Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
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.
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
3.
Eur J Med Chem ; 225: 113792, 2021 Dec 05.
Article in English | MEDLINE | ID: mdl-34530376

ABSTRACT

The lack of an effective treatment makes Alzheimer's disease a serious healthcare problem and a challenge for medicinal chemists. Herein we report interdisciplinary research on novel multifunctional ligands targeting proteins and processes involved in the development of the disease: BuChE, 5-HT6 receptors and ß-amyloid aggregation. Structure-activity relationship analyses supported by crystallography and docking studies led to the identification of a fused-type multifunctional ligand 50, with remarkable and balanced potencies against BuChE (IC50 = 90 nM) and 5-HT6R (Ki = 4.8 nM), and inhibitory activity against Aß aggregation (53% at 10 µM). In in vitro ADME-Tox and in vivo pharmacokinetic studies compound 50 showed good stability in the mouse liver microsomes, favourable safety profile and brain permeability with the brain to plasma ratio of 6.79 after p.o. administration in mice, thus being a promising candidate for in vivo pharmacology studies and a solid foundation for further research on effective anti-AD therapies.


Subject(s)
Alzheimer Disease/drug therapy , Butyrylcholinesterase/metabolism , Cholinesterase Inhibitors/pharmacology , Drug Development , Neuroprotective Agents/pharmacology , Receptors, Serotonin/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Animals , Cell Survival/drug effects , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/chemistry , Crystallography, X-Ray , Dose-Response Relationship, Drug , Electrophorus , Hep G2 Cells , Horses , Humans , Male , Mice , Models, Molecular , Molecular Structure , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/chemistry , Protein Aggregates/drug effects , Structure-Activity Relationship
4.
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
5.
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
6.
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
7.
ACS Chem Neurosci ; 9(5): 1195-1214, 2018 05 16.
Article in English | MEDLINE | ID: mdl-29384656

ABSTRACT

Alzheimer's disease (AD) is a major public health problem, which is due to its increasing prevalence and lack of effective therapy or diagnostics. The complexity of the AD pathomechanism requires complex treatment, e.g. multifunctional ligands targeting both the causes and symptoms of the disease. Here, we present new multitarget-directed ligands combining pharmacophore fragments that provide a blockade of serotonin 5-HT6 receptors, acetyl/butyrylcholinesterase inhibition, and amyloid ß antiaggregation activity. Compound 12 has displayed balanced activity as an antagonist of 5-HT6 receptors ( Ki = 18 nM) and noncompetitive inhibitor of cholinesterases (IC50 hAChE = 14 nM, IC50 eqBuChE = 22 nM). In further in vitro studies, compound 12 has shown amyloid ß antiaggregation activity (IC50 = 1.27 µM) and ability to permeate through the blood-brain barrier. The presented findings may provide an excellent starting point for further studies and facilitate efforts to develop new effective anti-AD therapy.


Subject(s)
Alzheimer Disease/etiology , Amyloid beta-Peptides/metabolism , Butyrylcholinesterase/pharmacology , Cholinesterase Inhibitors/pharmacology , Ligands , Alzheimer Disease/drug therapy , Drug Design , Humans , Models, Molecular , Molecular Docking Simulation , Peptide Fragments/metabolism , Structure-Activity Relationship
8.
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
9.
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
10.
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
11.
Eur J Med Chem ; 124: 63-81, 2016 Nov 29.
Article in English | MEDLINE | ID: mdl-27560283

ABSTRACT

As currently postulated, a complex treatment may be key to an effective therapy for Alzheimer's disease (AD). Recent clinical trials in patients with moderate AD have shown a superior effect of the combination therapy of donepezil (a selective acetylcholinesterase inhibitor) with idalopirdine (a 5-HT6 receptor antagonist) over monotherapy with donepezil. Here, we present the first report on the design, synthesis and biological evaluation of a novel class of multifunctional ligands that combines a 5-HT6 receptor antagonist with a cholinesterase inhibitor. Novel multi-target-directed ligands (MTDLs) were designed by combining pharmacophores directed against the 5-HT6 receptor (1-(phenylsulfonyl)-4-(piperazin-1-yl)-1H-indole) and cholinesterases (tacrine or N-benzylpiperidine analogues). In vitro evaluation led to the identification of tacrine derivative 12 with well-balanced potencies against the 5-HT6 receptor (Kb = 27 nM), acetylcholinesterase and butyrylcholinesterase (IC50hAChE = 12 nM, IC50hBuChE = 29 nM). The compound also showed good in vitro blood-brain-barrier permeability (PAMPA-BBB assay), which was confirmed in vivo (open field study). Central cholinomimetic activity was confirmed in vivo in rats using a scopolamine-induced hyperlocomotion model. A novel class of multifunctional ligands with compound 12 as the best derivative in a series represents an excellent starting point for the further development of an effective treatment for AD.


Subject(s)
Alzheimer Disease/diet therapy , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/pharmacology , Drug Design , Molecular Targeted Therapy , Receptors, Serotonin/metabolism , Acetylcholinesterase/metabolism , Alzheimer Disease/enzymology , Alzheimer Disease/metabolism , Animals , Blood-Brain Barrier/metabolism , Butyrylcholinesterase/metabolism , Catalytic Domain , Chemistry Techniques, Synthetic , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/metabolism , Humans , Kinetics , Ligands , Male , Models, Molecular , Protein Conformation , Rats , Rats, Wistar , Receptors, Serotonin/chemistry
12.
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
13.
Postepy Hig Med Dosw (Online) ; 69: 1423-34, 2015 Jan 02.
Article in Polish | MEDLINE | ID: mdl-27259214

ABSTRACT

Selective drugs directed at a single biological target often prove ineffective in the treatment of diseases with a complex pathomechanism, e.g. Alzheimer's disease (AD). This situation prompts researchers to design multi-target-directed ligands (MTDLs), capable of interacting with a number of selected biological targets. The paper outlines the concept of the multi-target-directed ligand design and examples of its use in the search of a cure for AD. In the knowledge-based approach for designing MTDLs, selective ligands of different targets are combined in one molecule. In the screening-based approach, libraries of compounds are screened against selected targets, which allows one to find molecules with a desirable pharmacological profile. It is also possible to obtain multifunctional ligands by performing optimization of a drug with known side activity and transforming it into the main activity, with a simultaneous decrease or complete removal of the original activity. The type of biological targets and applied MTDL design strategy affect the physicochemical and pharmacokinetic properties of the resulting molecules. AD is a multifactorial neurodegenerative disease of the central nervous system associated with the formation of neurofibrillary tangles within neurons, formed by the hyperphosphorylated τ proteins, and extracellular ß-amyloid deposits (senile plaques). Current AD therapy comprises symptomatic drugs that enhance cholinergic neurotransmission or inhibit glutamate receptors. The literature provides numerous examples of compounds which proved in in vitro tests to be multifunctional ligands. Most of them are derivatives of cholinesterase-inhibiting drugs, also capable of inhibiting the aggregation of Aß and showing neuroprotective effects in Aß-induced cytotoxicity assays.


Subject(s)
Alzheimer Disease/drug therapy , Ligands , Neurodegenerative Diseases/drug therapy , Alzheimer Disease/metabolism , Humans , Neurodegenerative Diseases/metabolism , Neurofibrillary Tangles , Neurons/metabolism , Neuroprotective Agents/pharmacology , Plaque, Amyloid , tau Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...