Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
1.
Eur J Med Chem ; 250: 115169, 2023 Mar 15.
Article in English | MEDLINE | ID: mdl-36753881

ABSTRACT

A set of twenty-five thioxanthene-9-one and xanthene-9-one derivatives, that were previously shown to inhibit cholinesterases (ChEs) and amyloid ß (Aß40) aggregation, were evaluated for the inhibition of tau protein aggregation. All compounds exhibited a good activity, and eight of them (5-8, 10, 14, 15 and 20) shared comparable low micromolar inhibitory potency versus Aß40 aggregation and human acetylcholinesterase (AChE), while inhibiting human butyrylcholinesterase (BChE) even at submicromolar concentration. Compound 20 showed outstanding biological data, inhibiting tau protein and Aß40 aggregation with IC50 = 1.8 and 1.3 µM, respectively. Moreover, at 0.1-10 µM it also exhibited neuroprotective activity against tau toxicity induced by okadoic acid in human neuroblastoma SH-SY5Y cells, that was comparable to that of estradiol and PD38. In preliminary toxicity studies, these interesting results for compound 20 are somewhat conflicting with a narrow safety window. However, compound 10, although endowed with a little lower potency for tau and Aß aggregation inhibition additionally demonstrated good inhibition of ChEs and rather low cytotoxicity. Compound 4 is also worth of note for its high potency as hBChE inhibitor (IC50 = 7 nM) and for the three order of magnitude selectivity versus hAChE. Molecular modelling studies were performed to explain the different behavior of compounds 4 and 20 towards hBChE. The observed balance of the inhibitory potencies versus the relevant targets indicates the thioxanthene-9-one derivatives as potential MTDLs for AD therapy, provided that the safety window will be improved by further structural variations, currently under investigation.


Subject(s)
Alzheimer Disease , Neuroblastoma , Humans , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Butyrylcholinesterase/metabolism , Amyloid beta-Peptides/metabolism , Acetylcholinesterase/metabolism , Cholinesterase Inhibitors/chemistry , Molecular Structure , Structure-Activity Relationship , Neuroblastoma/drug therapy , Drug Design , Molecular Docking Simulation
2.
Methods Mol Biol ; 2558: 197-205, 2023.
Article in English | MEDLINE | ID: mdl-36169865

ABSTRACT

The quantitative structure-activity relationship method based on the three-dimensional structure of the target molecules (3D-QSAR) represents a valuable predictive tool for the design of new bioactive agents. Herewith, a detailed procedure is described which uses a pool comprising 67 derivatives substituted at position 4 and 7 of the common coumarin scaffold as a benchmark for deriving a predictive 3D-QSAR model employed for guiding the rational design of 10 new potent and selective MAO B inhibitors.


Subject(s)
Coumarins , Quantitative Structure-Activity Relationship , Coumarins/pharmacology , Monoamine Oxidase/metabolism
3.
ACS Med Chem Lett ; 12(12): 1920-1924, 2021 Dec 09.
Article in English | MEDLINE | ID: mdl-34917255

ABSTRACT

In the face of the clinical challenge posed by non-small cell lung cancer (NSCLC), the present need for new therapeutic approaches is genuine. Up to now, no proof existed that 17ß-hydroxysteroid dehydrogenase type 1 (17ß-HSD1) is a viable target for treating this disease. Synthesis of a rationally designed library of 2,5-disubstituted furan derivatives followed by biological screening led to the discovery of 17ß-HSD1 inhibitor 1, capable of fully inhibiting human NSCLC Calu-1 cell proliferation. Its pharmacological profile renders it eligible for further in vivo studies. The very high selectivity of 1 over 17ß-HSD2 was investigated, revealing a rational approach for the design of selective inhibitors. 17ß-HSD1 and 1 hold promise in fighting NSCLC.

4.
Molecules ; 23(2)2018 Jan 27.
Article in English | MEDLINE | ID: mdl-29382051

ABSTRACT

Many naturally occurring substances, traditionally used in popular medicines around the world, contain the coumarin moiety. Coumarin represents a privileged scaffold for medicinal chemists, because of its peculiar physicochemical features, and the versatile and easy synthetic transformation into a large variety of functionalized coumarins. As a consequence, a huge number of coumarin derivatives have been designed, synthesized, and tested to address many pharmacological targets in a selective way, e.g., selective enzyme inhibitors, and more recently, a number of selected targets (multitarget ligands) involved in multifactorial diseases, such as Alzheimer's and Parkinson's diseases. In this review an overview of the most recent synthetic pathways leading to mono- and polyfunctionalized coumarins will be presented, along with the main biological pathways of their biosynthesis and metabolic transformations. The many existing and recent reviews in the field prompted us to make some drastic selections, and therefore, the review is focused on monoamine oxidase, cholinesterase, and aromatase inhibitors, and on multitarget coumarins acting on selected targets of neurodegenerative diseases.


Subject(s)
Coumarins/chemical synthesis , Drug Design , Enzyme Inhibitors/chemical synthesis , Neuroprotective Agents/chemical synthesis , Nootropic Agents/chemical synthesis , Alzheimer Disease/drug therapy , Aromatase/chemistry , Aromatase/metabolism , Biotransformation , Cholinesterases/chemistry , Cholinesterases/metabolism , Coumarins/pharmacology , Enzyme Inhibitors/pharmacology , Humans , Molecular Structure , Monoamine Oxidase/chemistry , Monoamine Oxidase/metabolism , Neuroprotective Agents/pharmacology , Nootropic Agents/pharmacology , Parkinson Disease/drug therapy , Structure-Activity Relationship
5.
Biochem Pharmacol ; 143: 39-52, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28709952

ABSTRACT

The LAT1 transporter is acknowledged as a pharmacological target of tumours since it is strongly overexpressed in many human cancers. The purpose of this work was to find novel compounds exhibiting potent and prolonged inhibition of the transporter. To this aim, compounds based on dithiazole and dithiazine scaffold have been screened in the proteoliposome experimental model. Inhibition was tested on the antiport catalysed by hLAT1 as transport of extraliposomal [3H]histidine in exchange with intraliposomal histidine. Out of 59 compounds tested, 8 compounds, showing an inhibition higher than 90% at 100µM concentration, were subjected to dose-response analysis. Two of them exhibited IC50 lower than 1µM. Inhibition kinetics, performed on the two best inhibitors, indicated a mixed type of inhibition with respect to the substrate. Furthermore, inhibition of the transporter was still present after removal of the compounds from the reaction mixture, but was reversed on addition of dithioerythritol, a S-S reducing agent, indicating the formation of disulfide(s) between the compounds and the protein. Molecular docking of the two best inhibitors on the hLAT1 homology structural model, highlighted interaction with the substrate binding site and formation of a covalent bond with the residue C407. Indeed, the inhibition was impaired in the hLAT1 mutant C407A confirming the involvement of that Cys residue. Treatment of SiHa cells expressing hLAT1 at relatively high level, with the two most potent inhibitors led to cell death which was not observed after treatment with a compound exhibiting very poor inhibitory effect.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Discovery/methods , Large Neutral Amino Acid-Transporter 1/metabolism , Thiazines/chemistry , Thiazoles/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Large Neutral Amino Acid-Transporter 1/genetics , Molecular Docking Simulation , Molecular Structure
6.
Bioorg Med Chem ; 25(9): 2625-2634, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28342691

