Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Arch Toxicol ; 98(5): 1457-1467, 2024 May.
Article in English | MEDLINE | ID: mdl-38492097

ABSTRACT

Cytochrome P450 (P450)-mediated bioactivation, which can lead to the hepatotoxicity through the formation of reactive metabolites (RMs), has been regarded as the major problem of drug failures. Herein, we purposed to establish machine learning models to predict the bioactivation of P450. On the basis of the literature-derived bioactivation dataset, models for Benzene ring, Nitrogen heterocycle and Sulfur heterocycle were developed with machine learning methods, i.e., Random Forest, Random Subspace, SVM and Naïve Bayes. The models were assessed by metrics like "Precision", "Recall", "F-Measure", "AUC" (Area Under the Curve), etc. Random Forest algorithms illustrated the best predictability, with nice AUC values of 0.949, 0.973 and 0.958 for the test sets of Benzene ring, Nitrogen heterocycle and Sulfur heterocycle models, respectively. 2D descriptors like topological indices, 2D autocorrelations and Burden eigenvalues, etc. contributed most to the models. Furthermore, the models were applied to predict the occurrence of bioactivation of an external verification set. Drugs like selpercatinib, glafenine, encorafenib, etc. were predicted to undergo bioactivation into toxic RMs. In vitro, IC50 shift experiment was performed to assess the potential of bioactivation to validate the prediction. Encorafenib and tirbanibulin were observed of bioactivation potential with shifts of 3-6 folds or so. Overall, this study provided a reliable and robust strategy to predict the P450-mediated bioactivation, which will be helpful to the assessment of adverse drug reactions (ADRs) in clinic and the design of new candidates with lower toxicities.


Subject(s)
Benzene , Carbamates , Drug-Related Side Effects and Adverse Reactions , Sulfonamides , Humans , Bayes Theorem , Cytochrome P-450 Enzyme System/metabolism , Machine Learning , Sulfur , Nitrogen
2.
J Steroid Biochem Mol Biol ; 225: 106196, 2023 01.
Article in English | MEDLINE | ID: mdl-36181991

ABSTRACT

ß-estradiol (ß-E2) and α-estradiol (α-E2) act as an endo- and an exon-estrogen in humans, respectively. There is a structural variation in C17-OH configuration of the two estrogens. UDP-glucuronosyltransferases (UGT) are responsible for termination of activities of a variety of endogenous hormones, clinical drugs, and environmental toxicants. The current study was conducted to investigate the effects of the two estrogens towards catalytic activities of UGTs. It was found that ß-E2 could decrease activities of UGT1A9, - 2B4 and - 2B7, with Ki values of a few micro-molars. ß-E2 could additionally accelerate the activity of UGT2B17 via promoting enzyme-substrate binding and increasing the turn over number. Comparatively, α-E2 displayed much stronger inhibitory potentials towards UGT2B7 and - 2B4, but showed little influence to UGT1A9 and - 2B17. The Ki values for inhibition of UGT2B7 in glucuronidation of different substrates by α-E2 were in a nanomolar range that is only about 1/100-1/50 of ß-E2. UGT2B7 structural model was fatherly constructed to explore the mechanism underlying dramatically different inhibition selectivity of the two estrogens. Compared to ß-E2, α-E2 formed more hydrophobic and hydrogen-bonded interactions with the residues in the active pocket. It is concluded that the configuration of E2-17-OH determines the inhibitory potentials towards UGTs. The results are useful in better understanding ligand selectivity of UGTs, as well as in further development of α-E2 in health protection.


Subject(s)
Estradiol , Glucuronosyltransferase , Humans , Glucuronosyltransferase/chemistry , Glucuronosyltransferase/metabolism , Estradiol/metabolism , UDP-Glucuronosyltransferase 1A9 , Kinetics , Estrogens , Uridine Diphosphate
3.
Anal Chem ; 94(43): 15057-15066, 2022 11 01.
Article in English | MEDLINE | ID: mdl-36262049

