Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Sci Rep ; 10(1): 14150, 2020 08 25.
Article in English | MEDLINE | ID: mdl-32843666

ABSTRACT

We herein report the synthesis, antioxidant power and neuroprotective properties of nine homo-bis-nitrones HBNs 1-9 as alpha-phenyl-N-tert-butylnitrone (PBN) analogues for stroke therapy. In vitro neuroprotection studies of HBNs 1-9 against Oligomycin A/Rotenone and in an oxygen-glucose-deprivation model of ischemia in human neuroblastoma cell cultures, indicate that (1Z,1'Z)-1,1'-(1,3-phenylene)bis(N-benzylmethanimine oxide) (HBN6) is a potent neuroprotective agent that prevents the decrease in neuronal metabolic activity (EC50 = 1.24 ± 0.39 µM) as well as necrotic and apoptotic cell death. HBN6 shows strong hydroxyl radical scavenger power (81%), and capacity to decrease superoxide production in human neuroblastoma cell cultures (maximal activity = 95.8 ± 3.6%), values significantly superior to the neuroprotective and antioxidant properties of the parent PBN. The higher neuroprotective ability of HBN6 has been rationalized by means of Density Functional Theory calculations. Calculated physicochemical and ADME properties confirmed HBN6 as a hit-agent showing suitable drug-like properties. Finally, the contribution of HBN6 to brain damage prevention was confirmed in a permanent MCAO setting by assessing infarct volume outcome 48 h after stroke in drug administered experimental animals, which provides evidence of a significant reduction of the brain lesion size and strongly suggests that HBN6 is a potential neuroprotective agent against stroke.


Subject(s)
Brain Ischemia/drug therapy , Cyclic N-Oxides/chemistry , Free Radical Scavengers/therapeutic use , Neurons/drug effects , Neuroprotection/drug effects , Neuroprotective Agents/therapeutic use , Nitrogen Oxides/therapeutic use , Animals , Apoptosis/drug effects , Brain Ischemia/chemically induced , Cell Line, Tumor , Disease Models, Animal , Drug Evaluation, Preclinical , Free Radical Scavengers/chemical synthesis , Free Radical Scavengers/pharmacology , Glucose/pharmacology , Infarction, Middle Cerebral Artery/drug therapy , Lipid Peroxidation/drug effects , Lipoxygenase Inhibitors/pharmacology , Male , Mice , Mice, Inbred C57BL , Molecular Structure , Neuroblastoma/pathology , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/pharmacology , Nitrogen Oxides/chemical synthesis , Nitrogen Oxides/pharmacology , Oligomycins/toxicity , Oxygen/pharmacology , Rotenone/toxicity
2.
J Med Chem ; 63(21): 12485-12510, 2020 11 12.
Article in English | MEDLINE | ID: mdl-32672958

ABSTRACT

3-n-Butylphthalide (NBP) as well as its derivatives and analogues (NBPs), in racemic or enantiomerically pure forms, possess potent and diverse pharmacological properties and have shown a great potential therapeutic interest for many human conditions, especially for cerebral ischemia. This Perspective outlines the synthesis and therapeutic applications of NBPs.


Subject(s)
Apium/chemistry , Benzofurans/chemistry , Brain Ischemia/drug therapy , Neuroprotective Agents/therapeutic use , Animals , Apium/metabolism , Asthma/drug therapy , Asthma/pathology , Benzofurans/chemical synthesis , Benzofurans/therapeutic use , Brain Ischemia/pathology , Humans , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/chemistry , Seeds/chemistry , Seeds/metabolism , Stereoisomerism , Stroke/drug therapy , Stroke/pathology
3.
Int J Mol Sci ; 21(11)2020 May 30.
Article in English | MEDLINE | ID: mdl-32486316

ABSTRACT

In this communication, we report the synthesis and cholinesterase (ChE)/monoamine oxidase (MAO) inhibition of 19 quinolinones (QN1-19) and 13 dihydroquinolinones (DQN1-13) designed as potential multitarget small molecules (MSM) for Alzheimer's disease therapy. Contrary to our expectations, none of them showed significant human recombinant MAO inhibition, but compounds QN8, QN9, and DQN7 displayed promising human recombinant acetylcholinesterase (hrAChE) and butyrylcholinesterase (hrBuChE) inhibition. In particular, molecule QN8 was found to be a potent and quite selective non-competitive inhibitor of hrAChE (IC50 = 0.29 µM), with Ki value in nanomolar range (79 nM). Pertinent docking analysis confirmed this result, suggesting that this ligand is an interesting hit for further investigation.