ABSTRACT

DNA intercalating agents are a consolidated therapeutic option in the treatment of tumor diseases. Starting from previous findings in the antiproliferative efficacy of a series of indeno[1,2-c]cinnoline-11-one derivatives, we performed a suitable decoration of this scaffold by means of a simple and straightforward chemistry, aiming to a) enlarge the planar core to a pentacyclic benzo[h]indeno[1,2-c]cinnoline-13-one and b) introduce a basic head tethered through a simple polymethylene chain. In fluorescence melting and fluorescence intercalator displacement assays, these new compounds displayed fair to very good intercalating properties on different nucleic acid strands, with preference for G-quadruplex sequences. Inhibition of human topoisomerase IIα and antiproliferative assays on HeLa and MCF7 tumor cell lines outlined a multitarget antiproliferative profile for tetracyclic 6 and pentacyclic derivative 20, both bearing a N,N-dimethylamine as the protonatable moiety. Particularly, compound 6 displayed a very potent inhibition of tumor cell proliferation, while 20 returned the highest thermal stabilization in melting experiments. In summary, these results outlined a potential of such highly planar scaffolds for nucleic acid binding and antiproliferative effects.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Topoisomerases, Type II/metabolism , G-Quadruplexes , Heterocyclic Compounds, 4 or More Rings/pharmacology , Intercalating Agents/pharmacology , Topoisomerase II Inhibitors/pharmacology , Antineoplastic Agents/chemical synthesis , Benzothiazoles/chemistry , DNA Topoisomerase IV/antagonists & inhibitors , HeLa Cells , Heterocyclic Compounds, 4 or More Rings/chemical synthesis , Humans , Intercalating Agents/chemical synthesis , Ligands , MCF-7 Cells , Quinolines/chemistry , Topoisomerase II Inhibitors/chemical synthesis
7.
Bioorg Med Chem Lett ; 27(5): 1179-1185, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28189420

ABSTRACT

A series of 3,4-dihydroquinazoline derivatives consisting of the selected compounds from our chemical library on the diversity basis and the new synthetic compounds were in vitro tested for their inhibitory activities for both acetylcholinesterase (AChE, from electric eel) and butyrylcholinesterase (BChE, from equine serum) enzymes. It was discovered that most of the compounds displayed weak AChE and strong BuChE inhibitory activities. In particular, compound 8b and 8d were the most active compounds in the series against BChE with IC50 values of 45nM and 62nM, as well as 146- and 161-fold higher affinity to BChE, respectively. To understand the excellent activity of these compounds, molecular docking simulations were performed to get better insights into the mechanism of binding of 3,4-dihydroquinazoline derivatives. As expected, compound 8b and 8d bind to both catalytic anionic site (CAS) and peripheral site (PS) of BChE with better interaction energy values than AChE, in agreement with our experimental data. Furthermore, the non-competitive/mixed-type inhibitions of both compounds further confirmed their dual binding nature in kinetic studies.


Subject(s)
Cholinesterase Inhibitors/pharmacology , Quinazolines/pharmacology , Alzheimer Disease/drug therapy , Cholinesterase Inhibitors/therapeutic use , Humans , Molecular Docking Simulation , Quinazolines/therapeutic use
8.
Open Med Chem J ; 11: 196-211, 2017.
Article in English | MEDLINE | ID: mdl-29387274

ABSTRACT

INTRODUCTION: The 1,3-dipolar cycloaddition reactions of nitrile oxides formed in situ (in the presence of NCS and Et3N) from the oximes of (purin-9-yl)acetaldehyde or (coumarinyloxy)acetaldehyde with allyloxycoumarins or 9-allylpurines, respectively resulted in 3,5-disubstituted isoxazolines. The similar reactions of propargyloxycoumarins or 9-propargylpurines led to 3,5-disubstituted isoxazoles by treatment with PIDA and catalytic amount of TFA. METHODS: The new compounds were tested in vitro as antioxidant agents and inhibitors of soybean lipoxygenase LO, AChE and MAO-B. RESULTS: The majority of the compounds showed significant hydroxyl radical scavenging activity. Compounds 4k and 4n presented LO inhibitory activity. CONCLUSION: Compound 13e presents an antioxidant significant profile combining anti-LO, anti-AChE and anti-MAO-B activities.

9.
Eur J Med Chem ; 123: 704-717, 2016 Nov 10.
Article in English | MEDLINE | ID: mdl-27521587

ABSTRACT

The quinoline motif fused with other heterocyclic systems plays an important role in the field of anticancer drug development. An extensive series of tetracyclic quinolino[3,4-b]quinoxalines N-5 or C-6 substituted with basic side chain and a limited number of tricyclic pyridazino[4,3-c]quinolines N-6 substituted were designed, synthesized and evaluated for topoisomerase IIα (Topo IIα) inhibitory activity, ability to bind and stabilize G-quadruplex structures and cytotoxic properties against two human cancer cell lines (HeLa and MCF-7). Almost all of the tested agents showed a high activity as Topo IIα inhibitors and G-quadruplex stabilizers. Among all the derivatives studied, the quinolino[3,4-b]quinoxalines 11 and 23, N-5 and C-6 substituted respectively, stand out as the most promising compounds. Derivative 11 resulted a selective binder to selected G-quadruplex sequences, while derivative 23 displayed the most interesting Topo IIα inhibitory activity (IC50 = 5.14 µM); both showed high cytotoxic activity (IC50 HeLa = 2.04 µM and 2.32 µM, respectively).


Subject(s)
Antigens, Neoplasm/metabolism , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/metabolism , G-Quadruplexes/drug effects , Quinolines/chemical synthesis , Quinolines/pharmacology , Quinoxalines/chemical synthesis , Quinoxalines/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Chemistry Techniques, Synthetic , DNA Topoisomerase IV/antagonists & inhibitors , HeLa Cells , Humans , Quinolines/chemistry , Quinolines/metabolism , Quinoxalines/chemistry , Quinoxalines/metabolism , Structure-Activity Relationship , Topoisomerase II Inhibitors/chemical synthesis , Topoisomerase II Inhibitors/chemistry , Topoisomerase II Inhibitors/metabolism , Topoisomerase II Inhibitors/pharmacology
10.
Mol Inform ; 35(8-9): 294-308, 2016 09.
Article in English | MEDLINE | ID: mdl-27546034

ABSTRACT

Computational methods have advanced toxicology towards the development of target-specific models based on a clear cause-effect rationale. However, the predictive potential of these models presents strengths and weaknesses. On the good side, in silico models are valuable cheap alternatives to in vitro and in vivo experiments. On the other, the unconscious use of in silico methods can mislead end-users with elusive results. The focus of this review is on the basic scientific and regulatory recommendations in the derivation and application of computational models. Attention is paid to examine the interplay between computational toxicology and drug discovery and development. Avoiding the easy temptation of an overoptimistic future, we report our view on what can, or cannot, realistically be done. Indeed, studies of safety/toxicity represent a key element of chemical prioritization programs carried out by chemical industries, and primarily by pharmaceutical companies.


Subject(s)
Computational Biology/methods , Drug Discovery/methods , Animals , Computer Simulation , Drug-Related Side Effects and Adverse Reactions/etiology , Humans , Toxicology/methods
11.
Methods Mol Biol ; 1425: 461-73, 2016.
Article in English | MEDLINE | ID: mdl-27311477

ABSTRACT

Predictive toxicology is a new emerging multifaceted research field aimed at protecting human health and environment from risks posed by chemicals. Such issue is of extreme public relevance and requires a multidisciplinary approach where the experience in medicinal chemistry is of utmost importance. Herein, we will survey some basic recommendations to gather good data and then will review three recent case studies to show how strategies of ligand- and structure-based molecular design, widely applied in medicinal chemistry, can be adapted to meet the more restrictive scientific and regulatory goals of predictive toxicology. In particular, we will report: Docking-based classification models to predict the estrogenic potentials of chemicals. Predicting the bioconcentration factor using biokinetics descriptors. Modeling oral sub-chronic toxicity using a customized k-nearest neighbors (k-NN) approach.


Subject(s)
Molecular Docking Simulation/methods , Toxicity Tests, Subchronic/methods , Algorithms , Computer Simulation , Drug Design , Humans , Models, Biological , Models, Theoretical , Quantitative Structure-Activity Relationship
12.
J Med Chem ; 59(14): 6791-806, 2016 07 28.
Article in English | MEDLINE | ID: mdl-27347731

ABSTRACT

Aiming at modulating two key enzymatic targets for Alzheimer's disease (AD), i.e., acetylcholinesterase (AChE) and monoamine oxidase B (MAO B), a series of multitarget ligands was properly designed by linking the 3,4-dimethylcoumarin scaffold to 1,3- and 1,4-substituted piperidine moieties, thus modulating the basicity to improve the hydrophilic/lipophilic balance. After in vitro enzymatic inhibition assays, multipotent inhibitors showing potencies in the nanomolar and in the low micromolar range for hMAO B and eeAChE, respectively, were prioritized and evaluated in human SH-SY5Y cell-based models for their cytotoxicity and neuroprotective effect against oxidative toxins (H2O2, rotenone, and oligomycin-A). The present study led to the identification of a promising multitarget hit compound (5b) exhibiting high hMAO B inhibitory activity (IC50 = 30 nM) and good MAO B/A selectivity (selectivity index, SI = 94) along with a micromolar eeAChE inhibition (IC50 = 1.03 µM). Moreover, 5b behaves as a water-soluble, brain-permeant neuroprotective agent against oxidative insults without interacting with P-gp efflux system.


Subject(s)
Acetylcholinesterase/metabolism , Cholinesterase Inhibitors/pharmacology , Coumarins/pharmacology , Monoamine Oxidase Inhibitors/pharmacology , Monoamine Oxidase/metabolism , Neuroprotective Agents/pharmacology , Animals , Cell Line, Tumor , Cell Survival/drug effects , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/chemistry , Coumarins/chemical synthesis , Coumarins/chemistry , Dogs , Dose-Response Relationship, Drug , Humans , Madin Darby Canine Kidney Cells , Molecular Structure , Monoamine Oxidase Inhibitors/chemical synthesis , Monoamine Oxidase Inhibitors/chemistry , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/chemistry , Oxidative Stress/drug effects , Solubility , Structure-Activity Relationship , Water/chemistry
13.
Molecules ; 21(3): 362, 2016 Mar 17.
Article in English | MEDLINE | ID: mdl-26999091

ABSTRACT

The need for developing real disease-modifying drugs against neurodegenerative syndromes, particularly Alzheimer's disease (AD), shifted research towards reliable drug discovery strategies to unveil clinical candidates with higher therapeutic efficacy than single-targeting drugs. By following the multi-target approach, we designed and synthesized a novel class of dual acetylcholinesterase (AChE)-monoamine oxidase B (MAO-B) inhibitors through the decoration of the 2H-chromen-2-one skeleton. Compounds bearing a propargylamine moiety at position 3 displayed the highest in vitro inhibitory activities against MAO-B. Within this series, derivative 3h emerged as the most interesting hit compound, being a moderate AChE inhibitor (IC50 = 8.99 µM) and a potent and selective MAO-B inhibitor (IC50 = 2.8 nM). Preliminary studies in human neuroblastoma SH-SY5Y cell lines demonstrated its low cytotoxicity and disclosed a promising neuroprotective effect at low doses (0.1 µM) under oxidative stress conditions promoted by two mitochondrial toxins (oligomycin-A and rotenone). In a Madin-Darby canine kidney (MDCK)II-MDR1 cell-based transport study, Compound 3h was able to permeate the BBB-mimicking monolayer and did not result in a glycoprotein-p (P-gp) substrate, showing an efflux ratio = 0.96, close to that of diazepam.


Subject(s)
Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/pharmacology , Drug Discovery , Monoamine Oxidase Inhibitors/chemistry , Monoamine Oxidase Inhibitors/pharmacology , Alzheimer Disease/drug therapy , Animals , Cell Line , Cholinesterase Inhibitors/chemical synthesis , Enzyme Activation/drug effects , Humans , Molecular Structure , Monoamine Oxidase Inhibitors/chemical synthesis , Structure-Activity Relationship
14.
Bioorg Med Chem Lett ; 26(1): 21-4, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26615885

ABSTRACT

Four different classes of new 17ß-hydroxysteroid dehydrogenase type 2 (17ß-HSD2) inhibitors were synthesized, in order to lower the cytotoxicity exhibited by the lead compound A, via disrupting the linearity and the aromaticity of the biphenyl moiety. Compounds 3, 4, 7a and 8 displayed comparable or better inhibitory activity and selectivity, as well as a lower cytotoxic effect, compared to the reference compound A. The best compound 4 (IC50=160nM, selectivity factor=168, LD50≈25µM) turned out as new lead compound for inhibition of 17ß-HSD2.


Subject(s)
Amides/pharmacology , Biphenyl Compounds/pharmacology , Cytotoxins/pharmacology , Drug Discovery , Enzyme Inhibitors/pharmacology , Estradiol Dehydrogenases/antagonists & inhibitors , Amides/chemical synthesis , Amides/chemistry , Biphenyl Compounds/chemical synthesis , Biphenyl Compounds/chemistry , Cell Survival/drug effects , Cytotoxins/chemical synthesis , Cytotoxins/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Estradiol Dehydrogenases/metabolism , HEK293 Cells , Humans , Molecular Structure , Structure-Activity Relationship
15.
Future Med Chem ; 7(14): 1921-36, 2015.
Article in English | MEDLINE | ID: mdl-26440057

ABSTRACT

BACKGROUND: The ethical and practical limitation of animal testing has recently promoted computational methods for the fast screening of huge collections of chemicals. RESULTS: The authors derived 24 reliable docking-based classification models able to predict the estrogenic potential of a large collection of chemicals provided by the US Environmental Protection Agency. Model performances were challenged by considering AUC, EF1% (EFmax = 7.1), -LR (at sensitivity = 0.75); +LR (at sensitivity = 0.25) and 37 reference compounds comprised within the training set. Moreover, external predictions were made successfully on ten representative known estrogenic chemicals and on a set consisting of >32,000 chemicals. CONCLUSION: The authors demonstrate that structure-based methods, widely applied to drug discovery programs, can be fairly adapted to exploratory toxicology studies.


Subject(s)
Molecular Docking Simulation , Receptors, Estrogen/chemistry , Animals , Area Under Curve , Binding Sites , Drug Discovery , Estrogen Antagonists/classification , Estrogen Antagonists/toxicity , Estrogen Receptor Modulators/classification , Estrogen Receptor Modulators/toxicity , Quantitative Structure-Activity Relationship , ROC Curve , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/metabolism , Software
16.
PLoS One ; 10(7): e0134754, 2015.
Article in English | MEDLINE | ID: mdl-26230928

ABSTRACT

Design and synthesis of a new class of inhibitors for the treatment of osteoporosis and its comparative h17ß-HSD2 and m17ß-HSD2 SAR study are described. 17a is the first compound to show strong inhibition of both h17ß-HSD2 and m17ß-HSD2, intracellular activity, metabolic stability, selectivity toward h17ß-HSD1, m17ß-HSD1 and estrogen receptors α and ß as well as appropriate physicochemical properties for oral bioavailability. These properties make it eligible for pre-clinical animal studies, prior to human studies.


Subject(s)
Enzyme Inhibitors/pharmacology , Estradiol Dehydrogenases/antagonists & inhibitors , Animals , Cell Line, Tumor , Enzyme Inhibitors/chemistry , Humans , Mice , Structure-Activity Relationship
17.
J Med Chem ; 58(14): 5561-78, 2015 Jul 23.
Article in English | MEDLINE | ID: mdl-26107513

ABSTRACT

The multifactorial nature of Alzheimer's disease calls for the development of multitarget agents addressing key pathogenic processes. To this end, by following a docking-assisted hybridization strategy, a number of aminocoumarins were designed, prepared, and tested as monoamine oxidases (MAOs) and acetyl- and butyryl-cholinesterase (AChE and BChE) inhibitors. Highly flexible N-benzyl-N-alkyloxy coumarins 2-12 showed good inhibitory activities at MAO-B, AChE, and BChE but low selectivity. More rigid inhibitors, bearing meta- and para-xylyl linkers, displayed good inhibitory activities and high MAO-B selectivity. Compounds 21, 24, 37, and 39, the last two featuring an improved hydrophilic/lipophilic balance, exhibited excellent activity profiles with nanomolar inhibitory potency toward hMAO-B, high hMAO-B over hMAO-A selectivity and submicromolar potency at hAChE. Cell-based assays of BBB permeation, neurotoxicity, and neuroprotection supported the potential of compound 37 as a BBB-permeant neuroprotective agent against H2O2-induced oxidative stress with poor interaction as P-gp substrate and very low cytotoxicity.


Subject(s)
Cholinesterases/metabolism , Coumarins/chemistry , Coumarins/pharmacology , Drug Design , Monoamine Oxidase/metabolism , Animals , Blood-Brain Barrier/metabolism , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/metabolism , Cholinesterase Inhibitors/pharmacology , Cholinesterase Inhibitors/toxicity , Cholinesterases/chemistry , Coumarins/metabolism , Coumarins/toxicity , Dogs , Humans , Inhibitory Concentration 50 , Madin Darby Canine Kidney Cells , Molecular Docking Simulation , Monoamine Oxidase/chemistry , Monoamine Oxidase Inhibitors/chemistry , Monoamine Oxidase Inhibitors/metabolism , Monoamine Oxidase Inhibitors/pharmacology , Monoamine Oxidase Inhibitors/toxicity , Permeability , Protein Conformation , Rats , Structure-Activity Relationship
18.
J Pharm Pharmacol ; 67(10): 1380-92, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26078032

ABSTRACT

OBJECTIVES: The aim of this work is to investigate whether and how two newly synthesized 3,4,5-trimethoxygalloyl-containing compounds 1 and 3 interfere with the mitogen-activated protein kinase (MAPK) signalling pathways involved in several pathological events, ranging from inflammatory diseases to cancer. METHODS: The effects on the phosphorylation of MAP kinases (c-Jun N-terminal kinases (JNKs), p38) and activation of nuclear factor-kappa B (NF-κB) pathways of 1 and its 1H-indazole-containing analogue 3, compared with those elicited by the known Adenosine Triphosphate (ATP)-competitive JNK inhibitor SP600125, were evaluated through Western blot analysis in murine fibroblasts NIH-3T3 and human endothelial cells EA.hy926 acutely treated with tumour necrosis factor-α (TNF-α). Their effects on cell viability were also assessed by 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) assay. KEY FINDINGS: In cultured murine fibroblasts, 1 inhibited JNK signalling with a different mechanism from SP600125. It reduced c-Jun phosphorylation without altering phosphorylation levels of JNK protein. Compound 3, showing a profile similar to SP600125, inhibited JNK phosphorylation and partially inhibited p38 MAPK at 50 µm concentration. Compound 3 and SP600125 showed similar behaviour in both cell cultures. In contrast, compound 1 in EA.hy926 cells significantly interfered with JNK phosphorylation, did not decrease phosphorylation of c-Jun (Ser73), whereas significantly suppressed phosphorylation of p38 MAPK and reversed degradation of NF-κB signalling components. CONCLUSIONS: 3,4,5-Trimethoxygalloyl-based compounds 1 and 3, which did not show significant cell toxicity, modulate the TNF-α-induced activation of MAPK signalling, mainly inhibiting phosphorylation of JNK, c-Jun and p38 MAPK, in murine fibroblasts and human endothelial cells with different MAPK selectivity profiles. These compounds deserve future investigation in specific cell-based disease models and in-vivo pharmacology.


Subject(s)
Benzamides/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Benzamides/chemistry , Benzamides/toxicity , Cell Survival/drug effects , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , MAP Kinase Signaling System/drug effects , Mice , Mitogen-Activated Protein Kinases/metabolism , NIH 3T3 Cells , Tumor Necrosis Factor-alpha/administration & dosage
19.
ChemMedChem ; 10(6): 1040-53, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25924599

ABSTRACT

Multitarget therapeutic leads for Alzheimer's disease were designed on the models of compounds capable of maintaining or restoring cell protein homeostasis and of inhibiting ß-amyloid (Aß) oligomerization. Thirty-seven thioxanthen-9-one, xanthen-9-one, naphto- and anthraquinone derivatives were tested for the direct inhibition of Aß(1-40) aggregation and for the inhibition of electric eel acetylcholinesterase (eeAChE) and horse serum butyrylcholinesterase (hsBChE). These compounds are characterized by basic side chains, mainly quinolizidinylalkyl moieties, linked to various bi- and tri-cyclic (hetero)aromatic systems. With very few exceptions, these compounds displayed inhibitory activity on both AChE and BChE and on the spontaneous aggregation of ß-amyloid. In most cases, IC50 values were in the low micromolar and sub-micromolar range, but some compounds even reached nanomolar potency. The time course of amyloid aggregation in the presence of the most active derivative (IC50 =0.84 µM) revealed that these compounds might act as destabilizers of mature fibrils rather than mere inhibitors of fibrillization. Many compounds inhibited one or both cholinesterases and Aß aggregation with similar potency, a fundamental requisite for the possible development of therapeutics exhibiting a multitarget mechanism of action. The described compounds thus represent interesting leads for the development of multitarget AD therapeutics.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/antagonists & inhibitors , Cholinesterase Inhibitors/therapeutic use , Quinolizidines/chemistry , Amyloid beta-Peptides/chemistry , Blood-Brain Barrier , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/pharmacokinetics , Circular Dichroism , Humans , Kinetics , Microscopy, Electron, Transmission , Structure-Activity Relationship
20.
ChemMedChem ; 10(6): 1054-70, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25924828

ABSTRACT

By following a multitarget ligand design approach, a library of 47 compounds was prepared, and they were tested as binders of selected G protein-coupled receptors (GPCRs) and inhibitors of acetyl and/or butyryl cholinesterase. The newly designed ligands feature pyridazinone-based tricyclic scaffolds connected through alkyl chains of variable length to proper amine moieties (e.g., substituted piperazines or piperidines) for GPCR and cholinesterase (ChE) molecular recognition. The compounds were tested at three different GPCRs, namely serotoninergic 5-HT1A, adrenergic α1A, and dopaminergic D2 receptors. Our main goal was the discovery of compounds that exhibit, in addition to ChE inhibition, antagonist activity at 5-HT1A because of its involvement in neuronal deficits typical of Alzheimer's and other neurodegenerative diseases. Ligands with nanomolar affinity for the tested GPCRs were discovered, but most of them behaved as dual antagonists of α1A and 5-HT1A receptors. Nevertheless, several compounds displaying this GPCR affinity profile also showed moderate to good inhibition of AChE and BChE, thus deserving further investigations to exploit the therapeutic potential of such unusual biological profiles.


Subject(s)
Cholinesterase Inhibitors/metabolism , Pyridazines/metabolism , Receptors, G-Protein-Coupled/metabolism , Acetylcholinesterase/drug effects , Animals , Butyrylcholinesterase/drug effects , Dogs , Humans , Ligands , Madin Darby Canine Kidney Cells
SELECTION OF CITATIONS
SEARCH DETAIL