ABSTRACT

Autophagy is a core recycling process for homeostasis, with its dysfunction associated with tumorigenesis and various diseases. Yet, its subtle intracellular details are covered due to the limited resolution of conventional microscopies. The major challenge for modern super-resolution microscopy deployment is the lack of a practical labeling system, which could provide robust fluorescence with fidelity in the context of the dynamic autophagy microenvironment. Herein, a representative autophagy marker LC3 protein is selected to develop two hybrid self-labeling systems with tetramethylrhodamine (TMR) fluorophores through SNAP/Halo-tag technologies. A systematic investigation indicated that the match of the LC3-Halo and TMR ligand remarkably outperforms that of LC3-SNAP, as the former Halo system exhibited more robust single-molecule brightness (440 vs 247), total photon numbers (45600 vs 13500), and dwell time of the initial bright state (0.82 vs 0.40 s) than the latter. With the aid of this desirable Halo system, for the first time, live-cell ferritinophagy is monitored with a spatial resolution of ∼50 nm, which disclosed reduced sizes of autophagosomes (∼650 nm, ferritinophagy) than those in nonselective (∼840 nm, mammalian target of rapamycin (mTOR)) and selective autophagy (∼900 nm, mitophagy).


Subject(s)
Autophagy , Fluorescent Dyes , Ligands , Mitophagy , Proteins
4.
Comput Biol Med ; 149: 105959, 2022 10.
Article in English | MEDLINE | ID: mdl-36063691

ABSTRACT

UDP-glucuronosyltransferase (UGT) 1A1, one of the most important isoforms in UGTs superfamily, has attracted increasing concerns for its special role in the clearance and detoxification of endogenous and exogenous substances. To avoid the clinical drug-drug interactions, it is of great importance to have the knowledge of the metabolic profile of UGT1A1 substrates early. Herein, we purposed to establish machine learning models to predict the metabolic propeties of UGT1A1 substrates. On the basis of the literature-derived substrates database of UGT1A1, automatic metabolism prediction models for the aromatic hydroxyl (ArOH) and carboxyl (COOH) groups were developed with eight machine learning methods, among which, three methods, i.e. Random Forest, Random Subspace and J48, illustrated the best performance either for the aromatic hydroxyl and the carboxyl model. The models illustrated good robustness when they were evaluated with functions like "Precision", "Recall", "F-Measure", "AUC", "MCC", etc. Nice accuracy was observed for the aromatic hydroxyl and carboxyl model of these methods, whose AUCs ranged from 0.901 to 0.997. Additionally, the ArOH model was applied to predict the UGT1A1-mediated metabolism of an external set. Two new unknown substrates, cytochrome P450 (CYPs)-mediated metabolites of gefitinib, were predicted and identified, which were validated by in vitro assays. In summary, this study provides a reliable and robust strategy to predict UGT1A1 metabolites, which will be helpful either in rational-optimization of drug metabolism or in avoiding drug-drug interactions in clinic.


Subject(s)
Cytochrome P-450 Enzyme System , Glucuronosyltransferase , Cytochrome P-450 Enzyme System/metabolism , Gefitinib , Glucuronosyltransferase/metabolism , Humans , Protein Isoforms , Uridine Diphosphate
5.
Front Pharmacol ; 13: 815235, 2022.
Article in English | MEDLINE | ID: mdl-35264954

ABSTRACT

Human UDP-glucuronosyltransferase 1A1 (hUGT1A1) is one of the most essential phase II enzymes in humans. Dysfunction or strong inhibition of hUGT1A1 may result in hyperbilirubinaemia and clinically relevant drug/herb-drug interactions (DDIs/HDIs). Recently, a high-throughput fluorescence-based assay was constructed by us to find the compounds/herbal extracts with strong inhibition against intracellular hUGT1A1. Following screening of over one hundred of herbal products, the extract of Ginkgo biloba leaves (GBL) displayed the most potent hUGT1A1 inhibition in HeLa-UGT1A1 cells (Hela cells overexpressed hUGT1A1). Further investigations demonstrated that four biflavones including bilobetin, isoginkgetin, sciadopitysin and ginkgetin, are key constituents responsible for hUGT1A1 inhibition in living cells. These biflavones potently inhibit hUGT1A1 in both human liver microsomes (HLM) and living cells, with the IC50 values ranging from 0.075 to 0.41 µM in living cells. Inhibition kinetic analyses and docking simulations suggested that four tested biflavones potently inhibit hUGT1A1-catalyzed NHPN-O-glucuronidation in HLM via a mixed inhibition manner, showing the K i values ranging from 0.07 to 0.74 µM. Collectively, our findings uncover the key constituents in GBL responsible for hUGT1A1 inhibition and decipher their inhibitory mechanisms against hUGT1A1, which will be very helpful for guiding the rational use of GBL-related herbal products in clinical settings.

6.
Int J Clin Pharmacol Ther ; 60(3): 146-158, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34846299

ABSTRACT

OBJECTIVE: Previous clinical studies and meta-analyses have shown controversial results on the association between C3435T polymorphism of the ABCB1 gene and anti-epileptic drug (AED) resistance. Based on the fact that sample size and confounding factors could contribute to the inconsistency, we performed an updated meta-analysis by including the most recent studies, and subgroup analysis was conducted to evaluate the effect of confounding factors on the association. MATERIALS AND METHODS: We searched articles in 6 electronic databases including PubMed, Medline, Embase, Web of science, Cochrane Library, CNKI (China National Knowledge Infrastructure) for relevant articles up to June 2020. RESULTS: The current analysis showed that the C allele of C3435T variant was a risk factor for drug resistance in the overall populations (C allele vs. T allele, OR: 1.13; 95% CI: 1.02 - 1.25; p = 0.02) and in the Caucasians (C allele vs. T allele, OR: 1.09; 95% CI: 1.09 - 1.43; p = 0.002), while no association was observed in Asians and Indians. Particularly, our study reported for the first time that the 3435T allele was more common in epilepsy patients with drug resistance in the Tunisian population (C allele vs. T allele, OR: 0.31; 95% CI: 0.15 - 0.65; p = 0.002). In addition, our present analysis suggested an association between C3435T and AED resistance in cryptogenic, symptomatic, but not in idiopathic patients. Subgroup studies based on age and gender showed no association. CONCLUSION: AED resistance in Caucasian and Tunisian populations may benefit from ABCB1 C3435T genotyping. We recommend that more details, such as gender and etiology of epilepsy, should be taken into account to draw a reliable conclusion in future studies.


Subject(s)
Anticonvulsants , Epilepsy , ATP Binding Cassette Transporter, Subfamily B/genetics , Anticonvulsants/adverse effects , Asian People/genetics , Drug Resistance/genetics , Epilepsy/drug therapy , Epilepsy/genetics , Genetic Predisposition to Disease , Humans , Polymorphism, Single Nucleotide
7.
Basic Clin Pharmacol Toxicol ; 129(6): 437-449, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34478607

ABSTRACT

Methylophiopogonanone A (MOA) is an abundant homoisoflavonoid in the Chinese herb Ophiopogonis Radix. Recent investigations revealed that MOA inhibited several human cytochrome P450 enzymes (CYPs) and stimulated OATP1B1. However, the inhibitory effects of MOA on phase II drug-metabolizing enzymes, such as human UDP-glucuronosyltransferases (hUGTs), have not been well investigated. Herein, the inhibition potentials of MOA on hUGTs were assessed. The results clearly demonstrated that MOA dose-dependently inhibited all tested hUGTs including UGT1A1 (IC50 = 1.23 µM), one of the most important detoxification enzymes in humans. Further investigations showed that MOA strongly inhibited UGT1A1-catalysed NHPH-O-glucuronidation in a range of biological settings including hUGT1A1, human liver microsomes (HLM) and HeLa cells overexpressing UGT1A1. Inhibition kinetic analyses demonstrated that MOA competitively inhibited UGT1A1-catalysed NHPH-O-glucuronidation in both hUGT1A1 and HLM, with Ki values of 0.52 and 1.22 µM, respectively. Collectively, our findings expanded knowledge of the interactions between MOA and human drug-metabolizing enzymes, which would be very helpful for guiding the use of MOA-related herbal products in clinical settings.


Subject(s)
Benzodioxoles/pharmacology , Enzyme Inhibitors/pharmacology , Glucuronosyltransferase/antagonists & inhibitors , Herb-Drug Interactions , Isoflavones/pharmacology , Benzodioxoles/administration & dosage , Dose-Response Relationship, Drug , Enzyme Inhibitors/administration & dosage , HeLa Cells , Humans , Inhibitory Concentration 50 , Isoflavones/administration & dosage , Microsomes, Liver/enzymology
8.
Seizure ; 91: 339-345, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34284302

ABSTRACT

OBJECTIVES: To assess the efficiency and safety profiles of levetiracetam and (fos)phenytoin (phenytoin or fosphenytoin) for second-line treatment of seizures by performing a meta-analysis of RCTs. METHODS: We systematically searched PubMed, Embase, Cochrane, FDA.gov, and ClinicalTrials.gov for RCTs (published before July 31, 2020; no language restrictions). Two independent reviewers screened abstracts and titles against inclusion and exclusion criteria published previously in the PROSPERO: CRD42020202736. Eleven studies fulfilled the established criteria. We assessed pooled data by using a random-effects model. Quality analysis was performed by using version 2 of the Cochrane risk-of-bias tool (RoB 2). RevMan v.5.3 was used to perform statistical analyses, and publication bias (egger's test) was assessed with Stata MP v.14.0. RESULTS: Levetiracetam was similar to (fos)phenytoin in seizure termination rate (risk ratio [RR] 0.94; 95% CI 0.87 to 1.01), time of seizure termination (mean difference [MD] 0.44; -0.60 to 1.49), and drug resistance ([RR] 1.12, 0.86 to 1.45). The safety outcome showed a significant statistical difference between fosphenytoin group and levetiracetam group ([RR] 1.44, 1.14 to 1.81), while there was no significant difference observed between phenytoin treatment and levetiracetam treatment ([RR] 1.26, 0.99 to 1.60). CONCLUSION: Levetiracetam was similar to (fos)phenytoin in cessation rate convulsive status epilepticus, and drug resistance, while it was superior (fos)phenytoin in pooled safety outcome. Further exploration is still needed as to whether it is the first choice for second-line drugs.


Subject(s)
Phenytoin , Status Epilepticus , Anticonvulsants/therapeutic use , Humans , Levetiracetam/therapeutic use , Phenytoin/therapeutic use , Randomized Controlled Trials as Topic , Status Epilepticus/drug therapy
9.
J Biol Inorg Chem ; 24(7): 1023-1033, 2019 10.
Article in English | MEDLINE | ID: mdl-31506822

ABSTRACT

In the present work, we performed Density Functional Theory calculations to explore the bioactivation mechanism of thiophene-containing molecules mediated by P450s. For this purpose, relatively large size compounds, 2,5-diaminothiophene derivatives were selected particularly for this investigation. Here we found the successive regio-selectivity triggered by conformational turn played a significant role in the occurrence of bioactivation. 2,5-Diaminothiophene was oxidized to a 2,5-diimine thiophene-reactive intermediate by Compound I (Cpd I) through successive activations of two N-H bonds (H3-N11 and H1-N6). This reaction exhibited three special characteristics: (1) self-controlled regio-selectivity during the oxidation process. There was a large scale of conformational turn in the abstraction of the first H atom which triggers the selection of the second H for abstraction. (2) Proton-shuttle mechanism. In high spin (HS) state, proton-shuttle mechanism was observed for the abstraction of the second H atom. (3) Spin-selective manner. In protein environment, the energy barrier in HS state was much lower than that in low spin state. The novel proposed bioactivation mechanism of 2,5-diaminothiophene compounds can help us in rational design of thiophene-contained drugs avoiding the occurrence of bioactivation.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Density Functional Theory , Thiophenes/chemistry , Thiophenes/metabolism , Biocatalysis , Models, Molecular , Molecular Conformation , Stereoisomerism , Substrate Specificity
10.
J Nat Prod ; 82(6): 1503-1509, 2019 06 28.
Article in English | MEDLINE | ID: mdl-31117520

ABSTRACT

Six new macrolides named myrothecines D-G (1-4), 16-hydroxymytoxin B (5), and 14'-dehydrovertisporin (6), including four 10,13-cyclotrichothecane derivatives, in addition to 12 known compounds (7-18), were isolated from three endophytic Myrothecium roridum, IFB-E008, IFB-E009, and IFB-E012. The isolated compounds were characterized by MS, NMR, CD, and single-crystal X-ray crystallography. The isolated macrolides exhibited an antiproliferation effect against chronic myeloid leukemia K562 and colorectal carcinoma SW1116 cell lines. Compounds 1-6 were cytotoxic, with IC50 values ranging between 56 nM and 16 µM. Since slight structural changes led to obvious activity differences, the CoMFA (comparative molecular field analysis) and CoMSIA (comparative molecular similarity indices analysis) methods were then used to explore the 3D QSAR (three-dimensional quantitative structure-activity relationship) of these macrolides. The result showed that the steric, electrostatic, hydrophobic, and H-bond acceptor factors were involved in their cytotoxicity and provided an in-depth understanding of the structure-activity relationships of these metabolites.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antineoplastic Agents/pharmacology , Hypocreales/chemistry , Macrolides/pharmacology , Mitosporic Fungi/chemistry , Protein Synthesis Inhibitors/pharmacology , Trichothecenes/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/isolation & purification , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , Crystallography, X-Ray , Macrolides/chemistry , Macrolides/isolation & purification , Magnetic Resonance Spectroscopy , Molecular Structure , Protein Synthesis Inhibitors/chemistry , Protein Synthesis Inhibitors/isolation & purification , Quantitative Structure-Activity Relationship , Trichothecenes/chemistry , Trichothecenes/isolation & purification
11.
Int J Biol Macromol ; 126: 653-661, 2019 Apr 01.
Article in English | MEDLINE | ID: mdl-30594625

ABSTRACT

Flavonoids are widely distributed phytochemicals in vegetables, fruits and medicinal plants. Recent studies demonstrate that some natural flavonoids are potent inhibitors of the human UDP-glucuronosyltransferase 1A1 (UGT1A1), a key enzyme in detoxification of endogenous harmful compounds such as bilirubin. In this study, the inhibitory effects of 56 natural and synthetic flavonoids on UGT1A1 were assayed, while the structure-inhibition relationships of flavonoids as UGT1A1 inhibitors were investigated. The results demonstrated that the C-3 and C-7 hydroxyl groups on the flavone skeleton would enhance UGT1A1 inhibition, while flavonoid glycosides displayed weaker inhibitory effects than their corresponding aglycones. Further investigation on inhibition kinetics of two strong flavonoid-type UGT1A1 inhibitors, acacetin and kaempferol, yielded interesting results. Both flavonoids were competitive inhibitors against UGT1A1-mediated NHPN-O-glucuronidation, but were mixed and competitive inhibitors toward UGT1A1-mediated NCHN-O-glucuronidation, respectively. Furthermore, docking simulations showed that the binding areas of NHPN, kaempferol and acacetin on UGT1A1 were highly overlapping, and convergence with the binding area of bilirubin within UGT1A1. In summary, detailed structure-inhibition relationships of flavonoids as UGT1A1 inhibitors were investigated carefully and the findings shed new light on the interactions between flavonoids and UGT1A1, and will contribute considerably to the development of flavonoid-type drugs without strong UGT1A1 inhibition.


Subject(s)
Flavonoids/pharmacology , Glucuronosyltransferase/antagonists & inhibitors , Catalytic Domain , Flavones/chemistry , Flavones/pharmacology , Flavonoids/chemistry , Fluorescent Dyes/metabolism , Glucuronosyltransferase/chemistry , Glucuronosyltransferase/metabolism , Humans , Inhibitory Concentration 50 , Kaempferols/chemistry , Kaempferols/pharmacology , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Molecular Docking Simulation , Substrate Specificity/drug effects
14.
Int J Clin Pharmacol Ther ; 56(7): 337-346, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29628024

ABSTRACT

OBJECTIVE: Therapeutic response to phenytoin (PHT), a first-line antiepileptic drug (AED), is highly variable, in part likely due to genetic factors. Genetic polymorphisms in cytochrome P450 (CYP) 2C9 and CYP2C19 are expected to affect the metabolism of PHT and consequently affect its maintenance doses. We aimed to clarify the effects of genetic polymorphisms in both enzymes on daily PHT maintenance dosage in Asian epileptic patients by meta-analysis. MATERIALS AND METHODS: A systematic literature search was conducted in PubMed and EMBASE for relevant studies published prior to April 14, 2017. RevMan 5.2.3 software was used to analyze the relationship between CYP2C9/2C19 polymorphisms and PHT maintenance doses. RESULTS: A total of 6 studies with 993 patients fulfilling the inclusion criteria were included in our meta-analysis. The homozygous and heterozygous CYP2C19 mutation group (i.e., CYP2C19*2/*2, CYP2C19*3/*3, or CYP2C19*2/*3 group) required significant decrease of PHT maintenance dose. The starting maintenance dose suggested in this group is 4.38 mg/kg/day. Patients with heterozygous CYP2C9 or both heterozygous CYP2C9 and CYP2C19 showed a trend but not a statistically-significant decrease of PHT dose, but dosage adjustment was recommended. CONCLUSION: The meta-analysis indicates that CYP2C9 and CYP2C19 polymorphisms are associated with lower PHT maintenance dosage in Asian epileptic patients. Ethnic differences can influence PHT maintenance dose.
.


Subject(s)
Anticonvulsants/administration & dosage , Cytochrome P-450 CYP2C19/genetics , Cytochrome P-450 CYP2C9/genetics , Epilepsy/drug therapy , Pharmacogenomic Variants , Phenytoin/administration & dosage , Polymorphism, Single Nucleotide , Anticonvulsants/adverse effects , Anticonvulsants/pharmacokinetics , Asian People/genetics , Chi-Square Distribution , Cytochrome P-450 CYP2C19/metabolism , Cytochrome P-450 CYP2C9/metabolism , Drug Monitoring , Epilepsy/enzymology , Epilepsy/ethnology , Epilepsy/genetics , Heterozygote , Homozygote , Humans , Pharmacogenetics , Phenytoin/adverse effects , Phenytoin/pharmacokinetics , Risk Factors , Treatment Outcome
15.
Cancer Cell Int ; 17: 103, 2017.
Article in English | MEDLINE | ID: mdl-29162985

ABSTRACT

BACKGROUND: Renal cell carcinoma (RCC) is the most common kidney cancer, accounting for approximately 80-90% of all primary kidney cancer. Treatment for patients with advanced RCC remains unsatisfactory. Rare cancer stem cells (CSCs) are proposed to be responsible for failure of current treatment. METHODS: OncoLnc was used as a tool for interactively exploring survival correlations. Gene manipulation and expression analysis were carried out using siRNA, RT-PCR and Western blotting. Wound healing and invasion assays were used for phenotypical characterization. Aldefluor assay and FACS sorting Sphere culture were used to determine the "stemness" of CSCs. Co-Immunoprecipitation (Co-IP) was used to examine the interaction between OCT4 and CBFA2T2. Student's t-test and Chi square test was used to analyze statistical significance. RESULTS: CBFA2T2 expression can significantly predict the survival of RCC patients. Knocking-down of CBFA2T2 can inhibit cell migration and invasion in RCC cells in vitro, and reduce ALDHhigh CSCs populations. CBFA2T2 expression is necessary for sphere-forming ability and cancer stem cells marker expression in RCC cell lines. CONCLUSIONS: Our data suggest that CBFA2T2 expression correlates with aggressive characteristics of RCC and CBFA2T2 is required for maintenance of "stemness" through regulation of stem cells factors, thereby highlighting CBFA2T2 as a potential therapeutic target for RCC treatment.

16.
J Med Chem ; 60(23): 9664-9675, 2017 12 14.
Article in English | MEDLINE | ID: mdl-29125289

ABSTRACT

This study aimed to develop a practical and high-affinity fluorescent probe for uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1), a key conjugative enzyme responsible for the elimination and detoxification of many potentially harmful compounds. Several substrates derived from N-butyl-4-phenyl-1,8-naphthalimide were designed and synthesized on the basis of the substrate preference of UGT1A1 and the principle of photoinduced electron transfer (PET). Following the preliminary screening, substrate 2 was found with a high specificity and high affinity toward UGT1A1, while such biotransformation brought remarkable changes in fluorescence emission. Both inhibition kinetic analyses and molecular docking simulations demonstrated that 2 could bind on UGT1A1 at the same ligand-binding site as bilirubin. Furthermore, this newly developed probe was successfully used for sensing UGT1A1 activities and the high-throughput screening of UGT1A1 modulators in complex biological samples. In conclusion, a practical and high-affinity fluorescent probe for UGT1A1 was designed and well-characterized, which could serve as a good surrogate for bilirubin to investigate UGT1A1-ligand interactions.


Subject(s)
Bilirubin/metabolism , Enzyme Assays/methods , Fluorescent Dyes/metabolism , Glucuronosyltransferase/metabolism , Bilirubin/analysis , Binding Sites , Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/pharmacology , Fluorescent Dyes/analysis , Glucuronosyltransferase/analysis , Glucuronosyltransferase/antagonists & inhibitors , Hep G2 Cells , High-Throughput Screening Assays/methods , Humans , Kinetics , Molecular Docking Simulation , Spectrometry, Fluorescence/methods
17.
PLoS One ; 12(6): e0179333, 2017.
Article in English | MEDLINE | ID: mdl-28628631

ABSTRACT

EGFR inhibitors, even with therapeutics superiorities in anticancer, can cause idiosyncratic pulmonary and hepatic toxicities that are associated with the reactive electrophile bioactivated by Cytochrome P450s (P450s). Until now, neither has the electrophilic intermediate been caught experimentally, nor has the subtle mechanism been declared. Herein, the underlying mechanism of bioactivation mediated by P450s was explored by DFT calculations for a case of EGFR inhibitor, Erlotinib. Based on the calculation and analysis, we suggest that with other metabolites, reactive electrophiles of Erlotinib: epoxide and quinine-imine, can be generated by several steps along the oxidative reaction pathway. The generation of epoxide needs two steps: (1) the addition of Erlotinib to Compound I (Cpd I) and (2) the rearrangement of protons. Whereas, quinine-imine needs a further oxidation step (3) via which quinone is generated and ultimately turns into quinine-imine. Although both reactive electrophiles can be produced for either face-on or side-on pose of Erlotinib, the analysis of energy barriers indicates that the side-on path is preferred in solvent environment. In the rate-determining step, e.g. the addition of Erlotinib to the porphyrin, the reaction barrier for side-on conformation is decreased in aqueous and protein environment compared with gas phase, whereas, the barrier for face-on pose is increased in solvent environment. The simulated mechanism is in good agreement with the speculation in previous experiment. The understanding of the subtle mechanism of bioactivation of Erlotinib will provide theoretical support for toxicological mechanism of EGFR inhibitors.


Subject(s)
Antineoplastic Agents/metabolism , Cytochrome P-450 Enzyme System/metabolism , Erlotinib Hydrochloride/metabolism , Antineoplastic Agents/chemistry , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP3A/metabolism , Epoxy Compounds/chemistry , Epoxy Compounds/metabolism , Erlotinib Hydrochloride/chemistry , Models, Molecular , Oxidation-Reduction , Quantum Theory , Quinones/chemistry , Quinones/metabolism , Thermodynamics
18.
Oncotarget ; 8(15): 24483-24490, 2017 Apr 11.
Article in English | MEDLINE | ID: mdl-28445934

ABSTRACT

Bladder cancer is the most common urologic malignancy in China, with an increase of the incidence and mortality rates over past decades. Recent studies suggest that bladder tumors are maintained by a rare fraction of cells with stem cell proprieties. Targeting these bladder tumor initiating cell (TICs) population can overcome the drug-resistance of bladder cancer. However, the molecular and genetic mechanisms regulating TICs in bladder cancer remain poorly defined. Jarid2 is implicated in signaling pathways regulating cancer cell epithelial-mesenchymal transition, and stem cell maintenance. The goal of our study was to examine whether Jarid2 plays a role in the regulation of TICs in bladder cancer. We found that knockdown of Jarid2 was able to inhibit the invasive ability and sphere-forming capacity in bladder cancer cells. Moreover, knockdown of Jarid2 reduced the proportion of TICs and impaired the tumorigenicity of bladder cancer TICs in vivo. Conversely, ectopic overexpression of Jarid2 promoted the invasive ability and sphere-forming capacity in bladder cancer cells. Mechanistically, reduced Jarid2 expression led to the upregulation of p16 and H3K27me3 level at p16 promoter region. Collectively, we provided evidence that Jarid2 via modulation of p16 is a putative novel therapeutic target for treating malignant bladder cancer.


Subject(s)
Neoplastic Stem Cells/metabolism , Polycomb Repressive Complex 2/genetics , Urinary Bladder Neoplasms/genetics , Cell Line, Tumor , Humans , Neoplastic Stem Cells/pathology , Polycomb Repressive Complex 2/metabolism , Signal Transduction , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology
19.
Oncotarget ; 7(39): 63561-63570, 2016 Sep 27.
Article in English | MEDLINE | ID: mdl-27566578

ABSTRACT

Long non-coding RNAs (lncRNAs) play an important role in gene regulation and are involving in diverse cellular processes. However, their roles in reprogramming of gene expression profiles during lineage commitment and maturation of mesenchymal stem cells (MSCs) remain poorly understood. In the current study, we characterize the expression of a lncRNA, HoxA-AS3, during the differentiation of MSCs. We showed that HoxA-AS3 is increased upon adipogenic induction of MSCs, while HoxA-AS3 remains unaltered during osteogenic induction. Silencing of HoxA-AS3 in MSCs resulted in decreased adipogenesis and expression of adipogenic markers, PPARG, CEBPA, FABP4 and ADIPOQ. Conversely, knockdown of HoxA-AS3 expression in MSCs exhibited an enhanced osteogenesis and osteogenic markers expression, including RUNX2, SP7, COL1A1, IBSP, BGLAP and SPP1. Mechanistically, HoxA-AS3 interacts with Enhancer Of Zeste 2 (EZH2) and is required for H3 lysine-27 trimethylation (H3K27me3) of key osteogenic transcription factor Runx2. Our data reveal that HoxA-AS3 acts as an epigenetic switch that determines the lineage specification of MSC.


Subject(s)
Adipogenesis/genetics , Cell Lineage/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Mesenchymal Stem Cells/cytology , Osteogenesis/genetics , RNA, Long Noncoding/metabolism , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Enhancer of Zeste Homolog 2 Protein/genetics , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Osteoblasts/cytology , Osteoblasts/metabolism , RNA, Long Noncoding/genetics
20.
Int J Mol Sci ; 16(9): 20118-38, 2015 Aug 25.
Article in English | MEDLINE | ID: mdl-26307982

ABSTRACT

Presently, 151 widely-diverse pyridinylimidazole-based compounds that show inhibitory activities at the TNF-α release were investigated. By using the distance comparison technique (DISCOtech), comparative molecular field analysis (CoMFA), and comparative molecular similarity index analysis (CoMSIA) methods, the pharmacophore models and the three-dimensional quantitative structure-activity relationships (3D-QSAR) of the compounds were explored. The proposed pharmacophore model, including two hydrophobic sites, two aromatic centers, two H-bond donor atoms, two H-bond acceptor atoms, and two H-bond donor sites characterizes the necessary structural features of TNF-α release inhibitors. Both the resultant CoMFA and CoMSIA models exhibited satisfactory predictability (with Q(2) (cross-validated correlation coefficient) = 0.557, R(2)ncv (non-cross-validated correlation coefficient) = 0.740, R(2)pre (predicted correlation coefficient) = 0.749 and Q(2) = 0.598, R(2)ncv = 0.767, R(2)pre = 0.860, respectively). Good consistency was observed between the 3D-QSAR models and the pharmacophore model that the hydrophobic interaction and hydrogen bonds play crucial roles in the mechanism of actions. The corresponding contour maps generated by these models provide more diverse information about the key intermolecular interactions of inhibitors with the surrounding environment. All these models have extended the understanding of imidazole-based compounds in the structure-activity relationship, and are useful for rational design and screening of novel 2-thioimidazole-based TNF-α release inhibitors.


Subject(s)
Imidazoles/chemistry , Models, Molecular , Quantitative Structure-Activity Relationship , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Drug Design , Enzyme Inhibitors/chemistry , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Molecular Conformation , Reproducibility of Results , Static Electricity
SELECTION OF CITATIONS
SEARCH DETAIL
...