Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
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.
Bioorg Med Chem ; 96: 117535, 2023 12 15.
Article in English | MEDLINE | ID: mdl-37956505

ABSTRACT

As Alzheimer's disease (AD) is a neurodegenerative disease with a complex pathogenesis, the exploration of multi-target drugs may be an effective strategy for AD treatment. Multifunctional small molecular agents can be obtained by connecting two or more active drugs or privileged pharmacophores by multicomponent reactions (MCRs). In this paper, two series of polysubstituted pyrazine derivatives with multifunctional moieties were designed as anti-AD agents and synthesized by Passerini-3CR and Ugi-4CR. Since the oxidative stress plays an important role in the pathological process of AD, the antioxidant activities of the newly synthesized compounds were first evaluated. Subsequently, selected active compounds were further screened in a series of AD-related bioassays, including Aß1-42 self-aggregation and deaggregation, BACE-1 inhibition, metal chelation, and protection of SH-SY5Y cells from H2O2-induced oxidative damage. Compound A3B3C1 represented the best one with multifunctional potencies. Mechanism study showed that A3B3C1 acted on Nrf2/ARE signaling pathway, thus increasing the expression of related antioxidant proteins NQO1 and HO-1 to normal cell level. Furthermore, A3B3C1 showed good in vitro human plasma and liver microsome stability, indicating a potential for further development as multifunctional anti-AD agent.


Subject(s)
Alzheimer Disease , Neuroblastoma , Neurodegenerative Diseases , Neuroprotective Agents , Humans , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Antioxidants/therapeutic use , Hydrogen Peroxide/pharmacology , Cholinesterase Inhibitors/pharmacology , Oxidative Stress , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Drug Design , Acetylcholinesterase/metabolism
3.
Eur J Med Chem ; 261: 115832, 2023 Dec 05.
Article in English | MEDLINE | ID: mdl-37837674

ABSTRACT

Alzheimer's disease (AD) is a global health problem in the medical sector that will increase over time. The limited treatment of AD leads to the search for a new clinical candidate. Considering the multifactorial nature of AD, a strategy targeting number of regulatory proteins involved in the development of the disease is an effective approach. Here, we present a discovery of new multi-target-directed ligands (MTDLs), purposely designed as GABA transporter (GAT) inhibitors, that successfully provide the inhibitory activity against butyrylcholinesterase (BuChE), ß-secretase (BACE1), amyloid ß aggregation and calcium channel blockade activity. The selected GAT inhibitors, 19c and 22a - N-benzylamide derivatives of 4-aminobutyric acid, displayed the most prominent multifunctional profile. Compound 19c (mGAT1 IC50 = 10 µM, mGAT4 IC50 = 12 µM and BuChE IC50 = 559 nM) possessed the highest hBACE1 and Aß40 aggregation inhibitory activity (IC50 = 1.57 µM and 99 % at 10 µM, respectively). Additionally, it showed a decrease in both the elongation and nucleation constants of the amyloid aggregation process. In contrast compound 22a represented the highest activity and a mixed-type of eqBuChE inhibition (IC50 = 173 nM) with hBACE1 (IC50 = 9.42 µM), Aß aggregation (79 % at 10 µM) and mGATs (mGAT1 IC50 = 30 µM, mGAT4 IC50 = 25 µM) inhibitory activity. Performed molecular docking studies described the mode of interactions with GATs and enzymatic targets. In ADMET in vitro studies both compounds showed acceptable metabolic stability and low neurotoxicity. Successfully, compounds 19c and 22a at the dose of 30 mg/kg possessed statistically significant antiamnesic properties in a mouse model of amnesia caused by scopolamine and assessed in the novel object recognition (NOR) task or the passive avoidance (PA) task.


Subject(s)
Alzheimer Disease , Butyrylcholinesterase , Mice , Animals , Butyrylcholinesterase/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid Precursor Protein Secretases/metabolism , GABA Plasma Membrane Transport Proteins/metabolism , Cholinesterase Inhibitors/metabolism , Molecular Docking Simulation , Structure-Activity Relationship , Drug Design , Aspartic Acid Endopeptidases/metabolism , Acetylcholinesterase/metabolism
4.
Int J Mol Sci ; 24(11)2023 May 23.
Article in English | MEDLINE | ID: mdl-37298087

ABSTRACT

Alzheimer's disease (AD) is a complex disease with an unknown etiology. Available treatments, limited to cholinesterase inhibitors and N-methyl-d-aspartate receptor (NMDAR) antagonists, provide symptomatic relief only. As single-target therapies have not proven effective, rational specific-targeted combination into a single molecule represents a more promising approach for treating AD, and is expected to yield greater benefits in alleviating symptoms and slowing disease progression. In the present study, we designed, synthesized, and biologically evaluated 24 novel N-methylpropargylamino-quinazoline derivatives. Initially, compounds were thoroughly inspected by in silico techniques determining their oral and CNS availabilities. We tested, in vitro, the compounds' effects on cholinesterases and monoamine oxidase A/B (MAO-A/B), as well as their impacts on NMDAR antagonism, dehydrogenase activity, and glutathione levels. In addition, we inspected selected compounds for their cytotoxicity on undifferentiated and differentiated neuroblastoma SH-SY5Y cells. We collectively highlighted II-6h as the best candidate endowed with a selective MAO-B inhibition profile, NMDAR antagonism, an acceptable cytotoxicity profile, and the potential to permeate through BBB. The structure-guided drug design strategy applied in this study imposed a novel concept for rational drug discovery and enhances our understanding on the development of novel therapeutic agents for treating AD.


Subject(s)
Alzheimer Disease , Neuroblastoma , Humans , Alzheimer Disease/drug therapy , Monoamine Oxidase Inhibitors/therapeutic use , Neuroblastoma/drug therapy , Cholinesterase Inhibitors/therapeutic use , Monoamine Oxidase/metabolism , Drug Design , Acetylcholinesterase/metabolism , Structure-Activity Relationship
5.
Bioorg Med Chem ; 88-89: 117333, 2023 06 06.
Article in English | MEDLINE | ID: mdl-37236021

ABSTRACT

Butyrylcholinesterase (BuChE) and amyloid ß (Aß) aggregation remain important biological target and mechanism in the search for effective treatment of Alzheimer's disease. Simultaneous inhibition thereof by the application of multifunctional agents may lead to improvement in terms of symptoms and causes of the disease. Here, we present the rational design, synthesis, biological evaluation and molecular modelling studies of novel series of fluorene-based BuChE and Aß inhibitors with drug-like characteristics and advantageous Central Nervous System Multiparameter Optimization scores. Among 17 synthesized and tested compounds, we identified 22 as the most potent eqBuChE inhibitor with IC50 of 38 nM and 37.4% of Aß aggregation inhibition at 10 µM. Based on molecular modelling studies, including molecular dynamics, we determined the binding mode of the compounds within BuChE and explained the differences in the activity of the two enantiomers of compound 22. A novel series of fluorenyl compounds meeting the drug-likeness criteria seems to be a promising starting point for further development as anti-Alzheimer agents.


Subject(s)
Alzheimer Disease , Butyrylcholinesterase , Humans , Butyrylcholinesterase/metabolism , Amyloid beta-Peptides/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Cholinesterase Inhibitors/pharmacology , Cholinesterase Inhibitors/therapeutic use , Structure-Activity Relationship , Molecular Dynamics Simulation , Acetylcholinesterase/metabolism , Drug Design , Molecular Structure , Molecular Docking Simulation
6.
Eur J Med Chem ; 249: 115135, 2023 Mar 05.
Article in English | MEDLINE | ID: mdl-36696766

ABSTRACT

The symptomatic and disease-modifying effects of butyrylcholinesterase (BuChE) inhibitors provide an encouraging premise for researching effective treatments for Alzheimer's disease. Here, we examined a series of compounds with a new chemical scaffold based on 3-(cyclohexylmethyl)amino-2-hydroxypropyl, and we identified a highly selective hBuChE inhibitor (29). Based on extensive in vitro and in vivo evaluations of the compound and its enantiomers, (R)-29 was identified as a promising candidate for further development. Compound (R)-29 is a potent hBuChE inhibitor (IC50 = 40 nM) with selectivity over AChE and relevant off-targets, including H1, M1, α1A and ß1 receptors. The compound displays high metabolic stability on human liver microsomes (90% of the parent compound after 2 h of incubation), and its safety was confirmed through examining the cytotoxicity on the HepG2 cell line (LC50 = 2.85 µM) and hERG inhibition (less than 50% at 10 µM). While (rac)-29 lacked an effect in vivo and showed limited penetration to the CNS in pharmacokinetics studies, compound (R)-29 exhibited a procognitive effect at 15 mg/kg in the passive avoidance task in scopolamine-treated mice.


Subject(s)
Alzheimer Disease , Butyrylcholinesterase , Mice , Animals , Humans , Butyrylcholinesterase/metabolism , Crystallography , Cholinesterase Inhibitors/chemistry , Alzheimer Disease/metabolism , Scopolamine/pharmacology , Acetylcholinesterase/metabolism , Structure-Activity Relationship , Molecular Docking Simulation
7.
Eur J Med Chem ; 225: 113783, 2021 Dec 05.
Article in English | MEDLINE | ID: mdl-34461507

ABSTRACT

Multifunctional ligands as an essential variant of polypharmacology are promising candidates for the treatment of multi-factorial diseases like Alzheimer's disease. Based on clinical evidence and following the paradigm of multifunctional ligands we have rationally designed and synthesized a series of compounds targeting processes involved in the development of the disease. The biological evaluation led to the discovery of two compounds with favorable pharmacological characteristics and ADMET profile. Compounds 17 and 35 are 5-HT6R antagonists (Ki = 13 nM and Ki = 15 nM respectively) and cholinesterase inhibitors with distinct mechanisms of enzyme inhibition. Compound 17, a tacrine derivative is a reversible inhibitor of acetyl- and butyrylcholinesterase (IC50 = 8 nM and IC50 = 24 nM respectively), while compound 35 with rivastigmine-derived phenyl N-ethyl-N-methylcarbamate fragment is a selective, pseudo-irreversible inhibitor of butyrylcholinesterase (IC50 = 455 nM). Both compounds inhibit aggregation of amyloid ß in vitro (75% for compound 17 and 68% for 35 at 10 µM) moreover, compound 35 is a potent tau aggregation inhibitor in cellulo (79%). In ADMET in vitro studies both compounds showed acceptable metabolic stability on mouse liver microsomes (28% and 60% for compound 17 and 35 respectively), no or little effect on CYP3A4 and 2D6 up to a concentration of 10 µM and lack of toxicity on HepG2 cell line (IC50 values of 80 and 21 µM, for 17 and 35 respectively). Based on the pharmacological characteristics and favorable pharmacokinetic properties, we propose compounds 17 and 35 as an excellent starting point for further optimization and in-depth biological studies.


Subject(s)
Cholinesterase Inhibitors/pharmacology , Drug Discovery , Indoles/pharmacology , Acetylcholinesterase/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Animals , Butyrylcholinesterase/metabolism , Cell Proliferation/drug effects , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/chemistry , Dose-Response Relationship, Drug , Electrophorus , Hep G2 Cells , Horses , Humans , Indoles/chemical synthesis , Indoles/chemistry , Ligands , Mice , Microsomes, Liver/chemistry , Microsomes, Liver/metabolism , Molecular Docking Simulation , Molecular Structure , Protein Aggregates/drug effects , Receptors, Serotonin/metabolism , Structure-Activity Relationship , tau Proteins/antagonists & inhibitors , tau Proteins/metabolism
8.
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
9.
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
10.
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
11.
Eur J Med Chem ; 187: 111916, 2020 Feb 01.
Article in English | MEDLINE | ID: mdl-31812794

ABSTRACT

Complex pathomechanism of Alzheimer's disease (AD) prompts researchers to develop multifunctional molecules in order to find effective therapy against AD. We designed and synthesized novel multifunctional ligands for which we assessed their activities towards butyrylcholinesterase, beta secretase, amyloid beta (Aß) and tau protein aggregation as well as antioxidant and metal-chelating properties. All compounds showed dual anti-aggregating properties towards Aß and tau protein in the in cellulo assay in Escherichia coli. Of particular interest are compounds 24b and 25b, which efficiently inhibit aggregation of Aß and tau protein at 10 µM (24b: 45% for Aß, 53% for tau; 25b: 49% for Aß, 54% for tau). They display free radical scavenging capacity and antioxidant activity in ABTS and FRAP assays, respectively, and selectively chelate copper ions. Compounds 24b and 25b are also the most potent inhibitors of BuChE with IC50 of 2.39 µM and 1.94 µM, respectively. Promising in vitro activities of the presented multifunctional ligands as well as their original scaffold are a very interesting starting point for further research towards effective anti-AD treatment.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/antagonists & inhibitors , Antioxidants/pharmacology , Butyrylcholinesterase/metabolism , Chelating Agents/pharmacology , Cholinesterase Inhibitors/pharmacology , Pyrrolidines/pharmacology , Acetylcholinesterase/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Antioxidants/chemical synthesis , Antioxidants/chemistry , Benzothiazoles/antagonists & inhibitors , Chelating Agents/chemical synthesis , Chelating Agents/chemistry , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/chemistry , Dose-Response Relationship, Drug , Electrophorus , Fluorescence Recovery After Photobleaching , Horses , Humans , Molecular Structure , Protein Aggregates/drug effects , Pyrrolidines/chemical synthesis , Pyrrolidines/chemistry , Structure-Activity Relationship , Sulfonic Acids/antagonists & inhibitors
12.
J Med Chem ; 62(24): 11416-11422, 2019 12 26.
Article in English | MEDLINE | ID: mdl-31724859

ABSTRACT

New tritarget small molecules combining Ca2+ channels blockade, cholinesterase, and H3 receptor inhibition were obtained by multicomponent synthesis. Compound 3p has been identified as a very promising lead, showing good Ca2+ channels blockade activity (IC50 = 21 ± 1 µM), potent affinity against hH3R (Ki = 565 ± 62 nM), a moderate but selective hBuChE inhibition (IC50 = 7.83 ± 0.10 µM), strong antioxidant power (3.6 TE), and ability to restore cognitive impairment induced by lipopolysaccharide.


Subject(s)
Alzheimer Disease/drug therapy , Calcium Channel Blockers/pharmacology , Cholinesterase Inhibitors/pharmacology , Neuroprotective Agents/pharmacology , Receptors, Histamine H3/chemistry , Small Molecule Libraries/pharmacology , Vasodilator Agents/pharmacology , Alzheimer Disease/metabolism , Animals , Calcium Channel Blockers/chemistry , Cholinesterase Inhibitors/chemistry , Humans , Memory Disorders/drug therapy , Memory Disorders/metabolism , Mice , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Neuroprotective Agents/chemistry , Small Molecule Libraries/chemistry , Tumor Cells, Cultured , Vasodilator Agents/chemistry
13.
Arch Pharm (Weinheim) ; 352(7): e1900041, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31162703

ABSTRACT

Serotonin 5-HT6 receptors, butyrylcholinesterase (BuChE) and oxidative stress are related to the pathophysiology of Alzheimer's disease. Inhibition of BuChE provides symptomatic treatment of the disease and the same effect was demonstrated for 5-HT 6 antagonists in clinical trials. Oxidative stress is regarded as a major and primary factor contributing to the development of Alzheimer's disease; therefore, antioxidant agents may provide a disease-modifying effect. Combining BuChE inhibition, 5-HT 6 antagonism, and antioxidant properties may result in multitarget-directed ligands providing cognition-enhancing properties with neuroprotective activity. On the basis of the screening of the library of 5-HT 6 antagonists against BuChE, we selected two compounds and designed their structural modifications that could lead to improved BuChE inhibitory activity. We synthesized two series of compounds and tested their affinity and functional activity at 5-HT 6 receptors, BuChE inhibitory activity and antioxidant properties. Compound 12 with K i and K b values against 5-HT 6 receptors of 41.8 and 74 nM, respectively, an IC 50 value of 5 µM against BuChE and antioxidant properties exceeding the activity of ascorbic acid is a promising lead structure for further development of anti-Alzheimer's agents.


Subject(s)
Antioxidants/pharmacology , Butyrylcholinesterase/metabolism , Cholinesterase Inhibitors/pharmacology , Receptors, Serotonin/metabolism , Serotonin Antagonists/pharmacology , Animals , Antioxidants/chemical synthesis , Antioxidants/chemistry , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/chemistry , Electrophorus , Horses , Humans , Models, Molecular , Molecular Structure , Oxidative Stress/drug effects , Serotonin Antagonists/chemical synthesis , Serotonin Antagonists/chemistry , Triazines/antagonists & inhibitors
14.
Molecules ; 23(2)2018 Feb 07.
Article in English | MEDLINE | ID: mdl-29414887

ABSTRACT

The complex nature of Alzheimer's disease calls for multidirectional treatment. Consequently, the search for multi-target-directed ligands may lead to potential drug candidates. The aim of the present study is to seek multifunctional compounds with expected activity against disease-modifying and symptomatic targets. A series of 15 drug-like various substituted derivatives of 2-(benzylamino-2-hydroxyalkyl)isoindoline-1,3-diones was designed by modification of cholinesterase inhibitors toward ß-secretase inhibition. All target compounds have been synthesized and tested against eel acetylcholinesterase (eeAChE), equine serum butyrylcholinesterase (eqBuChE), human ß-secretase (hBACE-1), and ß-amyloid (Aß-aggregation). The most promising compound, 12 (2-(5-(benzylamino)-4-hydroxypentyl)isoindoline-1,3-dione), displayed inhibitory potency against eeAChE (IC50 = 3.33 µM), hBACE-1 (43.7% at 50 µM), and Aß-aggregation (24.9% at 10 µM). Molecular modeling studies have revealed possible interaction of compound 12 with the active sites of both enzymes-acetylcholinesterase and ß-secretase. IN CONCLUSION: modifications of acetylcholinesterase inhibitors led to the discovery of a multipotent anti-Alzheimer's agent, with moderate and balanced potency, capable of inhibiting acetylcholinesterase, a symptomatic target, and disease-modifying targets: ß-secretase and Aß-aggregation.


Subject(s)
Drug Design , Isoindoles/chemistry , Isoindoles/pharmacology , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/chemistry , Butyrylcholinesterase/chemistry , Chemistry Techniques, Synthetic , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/pharmacology , Dose-Response Relationship, Drug , Hydrogen Bonding , Inhibitory Concentration 50 , Isoindoles/chemical synthesis , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Structure-Activity Relationship
15.
ACS Chem Neurosci ; 9(5): 1074-1094, 2018 05 16.
Article in English | MEDLINE | ID: mdl-29345897

ABSTRACT

The multitarget approach is a promising paradigm in drug discovery, potentially leading to new treatment options for complex disorders, such as Alzheimer's disease. Herein, we present the discovery of a unique series of 1-benzylamino-2-hydroxyalkyl derivatives combining inhibitory activity against butyrylcholinesterase, ß-secretase, ß-amyloid, and tau protein aggregation, all related to mechanisms which underpin Alzheimer's disease. Notably, diphenylpropylamine derivative 10 showed balanced activity against both disease-modifying targets, inhibition of ß-secretase (IC50  hBACE-1 = 41.60 µM), inhibition of amyloid ß aggregation (IC50 Aß = 3.09 µM), inhibition of tau aggregation (55% at 10 µM); as well as against symptomatic targets, butyrylcholinesterase inhibition (IC50  hBuChE = 7.22 µM). It might represent an encouraging starting point for development of multifunctional disease-modifying anti-Alzheimer's agents.


Subject(s)
Alzheimer Disease/drug therapy , Butyrylcholinesterase/pharmacology , Cholinesterase Inhibitors/pharmacology , Drug Design , Alzheimer Disease/metabolism , Amyloid beta-Peptides/drug effects , Amyloid beta-Peptides/metabolism , Humans , Molecular Docking Simulation/methods , Peptide Fragments/metabolism , Structure-Activity Relationship , tau Proteins/drug effects
16.
Future Med Chem ; 9(15): 1835-1854, 2017 10.
Article in English | MEDLINE | ID: mdl-28925729

ABSTRACT

The emergence of a multitarget design approach in the development of new potential anti-Alzheimer's disease agents has resulted in the discovery of many multifunctional compounds focusing on various targets. Among them the largest group comprises inhibitors of both cholinesterases, with additional anti-ß-amyloid aggregation activity. This review describes recent advances in this research area and presents the most interesting compounds reported over a 2-year span (2015-2016). The majority of hybrids possess heterodimeric structures obtained by linking structurally active fragments interacting with different targets. Multipotent cholinesterase inhibitors with ß-amyloid antiaggregating activity may additionally possess antioxidative, neuroprotective or metal-chelating properties or less common features such as anti-ß-secretase or τ-antiaggregation activity.


Subject(s)
Amyloid beta-Peptides/metabolism , Cholinesterases/metabolism , Alkaloids/chemistry , Alkaloids/metabolism , Alkaloids/therapeutic use , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/antagonists & inhibitors , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/metabolism , Cholinesterase Inhibitors/therapeutic use , Cholinesterases/chemistry , Donepezil , Humans , Indans/chemistry , Indans/metabolism , Inhibitory Concentration 50 , Piperidines/chemistry , Piperidines/metabolism , Rivastigmine/chemistry , Rivastigmine/metabolism , Tacrine/chemistry , Tacrine/metabolism
17.
Eur J Med Chem ; 126: 576-589, 2017 Jan 27.
Article in English | MEDLINE | ID: mdl-27918993

ABSTRACT

Herein we report an efficient two step synthesis and biological assessment of 12 racemic tetrahydropyranodiquinolin-8-amines derivatives as antioxidant, cholinesterase inhibitors and non-hepatotoxic agents. Based on the results of the primary screening, we identified 7-(3-methoxyphenyl)-9,10,11,12-tetrahydro-7H-pyrano[2,3-b:5,6-h']diquinolin-8-amine (2h) as a particularly interesting non-hepatotoxic compound that shows moderate antioxidant activity (1.83 equiv Trolox in the ORAC assay), a non competitive inhibition of hAChE (IC50 = 0.75 ± 0.01 µM), and brain permeable as determined by the PAMPA-Blood Brain Barrier assay.


Subject(s)
Aminoquinolines/pharmacology , Antioxidants/pharmacology , Cholinesterase Inhibitors/pharmacology , Acetylcholinesterase , Alzheimer Disease/drug therapy , Aminoquinolines/chemical synthesis , Antioxidants/chemistry , Blood-Brain Barrier/metabolism , Chemical and Drug Induced Liver Injury , Cholinesterase Inhibitors/chemistry , GPI-Linked Proteins/antagonists & inhibitors , Humans
18.
Eur J Med Chem ; 125: 676-695, 2017 Jan 05.
Article in English | MEDLINE | ID: mdl-27721153

ABSTRACT

The complexity of Alzheimer's disease (AD) calls for search of multifunctional compounds as potential candidates for effective therapy. A series of phthalimide and saccharin derivatives linked by different alicyclic fragments (piperazine, hexahydropyrimidine, 3-aminopyrrolidine or 3-aminopiperidine) with phenylalkyl moieties attached have been designed, synthesized, and evaluated as multifunctional anti-AD agents with cholinesterase, ß-secretase and ß-amyloid inhibitory activities. In vitro studies showed that the majority of saccharin derivatives with piperazine moiety and one phthalimide derivative with 3-aminopiperidine fragment exhibited inhibitory potency toward acetylcholinesterase (AChE) with EeAChE IC50 values ranging from 0.83 µM to 19.18 µM. The target compounds displayed inhibition of human ß-secretase-1 (hBACE1) ranging from 26.71% to 61.42% at 50 µM concentration. Among these compounds, two multifunctional agents (26, [2-(2-(4-benzylpiperazin-1-yl)ethyl)benzo[d]isothiazol-3(2H)-one 1,1-dioxide] and 52, 2-(2-(3-(3,5-difluorobenzylamino)piperidin-1-yl)ethyl)isoindoline-1,3-dione) have been identified. Compound 26 exhibited the highest inhibitory potency against EeAChE (IC50 = 0.83 µM) and inhibitory activity against hBACE1 (33.61% at 50 µM). Compound 52 is a selective AChE inhibitor (IC50 AChE = 6.47 µM) with BACE1 inhibitory activity (26.3% at 50 µM) and it displays the most significant Aß anti-aggregating properties among all the obtained compounds (39% at 10 µM). Kinetic and molecular modeling studies indicate that 26 may act as non-competitive AChE inhibitor able to interact with both catalytic and peripheral active site of the enzyme.


Subject(s)
Amines/chemistry , Amines/pharmacology , Phthalimides/chemistry , Phthalimides/pharmacology , Saccharin/chemistry , Saccharin/pharmacology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Binding Sites , Blood-Brain Barrier/drug effects , Cholinesterases/metabolism , Drug Delivery Systems , Drug Design , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Inhibitory Concentration 50 , Molecular Structure , Peptide Fragments/metabolism , Phthalimides/chemical synthesis , Protein Aggregation, Pathological , Protein Binding/drug effects , Saccharin/chemical synthesis
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL