Sun; Chunmei Jiang; Xiang Xue; Yuan Tian; Kristopher W. Krausz; Andrew D. Patterson; Yatrik M. Shah; Yue Wu; Changtao Jiang; Frank J. Gonzalez

Nonalcoholic fatty liver disease is becoming the most common chronic liver disease in Western countries, and limited therapeutic options are available. Here we uncovered a role for intestinal hypoxia-inducible factor (HIF) in hepatic steatosis. Human-intestine biopsies from individuals with or without obesity revealed that intestinal HIF-2α signaling was positively correlated with body-mass index and hepatic toxicity. The causality of this correlation was verified in mice with an intestine-specific disruption of Hif2a, in which high-fat-diet-induced hepatic steatosis and obesity were substantially lower as compared to control mice. PT2385, a HIF-2α-specific inhibitor, had preventive and therapeutic effects on metabolic disorders that were dependent on intestine HIF-2α. Intestine HIF-2α inhibition markedly reduced intestine and serum ceramide levels. Mechanistically, intestine HIF-2α regulates ceramide metabolism mainly from the salvage pathway, by positively regulating the expression of Neu3, the gene encoding neuraminidase 3. These results suggest that intestinal HIF-2α could be a viable target for hepatic steatosis therapy.


Biochemical Pharmacology | 2016

The antiandrogen flutamide is a novel aryl hydrocarbon receptor ligand that disrupts bile acid homeostasis in mice through induction of Abcc4.

Xiaoxia Gao; Cen Xie; Yuanyuan Wang; Yuhong Luo; Tomoki Yagai; Dong-Xue Sun; Xuemei Qin; Kristopher W. Krausz; Frank J. Gonzalez

Flutamide (FLU), an oral, nonsteroidal antiandrogen drug used in the treatment of prostate cancer, is associated with idiosyncratic hepatotoxicity that sometimes causes severe liver damage, including cholestasis, jaundice, and liver necrosis. To understand the mechanism of toxicity, a combination of aryl hydrocarbon receptor (Ahr)-deficient (Ahr-/-) mice, primary hepatocytes, luciferase reporter gene assays, in silico ligand docking and ultra-performance chromatography-quadrupole time-of-flight mass spectrometry-based metabolomics was used. A significant increase of liver weights, and liver and serum bile acid levels was observed after FLU treatment, indicating hepatomegaly and disrupted bile acid homeostasis. Expression of the AhR gene battery was markedly increased in livers of wild-type mice Ahr+/+ treated with FLU, while no change was noted in Ahr-/- mice. Messenger RNAs encoded by AhR target genes were induced in primary mouse hepatocytes cultured with FLU, which confirmed the in vivo results. Ligand-docking analysis further predicted that FLU is an AhR agonist ligand which was confirmed by luciferase reporter gene assays. Multivariate data analysis showed that bile acids were responsible for the separation of vehicle- and FLU-treated Ahr+/+ mice, while there was no separation in Ahr-/- mice. Expression of mRNA encoding the bile acid transporter ABCC4 was increased and farnesoid X receptor signaling was inhibited in the livers of Ahr+/+ mice, but not in Ahr-/- mice treated with FLU, in agreement with the observed downstream metabolic alterations. These findings provide new insights into the mechanism of liver injury caused by FLU treatment involving activation of AhR and the alterations of bile acid homeostasis, which could guide clinical application.


Chirality | 2015

Enantioselective Inhibition of Carprofen Towards UDP-glucuronosyltransferase (UGT) 2B7

Zhong-Ze Fang; Haina Wang; Yun-Feng Cao; Dong-Xue Sun; Li-Xuan Wang; Mo Hong; Ting Huang; Jian-Xing Chen; Jia Zeng

UDP-glucuronosyltransferases (UGTs)-catalyzed glucuronidation conjugation reaction plays an important role in the elimination of many important clinical drugs and endogenous substances. The present study aims to investigate the enantioselective inhibition of carprofen towards UGT isoforms. In vitro a recombinant UGT isoforms-catalyzed 4-methylumbelliferone (4-MU) glucuronidation incubation mixture was used to screen the inhibition potential of (R)-carprofen and (S)-carprofen towards multiple UGT isoforms. The results showed that (S)-carprofen exhibited stronger inhibition potential than (R)-carprofen towards UGT2B7. However, no significant difference was observed for the inhibition of (R)-carprofen and (S)-carprofen towards other UGT isoforms. Furthermore, the inhibition kinetic behavior was compared for the inhibition of (S)-carprofen and (R)-carprofen towards UGT2B7. A Lineweaver-Burk plot showed that both (S)-carprofen and (R)-carprofen exhibited competitive inhibition towards UGT2B7-catalyzed 4-MU glucuronidation. The inhibition kinetic parameter (Ki ) was calculated to be 7.0 μM and 31.1 μM for (S)-carprofen and (R)-carprofen, respectively. Based on the standard for drug-drug interaction, the threshold for (S)-carprofen and (R)-carprofen to induce a drug-drug interaction is 0.7 μM and 3.1 μM, respectively. In conclusion, enantioselective inhibition of carprofen towards UDP-glucuronosyltransferase (UGT) 2B7 was demonstrated in the present study. Using the in vitro inhibition kinetic parameter, the concentration threshold of (S)-carprofen and (R)-carprofen to possibly induce the drug-drug interaction was obtained. Therefore, clinical monitoring of the plasma concentration of (S)-carprofen is more important than (R)-carprofen to avoid a possible drug-drug interaction between carprofen and the drugs mainly undergoing UGT2B7-catalyzed metabolism.

Collaboration


Dive into the Dong-Xue Sun's collaboration.

Top Co-Authors

Avatar

Yun-Feng Cao

Dalian Institute of Chemical Physics

View shared research outputs
Top Co-Authors

Avatar

Zhong-Ze Fang

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Frank J. Gonzalez

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Ling Yang

Dalian Institute of Chemical Physics

View shared research outputs
Top Co-Authors

Avatar

Cen Xie

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Jun Yin

Shenyang Pharmaceutical University

View shared research outputs
Top Co-Authors

Avatar

Liang-Liang Zhu

Dalian Institute of Chemical Physics

View shared research outputs
Top Co-Authors

Avatar

Yan-Yan Zhang

Dalian Institute of Chemical Physics

View shared research outputs
Top Co-Authors

Avatar

Kristopher W. Krausz

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge