Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Molecules ; 29(11)2024 Jun 02.
Article in English | MEDLINE | ID: mdl-38893493

ABSTRACT

GSK-3ß, IKK-ß, and ROCK-1 kinases are implicated in the pathomechanism of Alzheimer's disease due to their involvement in the misfolding and accumulation of amyloid ß (Aß) and tau proteins, as well as inflammatory processes. Among these kinases, GSK-3ß plays the most crucial role. In this study, we present compound 62, a novel, remarkably potent, competitive GSK-3ß inhibitor (IC50 = 8 nM, Ki = 2 nM) that also exhibits additional ROCK-1 inhibitory activity (IC50 = 2.3 µM) and demonstrates anti-inflammatory and neuroprotective properties. Compound 62 effectively suppresses the production of nitric oxide (NO) and pro-inflammatory cytokines in the lipopolysaccharide-induced model of inflammation in the microglial BV-2 cell line. Furthermore, it shows neuroprotective effects in an okadaic-acid-induced tau hyperphosphorylation cell model of neurodegeneration. The compound also demonstrates the potential for further development, characterized by its chemical and metabolic stability in mouse microsomes and fair solubility.


Subject(s)
Alzheimer Disease , Glycogen Synthase Kinase 3 beta , I-kappa B Kinase , Thiazoles , rho-Associated Kinases , tau Proteins , tau Proteins/metabolism , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Animals , Thiazoles/pharmacology , Thiazoles/chemistry , Humans , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism , Mice , I-kappa B Kinase/metabolism , I-kappa B Kinase/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Neuroprotective Agents/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemistry , Cell Line , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Microglia/drug effects , Microglia/metabolism , Nitric Oxide/metabolism , Lipopolysaccharides , Protein Aggregates/drug effects , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism
2.
Future Med Chem ; 16(3): 197-204, 2024 02.
Article in English | MEDLINE | ID: mdl-38189171

ABSTRACT

Aims: Our research aimed to evaluate how the rigidification of the characteristic 3-aminopropyloxy linker by incorporating it into 1,5-benzoxazepines affects the potency of histamine H3 receptor (H3R) antagonists/inverse agonists. This research constitutes a starting point for the full characterization of the pharmacological properties of this group of compounds. Materials & methods: Several 1,5-benzoxazepine derivatives were synthesized and pharmacologically tested as potential H3R antagonist/inverse agonists. In a addition, the effect of the derivatives on acetylcholinesterase and butyrylcholinesterase inhibition and cytotoxicity were tested. Results: The studies indicated 1,5-benzoxazepine containing three carbon side chains as a compound for further modification. Conclusion: Further optimization of the lead structure is necessary, which will favorably affect biological targets.


Subject(s)
Histamine , Receptors, Histamine H3 , Butyrylcholinesterase/metabolism , Acetylcholinesterase/metabolism , Receptors, Histamine H3/chemistry , Drug Inverse Agonism , Structure-Activity Relationship
3.
Pharmaceuticals (Basel) ; 16(5)2023 Apr 30.
Article in English | MEDLINE | ID: mdl-37242458

ABSTRACT

This study examines the properties of novel guanidines, designed and synthesized as histamine H3R antagonists/inverse agonists with additional pharmacological targets. We evaluated their potential against two targets viz., inhibition of MDA-MB-231, and MCF-7 breast cancer cells viability and inhibition of AChE/BuChE. ADS10310 showed micromolar cytotoxicity against breast cancer cells, combined with nanomolar affinity at hH3R, and may represent a promising target for the development of an alternative method of cancer therapy. Some of the newly synthesized compounds showed moderate inhibition of BuChE in the single-digit micromolar concentration ranges. H3R antagonist with additional AChE/BuChE inhibitory effect might improve cognitive functions in Alzheimer's disease. For ADS10310, several in vitro ADME-Tox parameters were evaluated and indicated that it is a metabolically stable compound with weak hepatotoxic activity and can be accepted for further studies.

4.
Int J Mol Sci ; 23(16)2022 Aug 21.
Article in English | MEDLINE | ID: mdl-36012707

ABSTRACT

Neurodegeneration leading to Alzheimer's disease results from a complex interplay of a variety of processes including misfolding and aggregation of amyloid beta and tau proteins, neuroinflammation or oxidative stress. Therefore, to address more than one of these, drug discovery programmes focus on the development of multifunctional ligands, preferably with disease-modifying and symptoms-reducing potential. Following this idea, herein we present the design and synthesis of multifunctional ligands and biological evaluation of their 5-HT6 receptor affinity (radioligand binding assay), cholinesterase inhibitory activity (spectroscopic Ellman's assay), antioxidant activity (ABTS assay) and metal-chelating properties, as well as a preliminary ADMET properties evaluation. Based on the results we selected compound 14 as a well-balanced and potent 5-HT6 receptor ligand (Ki = 22 nM) and human BuChE inhibitor (IC50 = 16 nM) with antioxidant potential expressed as a reduction of ABTS radicals by 35% (150 µM). The study also revealed additional metal-chelating properties of compounds 15 and 18. The presented compounds modulating Alzheimer's disease-related processes might be further developed as multifunctional ligands against the disease.


Subject(s)
Alzheimer Disease , Cholinesterase Inhibitors , Acetylcholinesterase/metabolism , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Antioxidants/chemistry , Antioxidants/pharmacology , Butyrylcholinesterase/metabolism , Chelating Agents/chemistry , Chelating Agents/pharmacology , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/pharmacology , Drug Design , Humans , Ligands , Receptors, Serotonin/metabolism , Serotonin , Structure-Activity Relationship
5.
Eur J Med Chem ; 238: 114490, 2022 Aug 05.
Article in English | MEDLINE | ID: mdl-35660251

ABSTRACT

The ever-increasing number of bacteria resistant to the currently available antibacterial agents is a great medical problem today, and new antibiotics with novel mechanisms of action are urgently needed. Among the validated antibacterial drug targets against which new classes of antibiotics might be directed is bacterial type I signal peptidase (SPase I), an essential part of the Tat and Sec secretory systems. SPase I is responsible for the hydrolysis of the N-terminal signal peptides from proteins secreted across the cytoplasmic membrane and plays a key role in bacterial viability and virulence. This review focuses on the antibacterial activity of natural and synthetic SPase I inhibitors reported to date, namely ß-lactams, lipopeptides, and arylomycins, but also an example of SPase I activator was presented.


Subject(s)
Membrane Proteins , Serine Endopeptidases , Anti-Bacterial Agents/pharmacology , Bacteria/metabolism , Bacterial Proteins/metabolism , Serine Endopeptidases/metabolism
6.
ACS Chem Neurosci ; 12(11): 2057-2068, 2021 06 02.
Article in English | MEDLINE | ID: mdl-34019757

ABSTRACT

In Alzheimer's disease, neurons slowly degenerate due to the accumulation of misfolded amyloid ß and tau proteins. In our research, we performed extended studies directed at amyloid ß and tau aggregation inhibition using in cellulo (Escherichia coli model of protein aggregation), in silico, and in vitro kinetic studies. We tested our library of 1-benzylamino-2-hydroxyalkyl multifunctional anti-Alzheimer's agents and identified very potent dual aggregation inhibitors. Among the tested derivatives, we selected compound 18, which exhibited a unique profile of biological activity. This compound was the most potent and balanced dual aggregation inhibitor (Aß42 inhibition (inh.) 80.0%, tau inh. 68.3% in 10 µM), with previously reported in vitro inhibitory activity against hBuChE, hBACE1, and Aß (hBuChE IC50 = 5.74 µM; hBACE1 IC50 = 41.6 µM; Aß aggregation (aggr.) inh. IC50 = 3.09 µM). In docking studies for both proteins, we tried to explain the different structural requirements for the inhibition of Aß vs tau. Moreover, docking and kinetic studies showed that compound 18 could inhibit the amyloid aggregation process at several steps and also displayed disaggregating properties. These results may help to design the next generations of dual or selective aggregation inhibitors.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Alzheimer Disease/drug therapy , Cholinesterase Inhibitors/pharmacology , Computer Simulation , Drug Design , Humans , Kinetics , Peptide Fragments , Structure-Activity Relationship
7.
Eur J Med Chem ; 218: 113397, 2021 Jun 05.
Article in English | MEDLINE | ID: mdl-33838585

