Network


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

Hotspot


Dive into the research topics where Pei-Pei Dong is active.

Publication


Featured researches published by Pei-Pei Dong.


Drug Metabolism and Disposition | 2012

Characterization of Hepatic and Intestinal Glucuronidation of Magnolol: Application of the Relative Activity Factor Approach to Decipher the Contributions of Multiple UDP-Glucuronosyltransferase Isoforms

Liang-Liang Zhu; Guang-Bo Ge; Hongbo Zhang; Hui-Xin Liu; Guiyuan He; Si-Cheng Liang; Yan-Yan Zhang; Zhong-Ze Fang; Pei-Pei Dong; Moshe Finel; Ling Yang

Magnolol is a food additive that is often found in mints and gums. Human exposure to this compound can reach a high dose; thus, characterization of magnolol disposition in humans is very important. Previous studies indicated that magnolol can undergo extensive glucuronidation in humans in vivo. In this study, in vitro assays were used to characterize the glucuronidation pathway in human liver and intestine. Assays with recombinant human UDP-glucuronosyltransferase enzymes (UGTs) revealed that multiple UGT isoforms were involved in magnolol glucuronidation, including UGT1A1, -1A3, -1A7, -1A8, -1A9, -1A10, and -2B7. Magnolol glucuronidation by human liver microsomes (HLM), human intestine microsomes (HIM), and most recombinant UGTs exhibited strong substrate inhibition kinetics. The degree of substrate inhibition was relatively low in the case of UGT1A10, whereas the reaction catalyzed by UGT1A9 followed biphasic kinetics. Chemical inhibition studies and the relative activity factor (RAF) approach were used to identify the individual UGTs that played important roles in magnolol glucuronidation in HLM and HIM. The results indicate that UGT2B7 is mainly responsible for the reaction in HLM, whereas UGT2B7 and UGT1A10 are significant contributors in HIM. In summary, the current study clarifies the glucuronidation pathway of magnolol and demonstrates that the RAF approach can be used as an efficient method for deciphering the roles of individual UGTs in a given glucuronidation pathway in the native tissue that is catalyzed by multiple isoforms with variable and atypical kinetics.


Xenobiotica | 2012

Potent and selective inhibition of magnolol on catalytic activities of UGT1A7 and 1A9

Liang-Liang Zhu; Guang-Bo Ge; Yong Liu; Guiyuan He; Si-Cheng Liang; Zhong-Ze Fang; Pei-Pei Dong; Yun-Feng Cao; Ling Yang

Human exposure to magnolol can reach a high dose in daily life. Our previous studies indicated that magnolol showed high affinities to several UDP-glucuronosyltransferases (UGTs) This study was designed to examine the in vitro inhibitory effects of magnolol on UGTs, and further to evaluate the possibility of the in vivo inhibition that might happen. Assays with recombinant UGTs and human liver microsomes (HLM) indicated that magnolol (10 µM) can selectively inhibit activities of UGT1A9 and extra-hepatic UGT1A7. Inhibition of magnolol on UGT1A7 followed competitive inhibition mechanism, while the inhibition on UGT1A9 obeyed either competitive or mixed inhibition mechanism, depending on substrates. The Ki values for UGT1A7 and 1A9 are all in nanomolar ranges, lower than possible magnolol concentrations in human gut lumen and blood, indicating the in vivo inhibition on these two enzymes would likely occur. In conclusion, UGT1A7 and 1A9 can be strongly inhibited by magnolol, raising the alarm for safe application of magnolol and traditional Chinese medicines containing magnolol. Additionally, given that UGT1A7 is an extra-hepatic enzyme, magnolol can serve as a selective UGT1A9 inhibitor that will act as a new useful tool in future hepatic glucuronidation phenotyping.


Journal of Natural Products | 2015

Protostane Triterpenoids from the Rhizome of Alisma orientale Exhibit Inhibitory Effects on Human Carboxylesterase 2

Zhen-Peng Mai; Kun Zhou; Guang-Bo Ge; Chao Wang; Xiaokui Huo; Pei-Pei Dong; Sa Deng; Bao-Jing Zhang; Hou-Li Zhang; Shan-Shan Huang; Xiaochi Ma

Twelve new and 10 known protostane triterpenoids were isolated from the rhizome of Alisma orientale. Their structures were elucidated based on physical data analyses, including UV, HRESIMS, NMR experiments ((1)H, (13)C NMR, (1)H-(1)H COSY, HSQC, HMBC, and NOESY), and induced electronic circular dichroism. New compounds 1-12 were classified as protostanes (1-10), 29-norprotostane (11), and 24-norprotostane (12) by structure analyses. Furthermore, the inhibitory effects on human carboxylesterases (hCE-1, hCE-2) of compounds 1-22 were evaluated. Compounds 2, 6, 9, and 11 showed moderate inhibitory activities and were selective toward hCE-2 enzymes, with IC50 values of 8.68, 4.72, 4.58, and 2.02 μM, respectively. The inhibition kinetics of compound 11 toward hCE-2 were established, and the Ki value was determined as 1.76 μM using a mixed inhibition model. The interaction of bioactive compound 11 with hCE-2 was shown using molecular docking.


Phytotherapy Research | 2011

Cycloartane triterpenoids from Cimicifuga yunnanensis induce apoptosis of breast cancer cells (MCF7) via p53-dependent mitochondrial signaling pathway

Zhong-Ze Fang; Yin Nian; Wei Li; Jing-Jing Wu; Guang-Bo Ge; Pei-Pei Dong; Yan-Yan Zhang; Ming-Hua Qiu; Lei Liu; Ling Yang

The present study was carried out to investigate the antitumor activity of five cycloartane triterpenoids isolated from Cimicifuga yunnanensis on the breast cancer cell line MCF7 and its corresponding drug resistant subline R‐MCF7, including cimigenol‐3‐O‐β‐d‐xylopyranoside (compound 1), 25‐O‐acetylcimigenol‐3‐O‐β‐d‐xylopyranoside (compound 2), 25‐chlorodeoxycimigenol‐3‐O‐β‐d‐xylopyranoside (compound 3), 25‐O‐acetylcimigenol‐3‐O‐α‐l‐arabinopyranoside (compound 4) and 23‐O‐acetylcimigenol‐3‐O‐β‐d‐xylopyranoside (compound 5). The results showed that compounds 2–5 have relatively high antitumor activity on both MCF7 and R‐MCF7 cells. The involvement of apoptosis as a major cause of cycloartane triterpenoids‐induced cell death was further confirmed. The results of RT‐PCR showed that compounds 2–5 increased the expression of p53 and bax, which led to the loss of mitochondrial potential and then resulted in the activation of caspase‐7. These findings collectively demonstrated that compounds 2–5 induced apoptosis of MCF7 via p53‐dependent mitochondrial pathway. Copyright


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


Food and Chemical Toxicology | 2013

Inhibitory effects of sanguinarine on human liver cytochrome P450 enzymes

Xiao-Yi Qi; Si-Cheng Liang; Guang-Bo Ge; Yong Liu; Pei-Pei Dong; Jiang-Wei Zhang; Ao-Xue Wang; Jie Hou; Liang-Liang Zhu; Ling Yang; Cai-Xia Tu

Sanguinarine (SAG) has been recognized as an anticancer drug candidate. However, the drug-drug interactions (DDI) potential for SAG via the inhibition against human cytochrome P450 (CYP) enzymes remains unclear. In the present study, the inhibitory effects of SAG on seven major human CYP isoforms 1A2, 2A6, 2E1, 2D6, 2C8, 2C9 and 3A4 were investigated with human liver microsomes (HLM). The results showed that SAG was a potent noncompetitive inhibitor of CYP2C8 activity (Ki=8.9 μM), and competitive inhibitor of CYP1A2, CYP2C9 and CYP3A4 activities (Ki=2.7, 3.8 and 2.0 μM, respectively). Furthermore, SAG exhibited time- and NADPH-dependent inhibition towards CYP1A2 and CYP3A4 with KI/kinact values of 13.3/0.087 and 5.58/0.029 min(-1) μM(-1), respectively. Weak inhibition of SAG against CYP2E1, CYP2D6 and CYP2A6 was also observed. In vitro-in vivo extrapolation (IV-IVE) from HLM data showed that more than 35.9% of CYP1A2, CYP2C9, CYP2C8 and CYP3A4 activities in vivo could be inhibited by SAG, suggesting that harmful DDIs could occur when SAG or its medical preparations are co-administered with drugs primarily cleared by these CYP isoforms. Further in vivo studies are needed to evaluate the clinical significance of the data presented herein.