Subject(s)
Alzheimer Disease/drug therapy , Cholinesterase Inhibitors/pharmacology , Monoamine Oxidase Inhibitors/pharmacology , Quinolones/pharmacology , Acetylcholinesterase/metabolism , Drug Design , Drug Evaluation, Preclinical , Humans , Inhibitory Concentration 50 , Kinetics , Ligands , Magnetic Resonance Spectroscopy , Molecular Docking Simulation , Monoamine Oxidase/metabolism , Recombinant Proteins/metabolism , Structure-Activity Relationship
4.
Curr Alzheimer Res ; 16(9): 815-820, 2019.
Article in English | MEDLINE | ID: mdl-31660831

ABSTRACT

BACKGROUND: Many factors are involved in Alzheimer's Disease (AD) such as amyloid plaques, neurofibrillary tangles, cholinergic deficit and oxidative stress. To counter the complexity of the disease the new approach for drug development is to create a single molecule able to act simultaneously on different targets. OBJECTIVE: We conceived eight drug likeliness compounds targeting the inhibition of cholinesterases and the scavenging of radicals. METHODS: We synthesised the new molecules by the Passerini multicomponent reaction and evaluated their inhibitory activities against acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE) as well as their antioxidant activities by the Oxygen Radical Absorbance Capacity (ORAC) assay. The lipinski's rule for drug likeness and in silico ADME prediction was also performed. RESULTS: Compounds 4f [IC50 (EeAChE) = 0.30 µM; IC50 (eqBuChE) = 0.09 µM; ORAC = 0.64 TE] and 4h [IC50 (EeAChE) = 1 µM; IC50 (eqBuChE) = 0.03 µM; ORAC = 0.50 TE] were identified as hits for further development. CONCLUSION: The Passerini reaction allowed us the facile synthesis of ditarget molecules of interest for the treatment of AD.


Subject(s)
Alzheimer Disease/drug therapy , Cholinesterase Inhibitors/pharmacology , Chromones/pharmacology , Donepezil/pharmacology , Acetylcholinesterase/metabolism , Alzheimer Disease/metabolism , Butyrylcholinesterase/metabolism , Cholinesterase Inhibitors/chemical synthesis , Chromones/chemical synthesis , Donepezil/chemical synthesis , Drug Evaluation, Preclinical , Free Radical Scavengers/chemical synthesis , Free Radical Scavengers/pharmacology , Humans
5.
Biomolecules ; 9(8)2019 08 19.
Article in English | MEDLINE | ID: mdl-31430943

ABSTRACT

Tacrine was the first drug to be approved for Alzheimer's disease (AD) treatment, acting as a cholinesterase inhibitor. The neuropathological hallmarks of AD are amyloid-rich senile plaques, neurofibrillary tangles, and neuronal degeneration. The portfolio of currently approved drugs for AD includes acetylcholinesterase inhibitors (AChEIs) and N-methyl-d-aspartate (NMDA) receptor antagonist. Squaric acid is a versatile structural scaffold capable to be easily transformed into amide-bearing compounds that feature both hydrogen bond donor and acceptor groups with the possibility to create multiple interactions with complementary sites. Considering the relatively simple synthesis approach and other interesting properties (rigidity, aromatic character, H-bond formation) of squaramide motif, we combined this scaffold with different tacrine-based derivatives. In this study, we developed 21 novel dimers amalgamating squaric acid with either tacrine, 6-chlorotacrine or 7-methoxytacrine representing various AChEIs. All new derivatives were evaluated for their anti-cholinesterase activities, cytotoxicity using HepG2 cell line and screened to predict their ability to cross the blood-brain barrier. In this contribution, we also report in silico studies of the most potent AChE and BChE inhibitors in the active site of these enzymes.


Subject(s)
Acetylcholinesterase/metabolism , Butyrylcholinesterase/metabolism , Cholinesterase Inhibitors/pharmacology , Quinine/analogs & derivatives , Tacrine/pharmacology , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/chemistry , Dose-Response Relationship, Drug , Humans , Kinetics , Models, Molecular , Molecular Structure , Quinine/chemistry , Quinine/pharmacology , Structure-Activity Relationship , Tacrine/chemistry
6.
Bioorg Chem ; 91: 103205, 2019 10.
Article in English | MEDLINE | ID: mdl-31446330

ABSTRACT

In this work we describe the synthesis, Ca+2 channel blockade capacity and antioxidant power of N3,N5-bis(2-(5-methoxy-1H-indol-3-yl)ethyl)-2,6-dimethyl-4-aryl-1,4-dihydropyridine-3,5-dicarboxamides 1-9, a number of multi-target small 1,4-dihydropyridines (DHP), designed by juxtaposition of melatonin and nimodipine. As a result, we have identified antioxidant DHP 7 (Ca2+ channel blockade: 55%, and 8.78 Trolox/Equivalents), the most balanced DHP analyzed here, for potential Alzheimer's disease therapy.


Subject(s)
Alzheimer Disease/drug therapy , Antioxidants/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels/chemistry , Calcium/metabolism , Dihydropyridines/pharmacology , Neuroblastoma/drug therapy , Humans , Melatonin/pharmacology , Neuroblastoma/pathology , Nimodipine/pharmacology , Tumor Cells, Cultured
7.
ACS Chem Neurosci ; 9(3): 401-403, 2018 03 21.
Article in English | MEDLINE | ID: mdl-29465220

ABSTRACT

No selective drugs exist, and we have been designing, synthesizing, and evaluating multitarget-directed ligands since the beginning of modern medicinal chemistry, without knowing it, most possibly. The challenge to discover the efficient Multi-Target Small Molecules (MTSMs) for Alzheimer's disease (AD) therapy implies to identify the key combination of biological targets to modulate them, thus including in the design the corresponding pharmacophoric groups able to do it. Universal and polyvalent pharmacophoric groups, able to modulate diverse receptors or enzymatic systems, would simplify the drug discovery process leading to new and more efficient MTSMs for AD.


Subject(s)
Alzheimer Disease/drug therapy , Cholinesterase Inhibitors/pharmacology , Drug Design , Drug Discovery , Ligands , Humans
8.
Future Med Chem ; 9(8): 765-780, 2017 05.
Article in English | MEDLINE | ID: mdl-28498717

ABSTRACT

Melatonin is an indoleamine produced mainly in the pineal gland. The natural decline of melatonin levels with aging strongly contributes to the development of neurodegenerative disorders. Pleiotropic actions displayed by melatonin prevent several processes involved in neurodegeneration such as neuroinflammation, oxidative stress, excitotoxicity and/or apoptosis. This review focuses on a number of melatonin hybrids resulting from the juxtaposition of tacrine, berberine, tamoxifen, curcumin, N,N-dibenzyl(N-methyl)amine, among others, with potential therapeutic effects for the treatment of neurodegenerative diseases.


Subject(s)
Drug Design , Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/therapeutic use , Humans , Melatonin/chemical synthesis , Melatonin/chemistry , Melatonin/therapeutic use , Molecular Structure , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/chemistry
9.
Mini Rev Med Chem ; 15(8): 648-58, 2015.
Article in English | MEDLINE | ID: mdl-25694076

ABSTRACT

This paper describes our preliminary results on the ADMET, synthesis, biochemical evaluation, and molecular modeling of racemic HuperTacrines (HT), new hybrids resulting from the juxtaposition of huperzine A and tacrine for the potential treatment of Alzheimer's disease (AD). The synthesis of these HT was executed by Friedländer-type reactions of 2-amino-6-oxo-1,6-dihydropyridine-3-carbonitriles, or 7-amino-2-oxo-1,2,3,4-tetrahydro-1,6-naphthyridine- 8-carbonitriles, with cyclohexanone. In the biochemical evaluation, initial and particular attention was devoted to test their toxicity on human hepatoma cells, followed by the in vitro inhibition of human cholinesterases (hAChE, and hBuChE), and the kinetics/mechanism of the inhibition of the most potent HT; simultaneous molecular modeling on the best HT provided the key binding interactions with the human cholinesterases. >From these analyses, (±)-5-amino-3-methyl- 3,4,6,7,8,9-hexahydrobenzo[b][1,8]naphthyridin-2(1H)-one (HT1) and (±)-5-amino-3-(2,6-dichlorophenyl)-3,4,6,7,8,9- hexahydrobenzo[b][1,8]naphthyridin-2(1H)-one (HT3) have emerged as characterized by extremely low liver toxicity reversible mixed-type, selective hAChE and, quite selective irreversible hBuChEIs, respectively, showing also good druglike properties for AD-targeted drugs.


