Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 890
Filter
Add more filters

Publication year range
1.
Bioorg Chem ; 150: 107536, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38878751

ABSTRACT

Carboxylesterase 1 (CES1), a member of the serine hydrolase superfamily, is involved in a wide range of xenobiotic and endogenous substances metabolic reactions in mammals. The inhibition of CES1 could not only alter the metabolism and disposition of related drugs, but also be benefit for treatment of metabolic disorders, such as obesity and fatty liver disease. In the present study, we aim to develop potential inhibitors of CES1 and reveal the preferred inhibitor structure from a series of synthetic pyrazolones (compounds 1-27). By in vitro high-throughput screening method, we found compounds 25 and 27 had non-competitive inhibition on CES1-mediated N-alkylated d-luciferin methyl ester (NLMe) hydrolysis, while compound 26 competitively inhibited CES1-mediated NLMe hydrolysis. Additionally, Compounds 25, 26 and 27 can inhibit CES1-mediated fluorescent probe hydrolysis in live HepG2 cells with effect. Besides, compounds 25, 26 and 27 could effectively inhibit the accumulation of lipid droplets in mouse adipocytes cells. These data not only provided study basis for the design of newly CES1 inhibitors. The present study not only provided the basis for the development of lead compounds for novel CES1 inhibitors with better performance, but also offered a new direction for the explore of candidate compounds for the treatment of hyperlipidemia and related diseases.


Subject(s)
Adipocytes , Carboxylic Ester Hydrolases , Enzyme Inhibitors , Pyrazolones , Humans , Carboxylic Ester Hydrolases/metabolism , Carboxylic Ester Hydrolases/antagonists & inhibitors , Adipocytes/drug effects , Adipocytes/metabolism , Adipocytes/cytology , Animals , Mice , Pyrazolones/pharmacology , Pyrazolones/chemistry , Pyrazolones/chemical synthesis , Structure-Activity Relationship , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/chemical synthesis , Molecular Structure , Hep G2 Cells , Cell Differentiation/drug effects , Dose-Response Relationship, Drug , 3T3-L1 Cells
2.
Proc Natl Acad Sci U S A ; 118(41)2021 10 12.
Article in English | MEDLINE | ID: mdl-34607947

ABSTRACT

Plasticity in multicellular organisms involves signaling pathways converting contexts-either natural environmental challenges or laboratory perturbations-into context-specific changes in gene expression. Congruently, the interactions between the signaling molecules and transcription factors (TF) regulating these responses are also context specific. However, when a target gene responds across contexts, the upstream TF identified in one context is often inferred to regulate it across contexts. Reconciling these stable TF-target gene pair inferences with the context-specific nature of homeostatic responses is therefore needed. The induction of the Caenorhabditis elegans genes lipl-3 and lipl-4 is observed in many genetic contexts and is essential to survival during fasting. We find DAF-16/FOXO mediating lipl-4 induction in all contexts tested; hence, lipl-4 regulation seems context independent and compatible with across-context inferences. In contrast, DAF-16-mediated regulation of lipl-3 is context specific. DAF-16 reduces the induction of lipl-3 during fasting, yet it promotes it during oxidative stress. Through discrete dynamic modeling and genetic epistasis, we define that DAF-16 represses HLH-30/TFEB-the main TF activating lipl-3 during fasting. Contrastingly, DAF-16 activates the stress-responsive TF HSF-1 during oxidative stress, which promotes C. elegans survival through induction of lipl-3 Furthermore, the TF MXL-3 contributes to the dominance of HSF-1 at the expense of HLH-30 during oxidative stress but not during fasting. This study shows how context-specific diverting of functional interactions within a molecular network allows cells to specifically respond to a large number of contexts with a limited number of molecular players, a mode of transcriptional regulation we name "contextualized transcription."


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Fasting/physiology , Forkhead Transcription Factors/metabolism , Gene Expression Regulation/genetics , Lipase/metabolism , Oxidative Stress/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/antagonists & inhibitors , Caenorhabditis elegans Proteins/genetics , Carboxylic Ester Hydrolases/antagonists & inhibitors , Carboxylic Ester Hydrolases/genetics , Carboxylic Ester Hydrolases/metabolism , Lipase/genetics , Lipolysis/physiology , Signal Transduction/physiology , Transcription Factors/metabolism , Transcription, Genetic/genetics , Transcriptional Activation/physiology
3.
Environ Toxicol ; 39(7): 3856-3871, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38558378

ABSTRACT

Discharges to the aquatic environment of pharmaceuticals represent a hazard to the aquatic organisms. Subchronic assay with 17-alpha-ethinylestradiol (EE2) and in vitro essays with pharmaceuticals of environmental concern were conducted to examine the sensitivity of tissue acetylcholinesterase (AChE) and carboxylesterase (CbE) activities of Tinca tinca to them. Subchronic exposure to 17-alpha-EE2 caused significant effects on brain, liver, and muscle CbE, but no on AChE activities. Most of the pharmaceuticals tested in vitro were considered as weak inhibitors of tissular AChE activity. Depending on the tissues, some compounds were classified as moderate inhibitors of CbE activity while other were categorized as weak enzymatic inhibitors. An opposite trend was observed depending on the tissue, while brain and liver CbE activities were inhibited, the muscle CbE activity was induced. Changes experienced on enzymatic activities after exposure to pharmaceuticals might affect the physiological functions in which these enzymes are involved. In vitro exposure to 17-alpha-EE2 in tench could be an informative, but not a surrogate model to know the effect of this synthetic estrogen on AChE and CbE activities.


Subject(s)
Water Pollutants, Chemical , Animals , Water Pollutants, Chemical/toxicity , Liver/drug effects , Liver/enzymology , Cyprinidae , Acetylcholinesterase/metabolism , Brain/drug effects , Brain/enzymology , Cholinesterase Inhibitors/toxicity , Muscles/drug effects , Muscles/enzymology , Carboxylesterase/metabolism , Carboxylic Ester Hydrolases/metabolism , Carboxylic Ester Hydrolases/antagonists & inhibitors , Cholinesterases/metabolism
4.
J Enzyme Inhib Med Chem ; 37(1): 629-640, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35100926

ABSTRACT

Pancreatic lipase (PL) is a well-known key target for the prevention and treatment of obesity. Human carboxylesterase 1A (hCES1A) has become an important target for the treatment of hyperlipidaemia. Thus, the discovery of potent dual-target inhibitors based on PL and hCES1A hold great potential for the development of remedies for treating related metabolic diseases. In this study, a series of natural triterpenoids were collected and the inhibitory effects of these triterpenoids on PL and hCES1A were determined using fluorescence-based biochemical assays. It was found that oleanolic acid (OA) and ursolic acid (UA) have the excellent inhibitory effects against PL and hCES1A, and highly selectivity over hCES2A. Subsequently, a number of compounds based on the OA and UA skeletons were synthesised and evaluated. Structure-activity relationship (SAR) analysis of these compounds revealed that the acetyl group at the C-3 site of UA (compound 41) was very essential for both PL and hCES1A inhibition, with IC50 of 0.75 µM and 0.014 µM, respectively. In addition, compound 39 with 2-enol and 3-ketal moiety of OA also has strong inhibitory effects against both PL and hCES1A, with IC50 of 2.13 µM and 0.055 µM, respectively. Furthermore, compound 39 and 41 exhibited good selectivity over other human serine hydrolases including hCES2A, butyrylcholinesterase (BChE) and dipeptidyl peptidase IV (DPP-IV). Inhibitory kinetics and molecular docking studies demonstrated that both compounds 39 and 41 were effective mixed inhibitors of PL, while competitive inhibitors of hCES1A. Further investigations demonstrated that both compounds 39 and 41 could inhibit adipocyte adipogenesis induced by mouse preadipocytes. Collectively, we found two triterpenoid derivatives with strong inhibitory ability on both PL and hCES1A, which can be served as promising lead compounds for the development of more potent dual-target inhibitors targeting on PL and hCES1A.


Subject(s)
Carboxylic Ester Hydrolases/antagonists & inhibitors , Drug Discovery , Enzyme Inhibitors/pharmacology , Lipase/antagonists & inhibitors , Pancreas/enzymology , Triterpenes/pharmacology , Carboxylic Ester Hydrolases/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Lipase/metabolism , Molecular Structure , Structure-Activity Relationship , Triterpenes/chemical synthesis , Triterpenes/chemistry
5.
Angew Chem Int Ed Engl ; 60(6): 3071-3079, 2021 02 08.
Article in English | MEDLINE | ID: mdl-33035395

ABSTRACT

Herein, we report arylazopyrazole ureas and sulfones as a novel class of photoswitchable serine hydrolase inhibitors and present a chemoproteomic platform for rapid discovery of optically controlled serine hydrolase targets in complex proteomes. Specifically, we identify highly potent and selective photoswitchable inhibitors of the drug-metabolizing enzymes carboxylesterases 1 and 2 and demonstrate their pharmacological application by optically controlling the metabolism of the immunosuppressant drug mycophenolate mofetil. Collectively, this proof-of-concept study provides a first example of photopharmacological tools to optically control drug metabolism by modulating the activity of a metabolizing enzyme. Our arylazopyrazole ureas and sulfones offer synthetically accessible scaffolds that can be expanded to identify specific photoswitchable inhibitors for other serine hydrolases, including lipases, peptidases, and proteases. Our chemoproteomic platform can be applied to other photoswitches and scaffolds to achieve optical control over diverse protein classes.


Subject(s)
Carboxylesterase/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Pharmaceutical Preparations/metabolism , Ultraviolet Rays , Caco-2 Cells , Carboxylesterase/metabolism , Carboxylic Ester Hydrolases/antagonists & inhibitors , Carboxylic Ester Hydrolases/genetics , Carboxylic Ester Hydrolases/metabolism , Drug Evaluation, Preclinical , Enzyme Inhibitors/metabolism , Humans , Hydrolysis , Microscopy, Fluorescence , Pharmaceutical Preparations/chemistry , RNA Interference , RNA, Small Interfering/metabolism , Stereoisomerism , Sulfones/chemistry , Sulfones/metabolism , Urease/chemistry , Urease/metabolism
6.
Drug Metab Dispos ; 48(10): 993-1007, 2020 10.
Article in English | MEDLINE | ID: mdl-32591414

ABSTRACT

Carboxylesterase (CES) 1 is the predominant esterase expressed in the human liver and is capable of catalyzing the hydrolysis of a wide range of therapeutic agents, toxins, and endogenous compounds. Accumulating studies have demonstrated associations between the expression and activity of CES1 and the pharmacokinetics and/or pharmacodynamics of CES1 substrate medications (e.g., methylphenidate, clopidogrel, oseltamivir). Therefore, any perturbation of CES1 by coingested xenobiotics could potentially compromise treatment. Natural products are known to alter drug disposition by modulating cytochrome P450 and UDP-glucuronosyltransferase enzymes, but this issue is less thoroughly explored with CES1. We report the results of a systematic literature search and discuss natural products as potential modulators of CES1 activity. The majority of research reports reviewed were in vitro investigations that require further confirmation through clinical study. Cannabis products (Δ 9-tetrahydrocannabinol, cannabidiol, cannabinol); supplements from various plant sources containing naringenin, quercetin, luteolin, oleanolic acid, and asiatic acid; and certain traditional medicines (danshen and zhizhuwan) appear to pose the highest inhibition potential. In addition, ursolic acid, gambogic acid, and glycyrrhetic acid, if delivered intravenously, may attain high enough systemic concentrations to significantly inhibit CES1. The provision of a translational interpretation of in vitro assessments of natural product actions and interactions is limited by the dearth of basic pharmacokinetic data of the natural compounds exhibiting potent in vitro influences on CES1 activity. This is a major impediment to assigning even potential clinical significance. The modulatory effects on CES1 expression after chronic exposure to natural products warrants further investigation. SIGNIFICANCE STATEMENT: Modulation of CES1 activity by natural products may alter the course of treatment and clinical outcome. In this review, we have summarized the natural products that can potentially interact with CES1 substrate medications. We have also noted the limitations of existing reports and outlined challenges and future directions in this field.


Subject(s)
Biological Products/pharmacokinetics , Carboxylic Ester Hydrolases/antagonists & inhibitors , Administration, Intravenous , Administration, Oral , Biological Products/administration & dosage , Carboxylic Ester Hydrolases/metabolism , Clopidogrel/administration & dosage , Clopidogrel/pharmacokinetics , Drug Evaluation, Preclinical , Drug Interactions , Humans , Methylphenidate/administration & dosage , Methylphenidate/pharmacokinetics , Oseltamivir/administration & dosage , Oseltamivir/pharmacokinetics
7.
J Enzyme Inhib Med Chem ; 35(1): 261-264, 2020 Dec.
Article in English | MEDLINE | ID: mdl-31790606

ABSTRACT

Although paraoxonase-1 (PON1) activity has been demonstrated to be a reliable biomarker of various diseases, clinical studies have been based only on relative comparison of specific enzyme activities, which capture differences mainly due to (usually unknown) PON1 concentration. Hence, the aim of this report is to present for the first time the simple evaluation method for determining autonomous kinetic parameter of PON1 that could be also associated with polymorphic forms and diseases; i.e. the Michaelis constant which is enzyme concentration independent quantity. This alternative approach significantly reduces the number of experiments needed, and it yields the results with great accuracy.


Subject(s)
Aryldialkylphosphatase/metabolism , Carboxylic Ester Hydrolases/metabolism , Aryldialkylphosphatase/antagonists & inhibitors , Carboxylic Ester Hydrolases/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Hydroxyquinolines/chemistry , Hydroxyquinolines/pharmacology , Kinetics , Molecular Structure
8.
Biochem Biophys Res Commun ; 508(1): 320-325, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30502087

ABSTRACT

Pectin methylesterases (PMEs) catalyze pectin demethylation and facilitate the determination of the degree of methyl esterification of cell wall in higher plants. The regulation of PME activity through endogenous proteinaceous PME inhibitors (PMEIs) alters the status of pectin methylation and influences plant growth and development. In this study, we performed a PMEI screening assay using a chemical library and identified a strong inhibitor, phenylephrine (PE). PE, a small molecule, competitively inhibited plant PMEs, including orange PME and Arabidopsis PME. Physiologically, cultivation of Brassica campestris seedlings in the presence of PE showed root growth inhibition. Microscopic observation revealed that PE inhibits elongation and development of root hairs. Molecular studies demonstrated that Root Hair Specific 12 (RHS12) encoding a PME, which plays a role in root hair development, was inhibited by PE with a Ki value of 44.1 µM. The biochemical mechanism of PE-mediated PME inhibition as well as a molecular docking model between PE and RHS12 revealed that PE interacts within the catalytic cleft of RHS12 and interferes with PME catalytic activity. Taken together, these findings suggest that PE is a novel and non-proteinaceous PME inhibitor. Furthermore, PE could be a lead compound for developing a potent plant growth regulator in agriculture.


Subject(s)
Carboxylic Ester Hydrolases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Phenylephrine/pharmacology , Small Molecule Libraries/pharmacology , Brassica/drug effects , Brassica/growth & development , Brassica/metabolism , Carboxylic Ester Hydrolases/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Phenylephrine/chemistry , Seedlings/drug effects , Seedlings/metabolism , Small Molecule Libraries/chemistry , Structure-Activity Relationship
9.
Plant Physiol ; 176(3): 2202-2220, 2018 03.
Article in English | MEDLINE | ID: mdl-29363564

ABSTRACT

Pectins are major components of the primary plant cell wall, which functions as the primary barrier against pathogens. Pectin methylesterases (PMEs) catalyze the demethylesterification of the homogalacturonan domains of pectin in the plant cell wall. Their activity is regulated by PME inhibitors (PMEIs). Here, we provide evidence that the pectin methylesterase-inhibiting protein GhPMEI3 from cotton (Gossypium hirsutum) functions in plant responses to infection by the fungus Verticillium dahliae GhPMEI3 interacts with PMEs and regulates the expression of a specific fungal polygalacturonase (VdPG1). Ectopic expression of GhPMEI3 increased pectin methyl esterification and limited fungal disease in cotton, while also modulating root elongation. Enzymatic analyses revealed that GhPMEI3 efficiently inhibited the activity of cotton GhPME2/GhPME31. Experiments using transgenic Arabidopsis (Arabidopsis thaliana) plants expressing the GhPMEI3 gene under the control of the CaMV 35S promoter revealed that GhPMEI3 inhibits the endogenous PME activity in vitro. Moreover, the enhanced resistance to V. dahliae was associated with altered VdPG1 expression. Virus-induced silencing of GhPMEI3 resulted in increased susceptibility to V. dahliae Further, we investigated the interaction between GhPMEI3 and GhPME2/GhPME31 using inhibition assays and molecular docking simulations. The peculiar structural features of GhPMEI3 were responsible for the formation of a 1:1 stoichiometric complex with GhPME2/GhPME31. Together, these results suggest that GhPMEI3 enhances resistance to Verticillium wilt. Moreover, GhPMEI3-GhPMEs interactions would be needed before drawing the correlation between structure-function and are crucial for plant development against the ever-evolving fungal pathogens.


Subject(s)
Carboxylic Ester Hydrolases/antagonists & inhibitors , Carboxylic Ester Hydrolases/chemistry , Gossypium/genetics , Plant Proteins/pharmacology , Verticillium/pathogenicity , Arabidopsis/genetics , Arabidopsis/growth & development , Arabidopsis/metabolism , Arabidopsis/microbiology , Carboxylic Ester Hydrolases/genetics , Carboxylic Ester Hydrolases/metabolism , Gene Expression Regulation, Plant , Gossypium/microbiology , Host-Pathogen Interactions , Molecular Docking Simulation , Pectins/metabolism , Phylogeny , Plant Diseases/genetics , Plant Diseases/microbiology , Plant Proteins/chemistry , Plant Proteins/genetics , Plants, Genetically Modified , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Static Electricity
10.
Xenobiotica ; 49(11): 1260-1268, 2019 Nov.
Article in English | MEDLINE | ID: mdl-30486721

ABSTRACT

1. Schisandra chinensis, also called wuweizi in Chinese, is the fruit of Schisandra chinensis (Turcz.) Baill., and has been officially utilized as an astringent tonic for more than two thousand years in China. This study aims to evaluate the inhibition of carboxylesterases (CESs) by the major ingredients isolated from Schisandra chinensis, including Anwuligan, Schisandrol B, Schisanhenol, deoxyschizandrin, and Schisandrin B. 2. In vitro human liver microsomes (HLMs)-catalyzed hydrolysis of 2-(2-Benzoyl-3-methoxyphenyl) benzothiazole (BMBT) and fluorescein diacetate (FD) was employed as the probe reaction for CES1 and CES2, respectively. Initial screening, inhibition kinetics determination (inhibition type and parameters (Ki)), and in silico docking method were carried out. 3. Schisandrin B showed strong inhibition on the activity of CES1, and the activity of CES2 was strongly inhibited by Anwuligan and Schisandrin B. Schisandrin B exhibited noncompetitive inhibition towards CES1 and CES2. Anwuligan showed competitive inhibition towards CES2. The inhibition kinetic parameters (Ki) were calculated to be 29.8, 0.6, and 8.1 uM for the inhibition of Schisandrin B on CES1, Anwuligan on CES2, and Schisandrin B on CES2. In silico docking showed that hydrogen bonds and hydrophobic interactions contributed to the inhibition of Schisandrin B on CES1, Anwuligan on CES2, and Schisandrin B on CES2. All these information will be helpful for understanding the adverse effects of Schisandra chinensis due to the inhibition of CESs-catalyzed metabolism.


Subject(s)
Carboxylesterase/antagonists & inhibitors , Carboxylic Ester Hydrolases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Schisandra/chemistry , Carboxylesterase/chemistry , Carboxylesterase/metabolism , Carboxylic Ester Hydrolases/chemistry , Carboxylic Ester Hydrolases/metabolism , Cyclooctanes/pharmacology , Dioxoles/pharmacology , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Drug Interactions , Enzyme Inhibitors/chemistry , Humans , Lignans/pharmacology , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Molecular Docking Simulation , Polycyclic Compounds/pharmacology
11.
Biochem J ; 475(3): 621-642, 2018 02 09.
Article in English | MEDLINE | ID: mdl-29321244

ABSTRACT

Macrophage foam cells store excess cholesterol as cholesteryl esters, which need to be hydrolyzed for cholesterol efflux. We recently reported that silencing expression of carboxylesterase 1 (CES1) in human THP-1 macrophages [CES1KD (THP-1 cells with CES1 expression knocked down) macrophages] reduced cholesterol uptake and decreased expression of CD36 and scavenger receptor-A in cells loaded with acetylated low-density lipoprotein (acLDL). Here, we report that CES1KD macrophages exhibit reduced transcription of cytochrome P45027A1 (CYP27A1) in nonloaded and acLDL-loaded cells. Moreover, levels of CYP27A1 protein and its enzymatic product, 27-hydroxycholesterol, were markedly reduced in CES1KD macrophages. Transcription of LXRα (liver X receptor α) and ABCA1 (ATP-binding cassette transporter A1) was also decreased in acLDL-loaded CES1KD macrophages, suggesting reduced signaling through PPARγ-CYP27A1-LXRα. Consistent with this, treatment of CES1KD macrophages with agonists for PPARγ, RAR, and/or RAR/RXR partially restored transcription of CYP27A1 and LXRα, and repaired cholesterol influx. Conversely, treatment of control macrophages with antagonists for PPARγ and/or RXR decreased transcription of CYP27A1 and LXRα Pharmacologic inhibition of CES1 in both wild-type THP-1 cells and primary human macrophages also decreased CYP27A1 transcription. CES1 silencing did not affect transcript levels of PPARγ and RXR in acLDL-loaded macrophages, whereas it did reduce the catabolism of the endocannabinoid 2-arachidonoylglycerol. Finally, the gene expression profile of CES1KD macrophages was similar to that of PPARγ knockdown cells following acLDL exposures, further suggesting a mechanistic link between CES1 and PPARγ. These results are consistent with a model in which abrogation of CES1 function attenuates the CYP27A1-LXRα-ABCA1 signaling axis by depleting endogenous ligands for the nuclear receptors PPARγ, RAR, and/or RXR that regulate cholesterol homeostasis.


Subject(s)
ATP Binding Cassette Transporter 1/genetics , Carboxylic Ester Hydrolases/genetics , Cholestanetriol 26-Monooxygenase/genetics , Cholesterol/metabolism , Liver X Receptors/genetics , CD36 Antigens/genetics , Carboxylic Ester Hydrolases/antagonists & inhibitors , Cell Line , Foam Cells/metabolism , Gene Expression Regulation , Gene Silencing , Humans , Macrophages/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Retinoic Acid Receptor alpha/genetics , Retinoid X Receptor alpha/genetics , Scavenger Receptors, Class A/genetics
12.
Molecules ; 24(15)2019 Jul 29.
Article in English | MEDLINE | ID: mdl-31362390

ABSTRACT

The human carboxylesterase 1 (CES1), responsible for the biotransformation of many diverse therapeutic agents, may contribute to the occurrence of adverse drug reactions and therapeutic failure through drug interactions. The present study is designed to address the issue of potential drug interactions resulting from the inhibition of CES1. Based on an ensemble of 10 crystal structures complexed with different ligands and a set of 294 known CES1 ligands, we used docking (Autodock Vina) and machine learning methodologies (LDA, QDA and multilayer perceptron), considering the different energy terms from the scoring function to assess the best combination to enable the identification of CES1 inhibitors. The protocol was then applied on a library of 1114 FDA-approved drugs and eight drugs were selected for in vitro CES1 inhibition. An inhibition effect was observed for diltiazem (IC50 = 13.9 µM). Three others drugs (benztropine, iloprost and treprostinil), exhibited a weak CES1 inhibitory effects with IC50 values of 298.2 µM, 366.8 µM and 391.6 µM respectively. In conclusion, the binding site of CES1 is relatively flexible and can adapt its conformation to different types of ligands. Combining ensemble docking and machine learning approaches improves the prediction of CES1 inhibitors compared to a docking study using only one crystal structure.


Subject(s)
Carboxylic Ester Hydrolases/chemistry , Machine Learning , Molecular Docking Simulation , Molecular Dynamics Simulation , Protease Inhibitors/chemistry , Carboxylic Ester Hydrolases/antagonists & inhibitors , Drug Discovery , Enzyme Activation/drug effects , Humans , Protease Inhibitors/pharmacology , Quantitative Structure-Activity Relationship , ROC Curve , Reproducibility of Results , Small Molecule Libraries
13.
Molecules ; 24(8)2019 Apr 23.
Article in English | MEDLINE | ID: mdl-31018540

ABSTRACT

Jelly fig (Ficus awkeotsang Makino) is used to prepare drinks and desserts in Asia, owing to the gelling capability of its pectin via endogenous pectin methylesterase (PE) catalyzation. Meanwhile, substances with PE inhibitory activity (SPEI) in jelly fig achenes (JFA) residue were noticed to be able to impede the gelation. In this study, we characterized and isolated SPEI from JFA by a series of PE inhibition-guided isolations. Crude aqueous extract of JFA residue was mixed with acetone, and 90% acetone-soluble matter was further fractionated by Diaion HP-20 chromatography. The retained fraction with dominant PE inhibitory activity was collected from 100% methanol eluate. Results from high-performance liquid chromatography mass spectrometry (HPLC/MS) and hydrolysis-induced chromogenic transition revealed the SPEI as complex tannins. Total tannins content was determined in each isolated fraction, and was closely related to PE inhibitory activity. In addition, SPEI in this study could inhibit activities of digestive enzymes in vitro and may, therefore, be assumed to act as non-specific protein binding agent.


Subject(s)
Carboxylic Ester Hydrolases/antagonists & inhibitors , Enzyme Inhibitors/isolation & purification , Ficus/chemistry , Fruit/chemistry , Plant Proteins/antagonists & inhibitors , Tannins/isolation & purification , Acetone/chemistry , Beverages/analysis , Carboxylic Ester Hydrolases/chemistry , Carboxylic Ester Hydrolases/isolation & purification , Chromatography, Ion Exchange , Enzyme Assays , Enzyme Inhibitors/chemistry , Ficus/enzymology , Fruit/enzymology , Gels , Humans , Methanol/chemistry , Pectins/chemistry , Phase Transition , Plant Extracts/chemistry , Plant Proteins/chemistry , Plant Proteins/isolation & purification , Solvents/chemistry , Taiwan , Tannins/chemistry , Water/chemistry
14.
J Environ Sci Health B ; 54(11): 883-891, 2019.
Article in English | MEDLINE | ID: mdl-31311415

ABSTRACT

The characterization of soluble cholinesterases (ChEs) together with carboxylesterases (CEs) in Ficopomatus enigmaticus as suitable biomarkers of neurotoxicity was the main aim of this study. ChEs of F. enigmaticus were characterized considering enzymatic activity, substrate affinity (acetyl-, butyryl-, propionylthiocholine), kinetic parameters (Km and Vmax) and in vitro response to model inhibitors (eserine hemisulfate, iso-OMPA, BW284C51), and carbamates (carbofuran, methomyl, aldicarb, and carbaryl). CEs were characterized based on enzymatic activity, kinetic parameters and in vitro response to carbamates (carbofuran, methomyl, aldicarb, and carbaryl). Results showed that cholinesterases from F. enigmaticus showed a substrate preference for acetylthiocholine followed by propionylthiocholine; butyrylthioline was not hydrolyzed differently from other Annelida species. CE activity was in the same range of cholinesterase activity with acetylthiocholine as substrate; the enzyme activity showed high affinity for the substrate p-nytrophenyl butyrate. Carbamates inhibited ChE activity with propionylthiocholine as substrate to a higher extent than with acetylthiocoline. Also CE activity was inhibited by all tested carbamates except carbaryl. In vitro data highlighted the presence of active forms of ChEs and CEs in F. enigmaticus that could potentially be inhibited by pesticides at environmentally relevant concentration.


Subject(s)
Annelida/enzymology , Cholinesterase Inhibitors/toxicity , Cholinesterases/chemistry , Neurotoxins/toxicity , Animals , Annelida/drug effects , Biomarkers/chemistry , Carbamates/chemistry , Carbaryl/chemistry , Carbaryl/toxicity , Carbofuran/chemistry , Carbofuran/toxicity , Carboxylic Ester Hydrolases/antagonists & inhibitors , Carboxylic Ester Hydrolases/chemistry , Carboxylic Ester Hydrolases/metabolism , Cholinesterase Inhibitors/chemistry , Cholinesterases/metabolism , Kinetics , Methomyl/chemistry , Methomyl/toxicity , Neurotoxins/chemistry
15.
J Biol Chem ; 292(52): 21538-21547, 2017 12 29.
Article in English | MEDLINE | ID: mdl-29109147

ABSTRACT

Pectin methylesterases (PMEs) catalyze the demethylesterification of pectin, one of the main polysaccharides in the plant cell wall, and are of critical importance in plant development. PME activity generates highly negatively charged pectin and mutates the physiochemical properties of the plant cell wall such that remodeling of the plant cell can occur. PMEs are therefore tightly regulated by proteinaceous inhibitors (PMEIs), some of which become active upon changes in cellular pH. Nevertheless, a detailed picture of how this pH-dependent inhibition of PME occurs at the molecular level is missing. Herein, using an interdisciplinary approach that included homology modeling, MD simulations, and biophysical and biochemical characterizations, we investigated the molecular basis of PME3 inhibition by PMEI7 in Arabidopsis thaliana Our complementary approach uncovered how changes in the protonation of amino acids at the complex interface shift the network of interacting residues between intermolecular and intramolecular. These shifts ultimately regulate the stability of the PME3-PMEI7 complex and the inhibition of the PME as a function of the pH. These findings suggest a general model of how pH-dependent proteinaceous inhibitors function. Moreover, they enhance our understanding of how PMEs may be regulated by pH and provide new insights into how this regulation may control the physical properties and structure of the plant cell wall.


Subject(s)
Arabidopsis Proteins/metabolism , Carboxylic Ester Hydrolases/chemistry , Carboxylic Ester Hydrolases/metabolism , Amino Acid Sequence/genetics , Arabidopsis/metabolism , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/genetics , Carboxylic Ester Hydrolases/antagonists & inhibitors , Carboxylic Ester Hydrolases/genetics , Cell Membrane/metabolism , Cell Wall/metabolism , Gene Expression Regulation, Plant/genetics , Hydrogen-Ion Concentration , Pectins/metabolism , Plant Proteins/metabolism , Protein Interaction Domains and Motifs
16.
J Biol Chem ; 292(1): 146-160, 2017 Jan 06.
Article in English | MEDLINE | ID: mdl-27881673

ABSTRACT

The liver plays a key role in cholesterol metabolism. Impaired hepatic cholesterol homeostasis causes intracellular free cholesterol accumulation and hepatocyte injury. Sortilin 1 (SORT1) is a lysosomal trafficking receptor that was identified by genome-wide association studies (GWAS) as a novel regulator of cholesterol metabolism in humans. Here we report that SORT1 deficiency protected against cholesterol accumulation-induced liver injury and inflammation in mice. Using an LC-MS/MS-based proteomics approach, we identified liver carboxylesterase 1 (CES1) as a novel SORT1-interacting protein. Mechanistic studies further showed that SORT1 may regulate CES1 lysosomal targeting and degradation and that SORT1 deficiency resulted in higher liver CES1 protein abundance. Previous studies have established an important role of hepatic CES1 in promoting intracellular cholesterol mobilization, cholesterol efflux, and bile acid synthesis. Consistently, high cholesterol atherogenic diet-challenged Sort1 knock-out mice showed less hepatic free cholesterol accumulation, increased bile acid synthesis, decreased biliary cholesterol secretion, and the absence of gallstone formation. SORT1 deficiency did not alter hepatic ceramide and fatty acid metabolism in high cholesterol atherogenic diet-fed mice. Finally, knockdown of liver CES1 in mice markedly increased the susceptibility to high cholesterol diet-induced liver injury and abolished the protective effect against cholesterol lipotoxicity in Sort1 knock-out mice. In summary, this study identified a novel SORT1-CES1 axis that regulates cholesterol-induced liver injury, which provides novel insights that improve our current understanding of the molecular links between SORT1 and cholesterol metabolism. This study further suggests that therapeutic inhibition of SORT1 may be beneficial in improving hepatic cholesterol homeostasis in metabolic and inflammatory liver diseases.


Subject(s)
Adaptor Proteins, Vesicular Transport/physiology , Carboxylic Ester Hydrolases/metabolism , Chemical and Drug Induced Liver Injury/pathology , Cholesterol/toxicity , Hepatocytes/pathology , Inflammation/pathology , Animals , Carboxylic Ester Hydrolases/antagonists & inhibitors , Carboxylic Ester Hydrolases/genetics , Cells, Cultured , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/metabolism , Female , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Inflammation/etiology , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Small Interfering/genetics
17.
Cell Physiol Biochem ; 45(3): 1013-1022, 2018.
Article in English | MEDLINE | ID: mdl-29428958

ABSTRACT

BACKGROUND/AIMS: Neuropathy target esterase (NTE, also known as neurotoxic esterase) is proven to deacylate phosphatidylcholine (PC) to glycerophosphocholine as a phospholipase B. Recently; studies showed that artificial phosphatidylserine/PC microvesicles can induce preeclampsia (PE)-like changes in pregnant mice. However, it is unclear whether NTE plays a key role in the pathology of PE, a pregnancy-related disease, which was characterized by deficient trophoblast invasion and reduced trophoblast-mediated remodeling of spiral arteries. The aim of this study was to investigate the expression pattern of NTE in the placenta from women with PE and normal pregnancy, and the molecular mechanism of NTE involved in the development of PE. METHODS: NTE expression levels in placentas from 20 pregnant women with PE and 20 healthy pregnant women were detected using quantitative PCR and immunohistochemistry staining. The effect of NTE on trophoblast migration and invasion and the underlying mechanisms were examined in HTR-8/SVneo cell lines by transfection method. RESULTS: NTE mRNA and protein expression levels were significantly decreased in preeclamptic placentas than normal control. Over-expression of NTE in HTR-8/SVneo cells significantly promoted trophoblast cells migration and invasion and was associated with increased MMP-9 levels. Conversely, shRNA-mediated down-regulation of NTE markedly inhibited the cell migration and invasion. In addition, silencing NTE reduced the MMP-9 activity and phosphorylated Erk1/2 and AKT levels. CONCLUSIONS: Our results suggest that the decreased NTE may contribute to the development of PE through impairing trophoblast invasion by down-regulating MMP-9 via the Erk1/2 and AKT signaling pathway.


Subject(s)
Carboxylic Ester Hydrolases/metabolism , Matrix Metalloproteinase 9/metabolism , Pre-Eclampsia/pathology , Adult , Carboxylic Ester Hydrolases/antagonists & inhibitors , Carboxylic Ester Hydrolases/genetics , Cell Line , Cell Movement , Down-Regulation , Female , Gestational Age , Humans , Male , Matrix Metalloproteinase 2/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Placenta/metabolism , Pre-Eclampsia/metabolism , Pregnancy , RNA Interference , Signal Transduction , Trophoblasts/cytology , Trophoblasts/metabolism , Young Adult
18.
Plant Physiol ; 173(2): 1075-1093, 2017 02.
Article in English | MEDLINE | ID: mdl-28034952

ABSTRACT

The fine-tuning of the degree of methylesterification of cell wall pectin is a key to regulating cell elongation and ultimately the shape of the plant body. Pectin methylesterification is spatiotemporally controlled by pectin methylesterases (PMEs; 66 members in Arabidopsis [Arabidopsis thaliana]). The comparably large number of proteinaceous pectin methylesterase inhibitors (PMEIs; 76 members in Arabidopsis) questions the specificity of the PME-PMEI interaction and the functional role of such abundance. To understand the difference, or redundancy, between PMEIs, we used molecular dynamics (MD) simulations to predict the behavior of two PMEIs that are coexpressed and have distinct effects on plant development: AtPMEI4 and AtPMEI9. Simulations revealed the structural determinants of the pH dependence for the interaction of these inhibitors with AtPME3, a major PME expressed in roots. Key residues that are likely to play a role in the pH dependence were identified. The predictions obtained from MD simulations were confirmed in vitro, showing that AtPMEI9 is a stronger, less pH-independent inhibitor compared with AtPMEI4. Using pollen tubes as a developmental model, we showed that these biochemical differences have a biological significance. Application of purified proteins at pH ranges in which PMEI inhibition differed between AtPMEI4 and AtPMEI9 had distinct consequences on pollen tube elongation. Therefore, MD simulations have proven to be a powerful tool to predict functional diversity between PMEIs, allowing the discovery of a strategy that may be used by PMEIs to inhibit PMEs in different microenvironmental conditions and paving the way to identify the specific role of PMEI diversity in muro.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Carboxylic Ester Hydrolases/antagonists & inhibitors , Carboxylic Ester Hydrolases/metabolism , Computational Biology/methods , Enzyme Inhibitors/metabolism , Arabidopsis Proteins/genetics , Cell Wall/metabolism , Escherichia coli/metabolism , Gene Expression Regulation, Plant , Germination , Hydrogen Bonding , Hydrogen-Ion Concentration , Hypocotyl/growth & development , Hypocotyl/metabolism , Molecular Dynamics Simulation , Plant Roots/growth & development , Plant Roots/metabolism , Pollen Tube/growth & development , Pollen Tube/metabolism , Recombinant Proteins/metabolism
19.
Bioorg Med Chem Lett ; 28(6): 997-1000, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29503023

ABSTRACT

It is necessary to consider the affinity of prodrugs for metabolic enzymes for efficient activation of the prodrugs in the body. Although many prodrugs have been synthesized with consideration of these chemical properties, there has been little study on the design of a structure with consideration of biological properties such as substrate recognition ability of metabolic enzymes. In this report, chemical synthesis and evaluation of indomethacin prodrugs metabolically activated by human carboxylesterase 1 (hCES1) are described. The synthesized prodrugs were subjected to hydrolysis reactions in solutions of human liver microsomes (HLM), human intestine microsomes (HIM) and hCES1, and the hydrolytic parameters were investigated to evaluate the hydrolytic rates of these prodrugs and to elucidate the substrate recognition ability of hCES1. It was found that the hydrolytic rates greatly change depending on the steric hindrance and stereochemistry of the ester in HLM, HIM and hCES1 solutions. Furthermore, in a hydrolysis reaction catalyzed by hCES1, the Vmax value of n-butyl thioester with chemically high reactivity was significantly lower than that of n-butyl ester.


Subject(s)
Carboxylic Ester Hydrolases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Esters/pharmacology , Indomethacin/pharmacology , Prodrugs/pharmacology , Carboxylic Ester Hydrolases/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Esters/chemistry , Esters/metabolism , Humans , Indomethacin/chemistry , Indomethacin/metabolism , Molecular Structure , Prodrugs/chemistry , Prodrugs/metabolism , Structure-Activity Relationship
20.
J Nat Prod ; 81(11): 2410-2418, 2018 11 26.
Article in English | MEDLINE | ID: mdl-30351923

ABSTRACT

The roots of Salvia miltiorrhiza ("Danshen") have been used in Chinese herbal medicine for centuries for a host of different conditions. While the exact nature of the active components of this material are unknown, large amounts of tanshinones are present in extracts derived from these samples. Recently, the tanshinones have been demonstrated to be potent human carboxylesterase (CE) inhibitors, with the ability to modulate the biological activity of esterified drugs. During the course of these studies, we also identified more active, irreversible inhibitors of these enzymes. We have purified, identified, and synthesized these molecules and confirmed them to be the anhydride derivatives of the tanshinones. These compounds are exceptionally potent inhibitors ( Ki < 1 nM) and can inactivate human CEs both in vitro and in cell culture systems and can modulate the metabolism of the esterified drug oseltamivir. Therefore, the coadministration of Danshen extracts with drugs that contain the ester chemotype should be minimized since, not only is transient inhibition of CEs observed with the tanshinones, but also prolonged irreversible inhibition arises via interaction with the anhydrides.


Subject(s)
Abietanes/pharmacology , Carboxylic Ester Hydrolases/antagonists & inhibitors , Enzyme Inhibitors/isolation & purification , Salvia miltiorrhiza/chemistry , Abietanes/chemistry , Abietanes/isolation & purification , Animals , Cell Line, Tumor , Drugs, Chinese Herbal/chemistry , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Molecular Structure , Oseltamivir/antagonists & inhibitors , Spodoptera
SELECTION OF CITATIONS
SEARCH DETAIL