ABSTRACT

Looking for an effective anti-Alzheimer's agent is very challenging; however, a multifunctional ligand strategy may be a promising solution for the treatment of this complex disease. We herein present the design, synthesis and biological evaluation of novel hydroxyethylamine derivatives displaying unique, multiple properties that have not been previously reported. The original mechanism of action combines inhibitory activity against disease-modifying targets: ß-secretase enzyme (BACE1) and amyloid ß (Aß) aggregation, along with an effect on targets associated with symptom relief - inhibition of butyrylcholinesterase (BuChE) and γ-aminobutyric acid transporters (GATs). Among the obtained molecules, compound 36 exhibited the most balanced and broad activity profile (eeAChE IC50 = 2.86 µM; eqBuChE IC50 = 60 nM; hBuChE IC50 = 20 nM; hBACE1 IC50 = 5.9 µM; inhibition of Aß aggregation = 57.9% at 10 µM; mGAT1 IC50 = 10.96 µM; and mGAT2 IC50 = 19.05 µM). Moreover, we also identified 31 as the most potent mGAT4 and hGAT3 inhibitor (IC50 = 5.01 µM and IC50 = 2.95 µM, respectively), with high selectivity over other subtypes. Compounds 36 and 31 represent new anti-Alzheimer agents that can ameliorate cognitive decline and modify the progress of disease.


Subject(s)
Alzheimer Disease/drug therapy , Butyrylcholinesterase/metabolism , Cholinesterase Inhibitors/pharmacology , Drug Discovery , GABA Plasma Membrane Transport Proteins/metabolism , Neuroprotective Agents/pharmacology , Alzheimer Disease/metabolism , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/chemistry , Dose-Response Relationship, Drug , Humans , Molecular Structure , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/chemistry , Structure-Activity Relationship
8.
ACS Chem Neurosci ; 11(22): 3793-3801, 2020 11 18.
Article in English | MEDLINE | ID: mdl-33143412

ABSTRACT

Herein we report in vitro metabolic stability in human liver microsomes (HLMs), interactions with cytochrome P450 isoenzymes (CYP3A4, CYP2D6, and CYP2C9), and cytotoxicity analyses on HEK-293, HepG2, Huh7, and WTIIB cell lines of our most recent multitarget directed ligands PF9601N, ASS234, and contilisant. Based on these results, we conclude that (1) PF9601N and contilisant are metabolically stable in the HLM assay, in contrast to the very unstable ASS234; (2) CYP3A4 activity was decreased by PF9601N at all the tested concentrations and by ASS234 and contilisant only at the highest concentration; CYP2D6 activity was reduced by ASS234 at 1, 10, and 25 µM and by PF9601N at 10 and 25 µM, whereas contilisant increased its activity at the same concentrations; CYP2C9 was inhibited by the three compounds; (3) contilisant did not affect cell viability in the widest range of concentrations: up to 10 µM on HEK-293 cells, up to 30 µM on Huh7 cells, up to 50 µM on HepG2 cells, and up to 30 or 100 µM on WTIIB cells. Based on these results, we selected contilisant as a metabolically stable and nontoxic lead compound for further studies in Alzheimer's disease therapy.


Subject(s)
Monoamine Oxidase Inhibitors , Neurodegenerative Diseases , Computer Simulation , HEK293 Cells , Humans , Microsomes, Liver , Monoamine Oxidase Inhibitors/pharmacology
9.
J Enzyme Inhib Med Chem ; 35(1): 1944-1952, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33092411

ABSTRACT

Effective therapy of Alzheimer's disease (AD) requires treatment with a combination of drugs that modulate various pathomechanisms contributing to the disease. In our research, we have focused on the development of multi-target-directed ligands - 5-HT6 receptor antagonists and cholinesterase inhibitors - with disease-modifying properties. We have performed extended in vitro (FRET assay) and in cellulo (Escherichia coli model of protein aggregation) studies on their ß-secretase, tau, and amyloid ß aggregation inhibitory activity. Within these multifunctional ligands, we have identified compound 17 with inhibitory potency against tau and amyloid ß aggregation in in cellulo assay of 59% and 56% at 10 µM, respectively, hBACE IC50=4 µM, h5TH6 K i=94 nM, hAChE IC50=26 nM, and eqBuChE IC50=5 nM. This study led to the development of multifunctional ligands with a broad range of biological activities crucial not only for the symptomatic but also for the disease-modifying treatment of AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Cholinesterase Inhibitors/chemistry , Cholinesterases/metabolism , Receptors, Serotonin/metabolism , tau Proteins/metabolism , Alzheimer Disease/prevention & control , Cholinesterase Inhibitors/metabolism , Drug Design , Escherichia coli , Fluorescence Resonance Energy Transfer , Humans , Ligands , Models, Molecular , Protein Aggregates , Structure-Activity Relationship
10.
Bioorg Chem ; 90: 103084, 2019 09.
Article in English | MEDLINE | ID: mdl-31271942

ABSTRACT

In the search for new treatments for complex disorders such as Alzheimer's disease the Multi-Target-Directed Ligands represent a very promising approach. The aim of the present study was to identify multifunctional compounds among several series of non-imidazole histamine H3 receptor ligands, derivatives of 1-[2-thiazol-5-yl-(2-aminoethyl)]-4-n-propylpiperazine, 1-[2-thiazol-4-yl-(2-aminoethyl)]-4-n-propylpiperazine and 1-phenoxyalkyl-4-(amino)alkylopiperazine using in vitro and in vivo pharmacological evaluation and computational studies. Performed in vitro assays showed moderate potency of tested compounds against acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE). Molecular modeling studies have revealed possible interactions between the active compounds and both AChE and BuChE as well as the human H3 histamine receptor. Computational studies showed the high drug-likeness of selected compounds with very good physicochemical profiles. The parallel artificial membrane permeation assay proved outstanding blood-brain barrier penetration in test conditions. The most promising compound, A12, chemically methyl(4-phenylbutyl){2-[2-(4-propylpiperazin-1-yl)-1,3-thiazol-5-yl]ethyl}amine, possesses good balanced multifunctional profile with potency toward studied targets - H3 antagonist activity (pA2 = 8.27), inhibitory activity against both AChE (IC50 = 13.96 µM), and BuChE (IC50 = 14.62 µM). The in vivo pharmacological studies revealed the anti-amnestic properties of compound A12 in the passive avoidance test on mice.


Subject(s)
Alzheimer Disease/drug therapy , Amnesia/drug therapy , Cholinesterase Inhibitors/pharmacology , Disease Models, Animal , Piperazines/chemistry , Receptors, Histamine H3/metabolism , Acetylcholinesterase/chemistry , Adjuvants, Anesthesia/toxicity , Amnesia/chemically induced , Animals , Butyrylcholinesterase/chemistry , Cholinesterase Inhibitors/chemistry , Computational Biology , In Vitro Techniques , Ligands , Male , Mice , Models, Molecular , Molecular Structure , Receptors, Histamine H3/chemistry , Scopolamine/toxicity , Structure-Activity Relationship
11.
Eur J Med Chem ; 157: 1346-1360, 2018 Sep 05.
Article in English | MEDLINE | ID: mdl-30196059

ABSTRACT

Type I signal peptidase, with its vital role in bacterial viability, is a promising but underexploited antibacterial drug target. In the light of steadily increasing rates of antimicrobial resistance, we have developed novel macrocyclic lipopeptides, linking P2 and P1' by a boronic ester warhead, capable of inhibiting Escherichia coli type I signal peptidase (EcLepB) and exhibiting good antibacterial activity. Structural modifications of the macrocyclic ring, the peptide sequence and the lipophilic tail led us to 14 novel macrocyclic boronic esters. It could be shown that macrocyclization is well tolerated in terms of EcLepB inhibition and antibacterial activity. Among the synthesized macrocycles, potent enzyme inhibitors in the low nanomolar range (e.g. compound 42f, EcLepB IC50 = 29 nM) were identified also showing good antimicrobial activity (e.g. compound 42b, E. coli WT MIC = 16 µg/mL). The unique macrocyclic boronic esters described here were based on previously published linear lipopeptidic EcLepB inhibitors in an attempt to address cytotoxicity and hemolysis. We show herein that structural changes to the macrocyclic ring influence both the cytotoxicity and hemolytic activity suggesting that the P2 to P1' linker provide means for optimizing off-target effects. However, for the present set of compounds we were not able to separate the antibacterial activity and cytotoxic effect.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antineoplastic Agents/pharmacology , Escherichia coli/drug effects , Escherichia coli/enzymology , Membrane Proteins/antagonists & inhibitors , Serine Proteinase Inhibitors/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Boronic Acids/chemistry , Boronic Acids/pharmacology , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Esters/chemistry , Esters/pharmacology , Hep G2 Cells , Humans , Lipopeptides/chemistry , Lipopeptides/pharmacology , Macrocyclic Compounds/chemistry , Macrocyclic Compounds/pharmacology , Membrane Proteins/metabolism , Molecular Structure , Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/chemical synthesis , Serine Proteinase Inhibitors/chemistry , Structure-Activity Relationship
12.
Bioorg Med Chem ; 25(3): 897-911, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28038943

ABSTRACT

Type I signal peptidases are potential targets for the development of new antibacterial agents. Here we report finding potent inhibitors of E. coli type I signal peptidase (LepB), by optimizing a previously reported hit compound, decanoyl-PTANA-CHO, through modifications at the N- and C-termini. Good improvements of inhibitory potency were obtained, with IC50s in the low nanomolar range. The best inhibitors also showed good antimicrobial activity, with MICs in the low µg/mL range for several bacterial species. The selection of resistant mutants provided strong support for LepB as the target of these compounds. The cytotoxicity and hemolytic profiles of these compounds are not optimal but the finding that minor structural changes cause the large effects on these properties suggests that there is potential for optimization in future studies.


Subject(s)
Drug Design , Escherichia coli/enzymology , Membrane Proteins/antagonists & inhibitors , Oligopeptides/pharmacology , Cell Survival/drug effects , Dose-Response Relationship, Drug , Hep G2 Cells , Humans , Membrane Proteins/metabolism , Models, Molecular , Molecular Structure , Oligopeptides/chemical synthesis , Oligopeptides/chemistry , Serine Endopeptidases/metabolism , Structure-Activity Relationship
13.
Bioorg Med Chem Lett ; 26(16): 4140-5, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27445168

ABSTRACT

In recent years, multitarget-directed ligands have become an interesting strategy in a search for a new treatment of Alzheimer's disease. Combination of both: a histamine H3 receptor antagonist/inverse agonist and a cholinesterases inhibitor in one molecule could provide a new therapeutic opportunity. Here, we present biological evaluation of histamine H3 receptor ligands-chlorophenoxyalkylamine derivatives against cholinesterases: acetyl- and butyrylcholinesterase. The target compounds showed cholinesterase inhibitory activity in a low micromolar range. The most potent in this group was 1-(7-(4-chlorophenoxy)heptyl)homopiperidine (18) inhibiting the both enzymes (EeAChE IC50=1.93µM and EqBuChE IC50=1.64µM). Molecular modeling studies were performed to explain the binding mode of 18 with histamine H3 receptor as well as with cholinesterases.


Subject(s)
Acetylcholinesterase/metabolism , Amines/chemistry , Butyrylcholinesterase/metabolism , Cholinesterase Inhibitors/chemistry , Piperidines/chemistry , Receptors, Histamine H3/metabolism , Acetylcholinesterase/chemistry , Binding Sites , Butyrylcholinesterase/chemistry , Catalytic Domain , Humans , Kinetics , Ligands , Molecular Docking Simulation , Piperidines/chemical synthesis , Receptors, Histamine H3/chemistry , Structure-Activity Relationship
14.
Bioorg Med Chem ; 24(20): 4835-4854, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27396685

ABSTRACT

Different azides and alkynes have been coupled via Cu-catalyzed 1,3-dipolar Huisgen cycloaddition to afford a novel family of N1- and C5-substituted 1,2,3-triazole derivatives that feature the propargylamine group typical of irreversible MAO-B inhibitors at the C4-side chain of the triazole ring. All the synthesized compounds were evaluated against human MAO-A and MAO-B. Structure-activity relationships and molecular modeling were utilized to gain insight into the structural and chemical features that enhance the binding affinity and selectivity between the two enzyme isoforms. Several lead compounds, in terms of potency (submicromolar to low micromolar range), MAO-B selective recognition, and brain permeability, were identified. One of these leads (MAO-B IC50 of 3.54µM, selectivity MAO-A/MAO-B index of 27.7) was further subjected to reversibility and time-dependence inhibition studies, which disclosed a slow and irreversible inhibition of human MAO-B. Overall, the results support the suitability of the 4-triazolylalkyl propargylamine scaffold for exploring the design of multipotent anti-Alzheimer compounds endowed with irreversible MAO-B inhibitory activity.


Subject(s)
Drug Design , Monoamine Oxidase Inhibitors/pharmacology , Monoamine Oxidase/metabolism , Pargyline/pharmacology , Dose-Response Relationship, Drug , Humans , Molecular Docking Simulation , Molecular Structure , Monoamine Oxidase Inhibitors/chemistry , Pargyline/analogs & derivatives , Pargyline/chemical synthesis , Pargyline/chemistry , Structure-Activity Relationship
15.
Molecules ; 21(4): 410, 2016 Mar 26.
Article in English | MEDLINE | ID: mdl-27023510

ABSTRACT

Cholinesterases and amyloid beta are one of the major biological targets in the search for a new and efficacious treatment of Alzheimer's disease. The study describes synthesis and pharmacological evaluation of new compounds designed as dual binding site acetylcholinesterase inhibitors. Among the synthesized compounds, two deserve special attention--compounds 42 and 13. The former is a saccharin derivative and the most potent and selective acetylcholinesterase inhibitor (EeAChE IC50 = 70 nM). Isoindoline-1,3-dione derivative 13 displays balanced inhibitory potency against acetyl- and butyrylcholinesterase (BuChE) (EeAChE IC50 = 0.76 µM, EqBuChE IC50 = 0.618 µM), and it inhibits amyloid beta aggregation (35.8% at 10 µM). Kinetic studies show that the developed compounds act as mixed or non-competitive acetylcholinesterase inhibitors. According to molecular modelling studies, they are able to interact with both catalytic and peripheral active sites of the acetylcholinesterase. Their ability to cross the blood-brain barrier (BBB) was confirmed in vitro in the parallel artificial membrane permeability BBB assay. These compounds can be used as a solid starting point for further development of novel multifunctional ligands as potential anti-Alzheimer's agents.


Subject(s)
Acetylcholinesterase/therapeutic use , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/therapeutic use , Cholinesterase Inhibitors/therapeutic use , Protein Aggregation, Pathological/drug therapy , Acetylcholinesterase/chemistry , Alzheimer Disease/pathology , Amyloid beta-Peptides/chemical synthesis , Amyloid beta-Peptides/chemistry , Binding Sites , Blood-Brain Barrier/drug effects , Butyrylcholinesterase/chemistry , Butyrylcholinesterase/therapeutic use , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/chemistry , Humans , Isoindoles/chemical synthesis , Isoindoles/chemistry , Isoindoles/therapeutic use , Ligands , Models, Molecular , Protein Aggregation, Pathological/metabolism , Structure-Activity Relationship
16.
ChemMedChem ; 11(12): 1318-27, 2016 Jun 20.
Article in English | MEDLINE | ID: mdl-26804623

ABSTRACT

Given the complex nature of Alzheimer's disease (AD), compounds that are able to simultaneously address two or more AD-associated targets show greater promise for development into drugs for AD therapy. Herein we report an efficient two-step synthesis and biological evaluation of new racemic benzochromene derivatives as antioxidants, inhibitors of cholinesterase and ß-amyloid (Aß1-42 ) aggregation. Based on the results of the primary screening, we identified 15-(3-methoxyphenyl)-9,11,12,15-tetrahydro-10H,14H-benzo[5,6]chromeno[2,3-d]pyrido[1,2-a]pyrimidin-14-imine (3 e) and 16-(3-methoxyphenyl)-9,10,11,12,13,16-hexahydro-15H-benzo[5',6']chromeno[2',3':4,5]pyrimido[1,2-a]azepin-15-imine (3 f) as new potential multitarget-directed ligands for AD therapy. Further in-depth biological analysis showed that compound 3 f is a good human acetylcholinesterase inhibitor [IC50 =(0.36±0.02) µm], has strong antioxidant activity (3.61 µmol Trolox equivalents), and moderate Aß1-42 antiaggregating power (40.3 %).


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Antioxidants/chemical synthesis , Cholinesterase Inhibitors/therapeutic use , Cholinesterases/metabolism , Imines/chemistry , Peptide Fragments/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Antioxidants/chemistry , Antioxidants/therapeutic use , Binding Sites , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/chemistry , Cholinesterases/chemistry , Humans , Imines/chemical synthesis , Imines/therapeutic use , Ligands , Molecular Docking Simulation , Peptide Fragments/antagonists & inhibitors , Protein Structure, Tertiary
17.
Arch Pharm (Weinheim) ; 348(8): 556-63, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26032855

ABSTRACT

Alzheimer's disease (AD) is a fatal and complex neurodegenerative disorder for which effective treatment remains the unmet challenge. Using donepezil as a starting point, we aimed to develop novel potential anti-AD agents with a multidirectional biological profile. We designed the target compounds as dual binding site acetylcholinesterase inhibitors, where the N-benzylamine pharmacophore is responsible for interactions with the catalytic anionic site of the enzyme. The heteroaromatic fragment responsible for interactions with the peripheral anionic site was modified and three different heterocycles were introduced: isoindoline, isoindolin-1-one, and saccharine. Based on the results of the pharmacological evaluation, we identified compound 8b with a saccharine moiety as the most potent and selective human acetylcholinesterase inhibitor (IC50 = 33 nM) and beta amyloid aggregation inhibitor. It acts as a non-competitive acetylcholinesterase inhibitor and is able to cross the blood-brain barrier in vitro. We believe that compound 8b represents an important lead compound for further development as potential anti-AD agent.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Benzylamines/chemical synthesis , Benzylamines/pharmacology , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/pharmacology , Drug Design , Heterocyclic Compounds/chemical synthesis , Heterocyclic Compounds/pharmacology , Peptide Fragments/metabolism , Acetylcholinesterase/chemistry , Acetylcholinesterase/metabolism , Alzheimer Disease/diagnosis , Alzheimer Disease/enzymology , Amyloid beta-Peptides/chemistry , Binding Sites , Binding, Competitive , Blood-Brain Barrier/metabolism , Butyrylcholinesterase/metabolism , Capillary Permeability , Catalytic Domain , Cholinesterase Inhibitors/metabolism , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/chemistry , GPI-Linked Proteins/metabolism , Kinetics , Ligands , Molecular Docking Simulation , Molecular Targeted Therapy , Peptide Fragments/chemistry , Protein Aggregation, Pathological , Protein Binding , Protein Conformation , Saccharin/analogs & derivatives , Saccharin/chemical synthesis , Saccharin/pharmacology , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...