Food Chemistry | 2013

Biotransformation of imperatorin by Penicillium janthinellum. Anti-osteoporosis activities of its metabolites

Xia Lv; Dan Liu; Jie Hou; Pei-Pei Dong; Libin Zhan; Li Wang; Sa Deng; Changyuan Wang; Jihou Yao; Xiaohong Shu; Kexin Liu; Xiaochi Ma

Imperatorin (IMP) is a major constituent of many herbal medicines and possesses anti-osteoporosis activity. The present research work aimed to study the biotransformation processes of IMP and evaluated the anti-osteoporosis activity of the transformed metabolites. Among 18 strains of filamentous fungi screened, Penicillium janthinellum AS 3.510 exhibited good capability to metabolise IMP to the new derivatives. Ten transformed products were isolated and purified, and their structures were identified accurately based on spectroscopic data. Eight metabolites (2-8 and 10) were novel and previously unreported. The major biotransformation reactions involved hydroxylation of the prenyloxy side-chain and the lactone ring-opening reaction of furocoumarin skeleton. In addition, anti-osteoporosis activities of all products (1-10) were evaluated using MC3T3-E1 cells. The results showed that products 5 and 8 had the best bioactivities in increasing MC3T3-E1 cell growth. These products could be used in future therapeutic regimens for treating osteoporosis.


Acta Pharmacologica Sinica | 2011

Substrate-dependent modulation of the catalytic activity of CYP3A by erlotinib

Pei-Pei Dong; Zhong-Ze Fang; Yan-Yan Zhang; Guang-Bo Ge; Yu-Xi Mao; Liang-Liang Zhu; Yan-Qing Qu; Wei Li; Li-Ming Wang; Chang-xiao Liu; Ling Yang

Aim:To ascertain the effects of erlotinib on CYP3A, to investigate the amplitude and kinetics of erlotinib-mediated inhibition of seven major CYP isoforms in human liver microsomes (HLMs) for evaluating the magnitude of erlotinib in drug-drug interaction in vivo.Methods:The activities of 7 major CYP isoforms (CYP1A2, CYP2A6, CYP3A, CYP2C9, CYP2D6, CYP2C8, and CYP2E1) were assessed in HLMs using HPLC or UFLC analysis. A two-step incubation method was used to examine the time-dependent inhibition of erlotinib on CYP3A.Results:The activity of CYP2C8 was inhibited with an IC50 value of 6.17±2.0 μmol/L. Erlotinib stimulated the midazolam 1′-hydroxy reaction, but inhibited the formation of 6β-hydroxytestosterone and oxidized nifedipine. Inhibition of CYP3A by erlotinib was substrate-dependent: the IC50 values for inhibiting testosterone 6β-hydroxylation and nifedipine metabolism were 31.3±8.0 and 20.5±5.3 μmol/L, respectively. Erlotinib also exhibited the time-dependent inhibition on CYP3A, regardless of the probe substrate used: the value of KI and kinact were 6.3 μmol/L and 0.035 min−1 for midazolam; 9.0 μmol/L and 0.045 min−1 for testosterone; and 10.1 μmol/L and 0.058 min−1 for nifedipine.Conclusion:The inhibition of CYP3A by erlotinib was substrate-dependent, while its time-dependent inhibition on CYP3A was substrate-independent. The time-dependent inhibition of CYP3A may be a possible cause of drug-drug interaction, suggesting that attention should be paid to the evaluation of erlotinibs safety, especially in the context of combination therapy.


Molecular Cancer | 2014

Gamabufotalin, a bufadienolide compound from toad venom, suppresses COX-2 expression through targeting IKKβ/NF-κB signaling pathway in lung cancer cells

Zhenlong Yu; Wei Guo; Xiaochi Ma; Baojing Zhang; Pei-Pei Dong; Lin Huang; Xiuli Wang; Chao Wang; Xiaokui Huo; Wendan Yu; Canhui Yi; Yao Xiao; Wenjing Yang; Yu Qin; Yuhui Yuan; Songshu Meng; Quentin Liu; Wuguo Deng

BackgroundGamabufotalin (CS-6), a major bufadienolide of Chansu, has been used for cancer therapy due to its desirable metabolic stability and less adverse effect. However, the underlying mechanism of CS-6 involved in anti-tumor activity remains poorly understood.MethodsThe biological functions of gamabufotalin (CS-6) were investigated by migration, colony formation and apoptosis assays in NSCLC cells. The nuclear localization and interaction between transcriptional co-activator p300 and NF-κB p50/p65 and their binding to COX-2 promoter were analyzed after treatment with CS-6. Molecular docking study was used to simulate the interaction of CS-6 with IKKβ. The in vivo anti-tumor efficacy of CS-6 was also analyzed in xenografts nude mice. Western blot was used to detect the protein expression level.ResultsGamabufotalin (CS-6) strongly suppressed COX-2 expression by inhibiting the phosphorylation of IKKβ via targeting the ATP-binding site, thereby abrogating NF-κB binding and p300 recruitment to COX-2 promoter. In addition, CS-6 induced apoptosis by activating the cytochrome c and caspase-dependent apoptotic pathway. Moreover, CS-6 markedly down-regulated the protein levels of COX-2 and phosphorylated p65 NF-κB in tumor tissues of the xenograft mice, and inhibited tumor weight and size.ConclusionsOur study provides pharmacological evidence that CS-6 exhibits potential use in the treatment of COX-2-mediated diseases such as lung cancer.


Journal of Lipid Research | 2013

A model of in vitro UDP-glucuronosyltransferase inhibition by bile acids predicts possible metabolic disorders

Zhong-Ze Fang; Rong-Rong He; Yun-Feng Cao; Naoki Tanaka; Changtao Jiang; Kristopher W. Krausz; Yunpeng Qi; Pei-Pei Dong; Chun-Zhi Ai; Xiao-Yu Sun; Mo Hong; Guang-Bo Ge; Frank J. Gonzalez; Xiaochi Ma; Hong-Zhi Sun

Increased levels of bile acids (BAs) due to the various hepatic diseases could interfere with the metabolism of xenobiotics, such as drugs, and endobiotics including steroid hormones. UDP-glucuronosyltransferases (UGTs) are involved in the conjugation and elimination of many xenobiotics and endogenous compounds. The present study sought to investigate the potential for inhibition of UGT enzymes by BAs. The results showed that taurolithocholic acid (TLCA) exhibited the strongest inhibition toward UGTs, followed by lithocholic acid. Structure-UGT inhibition relationships of BAs were examined and in vitro-in vivo extrapolation performed by using in vitro inhibition kinetic parameters (Ki) in combination with calculated in vivo levels of TLCA. Substitution of a hydrogen with a hydroxyl group in the R1, R3, R4, R5 sites of BAs significantly weakens their inhibition ability toward most UGTs. The in vivo inhibition by TLCA toward UGT forms was determined with following orders of potency: UGT1A4 > UGT2B7 > UGT1A3 > UGT1A1 ∼ UGT1A7 ∼ UGT1A10 ∼ UGT2B15. In conclusion, these studies suggest that disrupted homeostasis of BAs, notably taurolithocholic acid, found in various diseases such as cholestasis, could lead to altered metabolism of xenobiotics and endobiotics through inhibition of UGT enzymes.

Collaboration


Dive into the Pei-Pei Dong's collaboration.

Top Co-Authors

Avatar

Xiaochi Ma

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Xiaokui Huo

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Chao Wang

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Baojing Zhang

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Sa Deng

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Ling Yang

Dalian Institute of Chemical Physics

View shared research outputs
Top Co-Authors

Avatar

Shan-Shan Huang

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Xiangge Tian

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Yan-Yan Zhang

Liaoning Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge