Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 23(24)2022 Dec 07.
Article in English | MEDLINE | ID: mdl-36555144

ABSTRACT

Fatty acid amide hydrolase (FAAH) plays a key role in the control of cannabinoid signaling and it represents a promising therapeutic strategy for the treatment of a wide range of diseases, including neuropathic pain and chronic inflammation. Starting from kinetics experiments carried out in our previous work for the most potent inhibitor 2-amino-3-chloropyridine amide (TPA14), we have investigated its non-competitive mechanism of action using molecular dynamics, thermodynamic integration and QM-MM/GBSA calculations. The computational studies highlighted the impact of mutations on the receptor binding pockets and elucidated the molecular basis of the non-competitive inhibition mechanism of TPA14, which prevents the endocannabinoid anandamide (AEA) from reaching its pro-active conformation. Our study provides a rationale for the design of non-competitive potent FAAH inhibitors for the treatment of neuropathic pain and chronic inflammation.


Subject(s)
Endocannabinoids , Neuralgia , Humans , Endocannabinoids/metabolism , Neuralgia/drug therapy , Amidohydrolases/metabolism , Polyunsaturated Alkamides/metabolism , Inflammation/drug therapy , Molecular Dynamics Simulation
2.
Molecules ; 27(17)2022 Aug 23.
Article in English | MEDLINE | ID: mdl-36080139

ABSTRACT

Here we report a small library of hydrazinocarbonyl-ureido and thioureido benzenesulfonamide derivatives, designed and synthesized as potent and selective human carbonic anhydrase inhibitors (hCAIs). The synthesized compounds were evaluated against isoforms hCA I, II, IX and XII using acetazolamide (AAZ) as standard inhibitor. Several urea and thiourea derivatives showed inhibitory activity at low nanomolar levels with selectivity against the cytosolic hCA II isoform, as well as the transmembrane, tumor-associated enzymes hCA IX and XII. The thiourea derivatives showed enhanced potency as compared to urea analogues. Additionally, eight compounds 5g, 5m, 5o, 5q, 6l, 6j, 6o and 6u were selected for docking analysis on isoform I, II, IX, XII to illustrate the potential interaction with the enzyme to better understand the activity against the different isoforms.


Subject(s)
Carbonic Anhydrase I , Carbonic Anhydrases , Carbonic Anhydrase IX , Carbonic Anhydrase Inhibitors/pharmacology , Carbonic Anhydrases/metabolism , Humans , Molecular Structure , Protein Isoforms , Structure-Activity Relationship , Sulfanilamide , Sulfonamides/pharmacology , Thiourea/pharmacology , Urea
3.
Chem Sci ; 13(13): 3674-3687, 2022 Mar 30.
Article in English | MEDLINE | ID: mdl-35432906

ABSTRACT

We report a fast-track computationally driven discovery of new SARS-CoV-2 main protease (Mpro) inhibitors whose potency ranges from mM for the initial non-covalent ligands to sub-µM for the final covalent compound (IC50 = 830 ± 50 nM). The project extensively relied on high-resolution all-atom molecular dynamics simulations and absolute binding free energy calculations performed using the polarizable AMOEBA force field. The study is complemented by extensive adaptive sampling simulations that are used to rationalize the different ligand binding poses through the explicit reconstruction of the ligand-protein conformation space. Machine learning predictions are also performed to predict selected compound properties. While simulations extensively use high performance computing to strongly reduce the time-to-solution, they were systematically coupled to nuclear magnetic resonance experiments to drive synthesis and for in vitro characterization of compounds. Such a study highlights the power of in silico strategies that rely on structure-based approaches for drug design and allows the protein conformational multiplicity problem to be addressed. The proposed fluorinated tetrahydroquinolines open routes for further optimization of Mpro inhibitors towards low nM affinities.

4.
J Enzyme Inhib Med Chem ; 36(1): 940-953, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33896320

ABSTRACT

Compounds combining dual inhibitory action against FAAH and cyclooxygenase (COX) may be potentially useful analgesics. Here, we describe a novel flurbiprofen analogue, N-(3-bromopyridin-2-yl)-2-(2-fluoro-(1,1'-biphenyl)-4-yl)propanamide (Flu-AM4). The compound is a competitive, reversible inhibitor of FAAH with a Ki value of 13 nM and which inhibits COX activity in a substrate-selective manner. Molecular modelling suggested that Flu-AM4 optimally fits a hydrophobic pocket in the ACB region of FAAH, and binds to COX-2 similarly to flurbiprofen. In vivo studies indicated that at a dose of 10 mg/kg, Flu-AM4 was active in models of prolonged (formalin) and neuropathic (chronic constriction injury) pain and reduced the spinal expression of iNOS, COX-2, and NFκB in the neuropathic model. Thus, the present study identifies Flu-AM4 as a dual-action FAAH/substrate-selective COX inhibitor with anti-inflammatory and analgesic activity in animal pain models. These findings underscore the potential usefulness of such dual-action compounds.


Subject(s)
Amides/pharmacology , Amidohydrolases/antagonists & inhibitors , Analgesics/pharmacology , Cyclooxygenase 2/metabolism , Enzyme Inhibitors/pharmacology , Flurbiprofen/pharmacology , Amides/chemical synthesis , Amides/chemistry , Amidohydrolases/metabolism , Analgesics/chemical synthesis , Analgesics/chemistry , Animals , Dose-Response Relationship, Drug , Drug Design , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Flurbiprofen/chemical synthesis , Flurbiprofen/chemistry , Male , Mice , Mice, Inbred Strains , Models, Molecular , Molecular Structure , Quantum Theory , Rats , Rats, Sprague-Dawley , Rats, Wistar , Static Electricity , Structure-Activity Relationship
6.
Bioorg Chem ; 101: 104034, 2020 08.
Article in English | MEDLINE | ID: mdl-32599361

ABSTRACT

In experimental animals, inhibition of fatty acid amide hydrolase (FAAH) reduces the gastrointestinal damage produced by non-steroidal anti-inflammatory agents that act by inhibition of cyclooxygenase (COX). This suggests that compounds able to inhibit both enzymes may be potentially useful therapeutic agents. In the present study, we have investigated eight novel amide analogues of carprofen, ketoprofen and fenoprofen as potential FAAH/COX dual action inhibitors. Carpro-AM1 (2-(6-Chloro-9H-carbazol-2-yl)-N-(3-methylpyridin-2-yl)propenamide) and Carpro-AM6 (2-(6-Chloro-9H-carbazol-2-yl)-N-(3-chloropyridin-2-yl)propenamide) were found to be fully reversible inhibitors of the hydrolysis of 0.5 µM [3H]anandamide in rat brain homogenates with IC50 values of 94 and 23 nM, respectively, i.e. 2-3 orders of magnitude more potent than carprofen in this respect. Both compounds inhibited the cyclooxygenation of arachidonic acid by ovine COX-1, and were more potent inhibitors of human recombinant COX-2 when 2-arachidonoylglycerol was used as substrate than when arachidonic acid was used. It is concluded that Carpro-AM1 and Carpro-AM6 are dual-acting FAAH/substrate-selective COX inhibitors.


Subject(s)
Amidohydrolases/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Carbazoles/therapeutic use , Cyclooxygenase Inhibitors/therapeutic use , Amidohydrolases/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Carbazoles/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Humans , Mice
7.
J Chem Inf Model ; 60(9): 4231-4245, 2020 09 28.
Article in English | MEDLINE | ID: mdl-32364713

ABSTRACT

The accuracy of structure-based (SB) virtual screening (VS) is heavily affected by the scoring function used to rank a library of screened compounds. Even in cases where the docked pose agrees with the experimental binding mode of the ligand, the limitations of current scoring functions may lead to sensible inaccuracies in the ability to discriminate between actives and inactives. In this context, the combination of SB and ligand-based (LB) molecular similarity may be a promising strategy to increase the hit rates in VS. This study explores different strategies that aim to exploit the synergy between LB and SB methods in order to mitigate the limitations of these techniques, and to enhance the performance of VS studies by means of a balanced combination between docking scores and three-dimensional (3D) similarity. Particularly, attention is focused to the use of measurements of molecular similarity with PharmScreen, which exploits the 3D distribution of atomic lipophilicity determined from quantum mechanical-based continuum solvation calculations performed with the MST model, in conjunction with three docking programs: Glide, rDock, and GOLD. Different strategies have been explored to combine the information provided by docking and similarity measurements for re-ranking the screened ligands. For a benchmarking of 44 datasets, including 41 targets, the hybrid methods increase the identification of active compounds, according to the early (ROCe%) and total (AUC) enrichment metrics of VS, compared to pure LB and SB methods. Finally, the hybrid approaches are also more effective in enhancing the chemical diversity of active compounds. The datasets employed in this work are available in https://github.com/Pharmacelera/Molecular-Similarity-and-Docking.


Subject(s)
Ligands , Molecular Docking Simulation , Protein Binding
8.
J Enzyme Inhib Med Chem ; 35(1): 815-823, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32200655

ABSTRACT

Inhibition of fatty acid amide hydrolase (FAAH) reduces the gastrointestinal damage produced by non-steroidal anti-inflammatory agents such as sulindac and indomethacin in experimental animals, suggesting that a dual-action FAAH-cyclooxygenase (COX) inhibitor could have useful therapeutic properties. Here, we have investigated 12 novel amide analogues of ibuprofen as potential dual-action FAAH/COX inhibitors. N-(3-Bromopyridin-2-yl)-2-(4-isobutylphenyl)propanamide (Ibu-AM68) was found to inhibit the hydrolysis of [3H]anandamide by rat brain homogenates by a reversible, mixed-type mechanism of inhibition with a Ki value of 0.26 µM and an α value of 4.9. At a concentration of 10 µM, the compound did not inhibit the cyclooxygenation of arachidonic acid by either ovine COX-1 or human recombinant COX-2. However, this concentration of Ibu-AM68 greatly reduced the ability of the COX-2 to catalyse the cyclooxygenation of the endocannabinoid 2-arachidonoylglycerol. It is concluded that Ibu-AM68 is a dual-acting FAAH/substrate-selective COX inhibitor.


Subject(s)
Amides/pharmacology , Amidohydrolases/antagonists & inhibitors , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Enzyme Inhibitors/pharmacology , Ibuprofen/pharmacology , Amides/chemical synthesis , Amides/chemistry , Amidohydrolases/metabolism , Animals , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Ibuprofen/chemical synthesis , Ibuprofen/chemistry , Molecular Docking Simulation , Molecular Structure , Rats , Rats, Sprague-Dawley , Rats, Wistar , Structure-Activity Relationship
9.
Bioorg Chem ; 98: 103728, 2020 05.
Article in English | MEDLINE | ID: mdl-32182519

ABSTRACT

Herein we report on a new series of hydrazidoureidobenzensulfonamides investigated as inhibitors of the cytosolic human (h) hCA I and II isoforms, as well as the transmembrane, tumor-associated enzymes hCA IX and XII. The reported derivatives contain a 4-substituted piperidine fragment in which the hydrazidoureido linker has been involved as spacer between the benzenesulfonamide fragment which binds the zinc ion from the active site, and the tail of the inhibitor. Depending on the substitution pattern at the piperidine ring, low nanomolar inhibitors were detected against hCA II, hCA IX and hCA XII, making the new class of sulfonamides of interest for various pharmacologic applications.


Subject(s)
Carbonic Anhydrase Inhibitors/pharmacology , Hydrazines/pharmacology , Molecular Docking Simulation , Piperidines/pharmacology , Sulfonamides/pharmacology , Antigens, Neoplasm/metabolism , Carbonic Anhydrase II/antagonists & inhibitors , Carbonic Anhydrase II/metabolism , Carbonic Anhydrase IX/antagonists & inhibitors , Carbonic Anhydrase IX/metabolism , Carbonic Anhydrase Inhibitors/chemical synthesis , Carbonic Anhydrase Inhibitors/chemistry , Carbonic Anhydrases/metabolism , Dose-Response Relationship, Drug , Humans , Hydrazines/chemistry , Molecular Structure , Piperidines/chemistry , Structure-Activity Relationship , Sulfonamides/chemistry , Benzenesulfonamides
10.
Eur J Med Chem ; 186: 111896, 2020 Jan 15.
Article in English | MEDLINE | ID: mdl-31784185

ABSTRACT

We report here a thorough structure-activity relationship (SAR) with piperazinylureido sulfamates as inhibitors of human (h) carbonic anhydrase (CA, EC 4.2.1.1). A SAR investigation over the structure of reported anti-cancer zinc-binder CAIs such as SLC-0111 and S4 was carried out by including the urea outer nitrogen atom into a substituted piperazine ring reducing the linker flexibility. The derivatives were assessed for the inhibition of CA I, II and IV (off-target isoforms) and the tumor-associated CA IX (anticancer drug target). CA I and IV were not effectively inhibited, whereas many low nanomolar inhibitors were evidenced against CA II (KIs in the range of 1.0-705.5 nM), and IX (KIs in the range of 0.91-155.9 nM). Interestingly, a subset of CA II/IX selective inhibitors was detected which might represent interesting lead for the development of new anticancer strategies.


Subject(s)
Carbonic Anhydrase Inhibitors/pharmacology , Carbonic Anhydrases/metabolism , Piperazines/pharmacology , Sulfonamides/pharmacology , Sulfonic Acids/pharmacology , Urea/pharmacology , Carbonic Anhydrase Inhibitors/chemical synthesis , Carbonic Anhydrase Inhibitors/chemistry , Dose-Response Relationship, Drug , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Molecular Docking Simulation , Molecular Structure , Piperazines/chemistry , Structure-Activity Relationship , Sulfonamides/chemistry , Sulfonic Acids/chemistry , Urea/analogs & derivatives , Urea/chemistry
11.
Eur J Med Chem ; 182: 111638, 2019 Nov 15.
Article in English | MEDLINE | ID: mdl-31472471

ABSTRACT

Four new series of aromatic sulfamates were synthesized and investigated for the inhibition of four human (h) isoforms of zinc enzyme carbonic anhydrase (CA, EC 4.2.1.1), hCA I, II, IX, and XII. The reported derivatives, obtained by a sulfamoylation reaction of the corresponding phenolic precursors, bear 3,5-diarylpyrazoline moieties as spacers between the benzenesulfamate fragment which binds the zinc ion from the active site, and the tail of the inhibitor. Pyrazolines are biologically privileged scaffolds, endowed with versatile biological activity, such as an anti-proliferative action. The derivatives were tested for the inhibition of the cytosolic, hCA I and II (off target isoforms) and the trans-membrane, tumor-associated hCA IX and XII enzymes (anticancer drug targets). Generally, hCA I was not effectively inhibited, whereas many low nanomolar inhibitors were evidenced against hCA II (KIs in the range of 0.42-90.1 nM), IX (KIs in the range of 0.72-63.6 nM), and XII (KIs in the range of 0.88-85.2 nM). The best substitution fragments at the pyrazoline ring included for CA II a 4-sulfamic group on the 3-aryl and halogens on the 5-aryl or a methoxy group on the 3-aryl and a 4-sulfamate group on the 5-aryl; for CA IX and CA XII they included the sulfamic group on the 3- or 4-position of the 5-aryl and an electronwithdrawing group on the 4-postion of the 3-aryl ring.


Subject(s)
Carbonic Anhydrase Inhibitors/pharmacology , Carbonic Anhydrases/metabolism , Pyrazoles/pharmacology , Sulfonic Acids/pharmacology , Carbonic Anhydrase Inhibitors/chemical synthesis , Carbonic Anhydrase Inhibitors/chemistry , Dose-Response Relationship, Drug , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Models, Molecular , Molecular Structure , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Structure-Activity Relationship , Sulfonic Acids/chemical synthesis , Sulfonic Acids/chemistry
12.
Eur J Med Chem ; 182: 111614, 2019 Nov 15.
Article in English | MEDLINE | ID: mdl-31422224

ABSTRACT

Two new piperazinyl-ureido single ring aryl sulfamate-based inhibitor series were designed against the emerging oncology drug target steroid sulfatase (STS), for which there are existing potent steroidal and non-steroidal agents in clinical trials. 4-(Piperazinocarbonyl)aminosulfamates (5-31) were obtained by reacting 4-hydroxyarylamines with phenylchloroformate, subsequent sulfamoylation of the resulting hydroxyarylcarbamates and coupling of the product with 1-substituted piperazines. Pyrimidinyl-piperazinourea sulfamates (35-42) were synthesized by pyrimidine ring closure of 4-Boc-piperazine-1-carboxamidine with 3-(dimethylamino)propenones, deprotection and coupling with the sulfamoylated building block. Target ureidosulfamates 5-31 and 35-42 were evaluated both as STS inhibitors in vitro using a lysate of JEG-3 human placenta choriocarcinoma cell line and in a whole cell assay. SAR conclusions were drawn from both series. In series 35-42 the best inhibitory activity is related to the presence of a benzofuryl on the pyrimidine ring. In series 5-31 the best inhibitory activity was shown by the ureas bearing 4-chlorophenyl, 3,4-dichlorophenyl groups or aliphatic chains at the piperazino 4-nitrogen displaying IC50 in the 33-94 nM concentration range. Final optimization to the low nanomolar level was achieved through substitution of the arylsulfamate ring with halogens. Four halogenated arylsulfamates of high potency were achieved and two of these 19 and 20 had IC50 values of 5.1 and 8.8 nM respectively and are attractive for potential in vivo evaluation and further development. We demonstrate the optimization of this new series to low nanomolar potency, employing fluorine substitution, providing potent membrane permeant inhibitors with further development potential indicating piperazinyl-ureido aryl sulfamate derivatives as an attractive new class of STS inhibitors.


Subject(s)
Enzyme Inhibitors/pharmacology , Piperazines/pharmacology , Steryl-Sulfatase/antagonists & inhibitors , Sulfonic Acids/pharmacology , Urea/pharmacology , Cell Line, Tumor , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Molecular Structure , Piperazines/chemical synthesis , Piperazines/chemistry , Steryl-Sulfatase/metabolism , Structure-Activity Relationship , Sulfonic Acids/chemical synthesis , Sulfonic Acids/chemistry , Urea/analogs & derivatives , Urea/chemistry
13.
J Enzyme Inhib Med Chem ; 34(1): 562-576, 2019 Dec.
Article in English | MEDLINE | ID: mdl-30688118

ABSTRACT

Fatty Acid Amide Hydrolase (FAAH) is a serine hydrolase that plays a key role in controlling endogenous levels of endocannabinoids. FAAH inhibition is considered a powerful approach to enhance the endocannabinoid signalling, and therefore it has been largely studied as a potential target for the treatment of neurological disorders such as anxiety or depression, or of inflammatory processes. We present two novel series of amide derivatives of ibuprofen designed as analogues of our reference FAAH inhibitor Ibu-AM5 to further explore its structure-activity relationships. In the new amides, the 2-methylpyridine moiety of Ibu-AM5 was substituted by benzylamino and piperazinoaryl moieties. The obtained benzylamides and piperazinoarylamides showed FAAH inhibition ranging from the low to high micromolar potency. The binding of the new amides in the active site of FAAH, estimated using the induced fit protocol, indicated arylpiperazinoamides binding the ACB channel and the cytosolic port, and benzylamides binding the ACB channel.


Subject(s)
Amides/pharmacology , Amidohydrolases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Ibuprofen/pharmacology , Piperazine/pharmacology , Amides/chemistry , Amidohydrolases/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Ibuprofen/chemical synthesis , Ibuprofen/chemistry , Models, Molecular , Molecular Structure , Piperazine/chemistry , Structure-Activity Relationship
14.
J Chem Inf Model ; 58(8): 1596-1609, 2018 08 27.
Article in English | MEDLINE | ID: mdl-30010337

ABSTRACT

Molecular alignment is a standard procedure for three-dimensional (3D) similarity measurements and pharmacophore elucidation. This process is influenced by several factors, such as the physicochemical descriptors utilized to account for the molecular determinants of biological activity and the reference templates. Relying on the hypothesis that the maximal achievable binding affinity for a drug-like molecule is largely due to desolvation, we explore a novel strategy for 3D molecular overlays that exploits the partitioning of molecular hydrophobicity into atomic contributions in conjunction with information about the distribution of hydrogen-bond (HB) donor/acceptor groups. A brief description of the method, as implemented in the software package PharmScreen, including the derivation of the fractional hydrophobic contributions within the quantum mechanical version of the Miertus-Scrocco-Tomasi (MST) continuum model, and the procedure utilized for the optimal superposition between molecules, is presented. The computational procedure is calibrated by using a data set of 402 molecules pertaining to 14 distinct targets taken from the literature and validated against the AstraZeneca test, which comprises 121 experimentally derived sets of molecular overlays. The results point out the suitability of the MST-based hydrophobic parameters for generating molecular overlays, as correct predictions were obtained for 94%, 79%, and 54% of the molecules classified into easy, moderate, and hard sets, respectively. Moreover, the results point out that this accuracy is attained at a much lower degree of identity between the templates used by hydrophobic/HB fields and electrostatic/steric ones. These findings support the usefulness of the hydrophobic/HB descriptors to generate complementary overlays that may be valuable to rationalize structure-activity relationships and for virtual screening campaigns.


Subject(s)
Computer-Aided Design , Drug Design , Drug Discovery/methods , Pharmaceutical Preparations/chemistry , Small Molecule Libraries/chemistry , Animals , Databases, Protein , Humans , Hydrophobic and Hydrophilic Interactions , Models, Chemical , Models, Molecular , Molecular Conformation , Proteins/metabolism , Quantitative Structure-Activity Relationship , Small Molecule Libraries/pharmacology , Static Electricity
15.
Pathog Dis ; 75(6)2017 08 31.
Article in English | MEDLINE | ID: mdl-28859311

ABSTRACT

In the continuous effort to identify new HIV-1 inhibitors endowed with innovative mechanisms, the dual inhibition of different viral functions would provide a significant advantage against drug-resistant variants. The HIV-1 reverse transcriptase (RT)-associated ribonuclease H (RNase H) is the only viral-encoded enzymatic activity that still lacks an efficient inhibitor. We synthesized a library of 3,5-diamino-N-aryl-1H-pyrazole-4-carbothioamide and 4-amino-5-benzoyl-N-phenyl-2-(substituted-amino)-1H-pyrrole-3-carbothioamide derivatives and tested them against RNase H activity. We identified the pyrazolecarbothioamide derivative A15, able to inhibit viral replication and both RNase H and RNA-dependent DNA polymerase (RDDP) RT-associated activities in the low micromolar range. Docking simulations hypothesized its binding to two RT pockets. Site-directed mutagenesis experiments showed that, with respect to wt RT, V108A substitution strongly reduced A15 IC50 values (12.6-fold for RNase H inhibition and 4.7-fold for RDDP), while substitution A502F caused a 9.0-fold increase in its IC50 value for RNase H, not affecting the RDDP inhibition, reinforcing the hypothesis of a dual-site inhibition. Moreover, A15 retained good inhibition potency against three non-nucleoside RT inhibitor (NNRTI)-resistant enzymes, confirming a mode of action unrelated to NNRTIs and suggesting its potential as a lead compound for development of new HIV-1 RT dual inhibitors active against drug-resistant viruses.


Subject(s)
Anti-HIV Agents/pharmacology , Enzyme Inhibitors/pharmacology , HIV Reverse Transcriptase/antagonists & inhibitors , Pyrazoles/pharmacology , Ribonuclease H/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Thioamides/pharmacology , Amino Acid Substitution , Anti-HIV Agents/chemistry , Anti-HIV Agents/isolation & purification , Binding Sites , Cell Line , Cloning, Molecular , Drug Design , Drug Resistance, Viral/drug effects , Drug Resistance, Viral/genetics , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/isolation & purification , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression Regulation , HIV Reverse Transcriptase/chemistry , HIV Reverse Transcriptase/genetics , HIV Reverse Transcriptase/metabolism , HIV-1/drug effects , HIV-1/enzymology , HIV-1/growth & development , Humans , Molecular Docking Simulation , Mutagenesis, Site-Directed , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Pyrazoles/chemical synthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Ribonuclease H/chemistry , Ribonuclease H/genetics , Ribonuclease H/metabolism , Small Molecule Libraries/chemical synthesis , T-Lymphocytes/drug effects , T-Lymphocytes/virology , Thioamides/chemical synthesis
16.
Eur J Med Chem ; 136: 523-542, 2017 Aug 18.
Article in English | MEDLINE | ID: mdl-28535469

ABSTRACT

Fatty acid amide hydrolase (FAAH) has a key role in the control of the cannabinoid signaling, through the hydrolysis of the endocannabinoids anandamide and in some tissues 2-arachidonoylglycerol. FAAH inhibition represents a promising strategy to activate the cannabinoid system, since it does not result in the psychotropic and peripheral side effects characterizing the agonists of the cannabinoid receptors. Here we present the discovery of a novel class of profen derivatives, the N-(heteroaryl)-2-(4-((2-(trifluoromethyl)pyridin-4-yl)amino)phenyl)propanamides, as FAAH inhibitors. Enzymatic assays showed potencies toward FAAH ranging from nanomolar to micromolar range, and the most compounds lack activity toward the two isoforms of cyclooxygenase. Extensive structure-activity studies and the definition of the binding mode for the lead compound of the series are also presented. Kinetic assays in rat and mouse FAAH on selected compounds of the series demonstrated that slight modifications of the chemical structure could influence the binding mode and give rise to competitive (TPA1) or non-competitive (TPA14) inhibition modes.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Ibuprofen/pharmacology , Amidohydrolases/metabolism , Animals , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Ibuprofen/chemical synthesis , Ibuprofen/chemistry , Male , Mice , Models, Molecular , Molecular Structure , Quantum Theory , Rats , Rats, Sprague-Dawley , Rats, Wistar , Structure-Activity Relationship , Thermodynamics
17.
J Mol Graph Model ; 70: 140-152, 2016 11.
Article in English | MEDLINE | ID: mdl-27723562

ABSTRACT

With the present work we quantitatively studied the modellability of the inactive state of Class A G protein-coupled receptors (GPCRs). Specifically, we constructed models of one of the Class A GPCRs for which structures solved in the inactive state are available, namely the ß2 AR, using as templates each of the other class members for which structures solved in the inactive state are also available. Our results showed a detectable linear correlation between model accuracy and model/template sequence identity. This suggests that the likely accuracy of the homology models that can be built for a given receptor can be generally forecasted on the basis of the available templates. We also probed whether sequence alignments that allow for the presence of gaps within the transmembrane domains to account for structural irregularities afford better models than the classical alignment procedures that do not allow for the presence of gaps within such domains. As our results indicated, although the overall differences are very subtle, the inclusion of internal gaps within the transmembrane domains has a noticeable a beneficial effect on the local structural accuracy of the domain in question.


Subject(s)
Models, Molecular , Receptors, G-Protein-Coupled/chemistry , Structural Homology, Protein , Amino Acid Sequence , Phylogeny , Protein Conformation , Sequence Alignment
18.
J Agric Food Chem ; 64(24): 4876-81, 2016 Jun 22.
Article in English | MEDLINE | ID: mdl-27249054

ABSTRACT

Different maleimide derivatives were synthesized and assayed for their in vitro activity on the soil inhabiting, plant-parasitic nematode Meloidogyne incognita, also known as root-knot nematode. The compounds maleimide, N-ethylmaleimide, N-isopropylmaleimide, and N-isobutylmaleimide showed the strongest nematicidal activity on the second stage juveniles of the root-knot nematode with EC50/72h values of 2.6 ± 1.3, 5.1 ± 3.4, 16.2 ± 5.4, and 19.0 ± 9.0 mg/L, respectively. We also determined the nematicidal activity of copper sulfate, finding an EC50 value of 48.6 ± 29.8 mg/L. When maleimide at 1 mg/L was tested in combination with copper sulfate at 50 mg/L, we observed 100% mortality of the nematodes. We performed a GC-MS metabolomics analysis after treating nematodes with maleimide at 8 mg/L for 24 h. This analysis revealed altered fatty acids and diglyceride metabolites such as oleic acid, palmitic acid, and 1-monopalmitin. Our results suggest that maleimide may be used as a new interesting building block for developing new nematicides in combination with copper salts.


Subject(s)
Antinematodal Agents/pharmacology , Maleimides/pharmacology , Tylenchoidea/drug effects , Animals , Antinematodal Agents/chemistry , Maleimides/chemistry , Structure-Activity Relationship , Tylenchoidea/genetics , Tylenchoidea/growth & development , Tylenchoidea/metabolism
19.
Molecules ; 21(5)2016 Apr 30.
Article in English | MEDLINE | ID: mdl-27144551

ABSTRACT

The synthesis and antiproliferative activity of new benzimidazole derivatives bearing an hydrazone mojety at the 2-position is described. The new N'-(4-arylidene)-1H-benzo[d]imidazole-2-carbohydrazides were evaluated for their cytostatic activity toward the murine leukemia (L1210), human T-cell leukemia (CEM), human cervix carcinoma (HeLa) and human pancreas carcinoma cells (Mia Paca-2). A preliminary structure-activity relationship could be defined. Some of the compounds possess encouraging and consistent antiproliferative activity, having IC50 values in the low micromolar range.


Subject(s)
Antineoplastic Agents/chemical synthesis , Benzimidazoles/chemical synthesis , Cell Proliferation/drug effects , Hydrazones/chemical synthesis , Animals , Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Cell Line, Tumor , Drug Design , Drug Screening Assays, Antitumor , HeLa Cells , Humans , Hydrazones/pharmacology , Mice , Molecular Structure , Structure-Activity Relationship
20.
PLoS One ; 10(11): e0142711, 2015.
Article in English | MEDLINE | ID: mdl-26565710

ABSTRACT

BACKGROUND: Combined fatty acid amide hydrolase (FAAH) and cyclooxygenase (COX) inhibition is a promising approach for pain-relief. The Flu-AM1 and Ibu-AM5 derivatives of flurbiprofen and ibuprofen retain similar COX-inhibitory properties and are more potent inhibitors of FAAH than the parent compounds. However, little is known as to the nature of their interaction with FAAH, or to the importance of their chirality. This has been explored here. METHODOLOGY/PRINCIPAL FINDINGS: FAAH inhibitory activity was measured in rat brain homogenates and in lysates expressing either wild-type or FAAH(T488A)-mutated enzyme. Molecular modelling was undertaken using both docking and molecular dynamics. The (R)- and (S)-enantiomers of Flu-AM1 inhibited rat FAAH with similar potencies (IC50 values of 0.74 and 0.99 µM, respectively), whereas the (S)-enantiomer of Ibu-AM5 (IC50 0.59 µM) was more potent than the (R)-enantiomer (IC50 5.7 µM). Multiple inhibition experiments indicated that both (R)-Flu-AM1 and (S)-Ibu-AM5 inhibited FAAH in a manner mutually exclusive to carprofen. Computational studies indicated that the binding site for the Flu-AM1 and Ibu-AM5 enantiomers was located between the acyl chain binding channel and the membrane access channel, in a site overlapping the carprofen binding site, and showed a binding mode in line with that proposed for carprofen and other non-covalent ligands. The potency of (R)-Flu-AM1 was lower towards lysates expressing FAAH mutated at the proposed carprofen binding area than in lysates expressing wild-type FAAH. CONCLUSIONS/SIGNIFICANCE: The study provides kinetic and structural evidence that the enantiomers of Flu-AM1 and Ibu-AM5 bind in the substrate channel of FAAH. This information will be useful in aiding the design of novel dual-action FAAH: COX inhibitors.


Subject(s)
Amidohydrolases/metabolism , Aminopyridines/chemistry , Brain/enzymology , Flurbiprofen/analogs & derivatives , Ibuprofen/analogs & derivatives , Prostaglandin-Endoperoxide Synthases/metabolism , Amidohydrolases/antagonists & inhibitors , Animals , Cyclooxygenase Inhibitors/chemistry , Drug Design , Flurbiprofen/chemistry , HeLa Cells , Humans , Hydrolysis , Ibuprofen/chemistry , Inhibitory Concentration 50 , Kinetics , Male , Mice , Molecular Dynamics Simulation , Protein Binding , Rats , Rats, Sprague-Dawley , Rats, Wistar , Recombinant Proteins/metabolism , Stereoisomerism
SELECTION OF CITATIONS
SEARCH DETAIL
...