Subject(s)
Alkaloids/chemistry , Alkaloids/pharmacology , Alzheimer Disease/drug therapy , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/pharmacology , Sesquiterpenes/chemistry , Sesquiterpenes/pharmacology , Tacrine/chemistry , Tacrine/pharmacology , Acetylcholinesterase/metabolism , Alkaloids/toxicity , Alzheimer Disease/enzymology , Cholinesterase Inhibitors/toxicity , Cholinesterases/metabolism , Drug Discovery , Hep G2 Cells , Humans , Models, Molecular , Nootropic Agents/chemistry , Nootropic Agents/pharmacology , Nootropic Agents/toxicity , Sesquiterpenes/toxicity , Tacrine/toxicity
10.
ACS Chem Neurosci ; 4(9): 1267-77, 2013 Sep 18.
Article in English | MEDLINE | ID: mdl-23763493

ABSTRACT

ITH12246 (ethyl 5-amino-2-methyl-6,7,8,9-tetrahydrobenzo[b][1,8]naphthyridine-3-carboxylate) is a 1,8-naphthyridine described to feature an interesting neuroprotective profile in in vitro models of Alzheimer's disease. These effects were proposed to be due in part to a regulatory action on protein phosphatase 2A inhibition, as it prevented binding of its inhibitor okadaic acid. We decided to investigate the pharmacological properties of ITH12246, evaluating its ability to counteract the memory impairment evoked by scopolamine, a muscarinic antagonist described to promote memory loss, as well as to reduce the infarct volume in mice suffering phototrombosis. Prior to conducting these experiments, we confirmed its in vitro neuroprotective activity against both oxidative stress and Ca(2+) overload-derived excitotoxicity, using SH-SY5Y neuroblastoma cells and rat hippocampal slices. Using a predictive model of blood-brain barrier crossing, it seems that the passage of ITH12246 is not hindered. Its potential hepatotoxicity was observed only at very high concentrations, from 0.1 mM. ITH12246, at the concentration of 10 mg/kg i.p., was able to improve the memory index of mice treated with scopolamine, from 0.22 to 0.35, in a similar fashion to the well-known Alzheimer's disease drug galantamine 2.5 mg/kg. On the other hand, ITH12246, at the concentration of 2.5 mg/kg, reduced the phototrombosis-triggered infarct volume by 67%. In the same experimental conditions, 15 mg/kg melatonin, used as control standard, reduced the infarct volume by 30%. All of these findings allow us to consider ITH12246 as a new potential drug for the treatment of neurodegenerative diseases, which would act as a multifactorial neuroprotectant.


Subject(s)
Brain Ischemia/prevention & control , Cerebral Infarction/prevention & control , Memory Disorders/prevention & control , Naphthyridines/therapeutic use , Nerve Tissue Proteins/drug effects , Neuroprotective Agents/therapeutic use , Protein Phosphatase 2/drug effects , Animals , Blood-Brain Barrier , Calcium Signaling/drug effects , Cell Line , Cerebral Infarction/pathology , Chemical and Drug Induced Liver Injury/etiology , Disease Models, Animal , Drug Evaluation, Preclinical , Hippocampus/drug effects , Mice , Molecular Structure , Molecular Targeted Therapy , Naphthyridines/chemistry , Naphthyridines/pharmacology , Nerve Tissue Proteins/physiology , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Oligomycins/toxicity , Oxidative Stress/drug effects , Phosphorylation/drug effects , Protein Phosphatase 2/physiology , Protein Processing, Post-Translational/drug effects , Rats , Rotenone/toxicity , Scopolamine/antagonists & inhibitors , Scopolamine/toxicity , tau Proteins/metabolism
11.
Med Res Rev ; 33(1): 139-89, 2013 Jan.
Article in English | MEDLINE | ID: mdl-21793014

ABSTRACT

With 27 million cases worldwide documented in 2006, Alzheimer's disease (AD) constitutes an overwhelming health, social, economic, and political problem to nations. Unless a new medicine capable to delay disease progression is found, the number of cases will reach 107 million in 2050. So far, the therapeutic paradigm one-compound-one-target has failed. This could be due to the multiple pathogenic mechanisms involved in AD including amyloid ß (Aß) aggregation to form plaques, τ hyperphosphorylation to disrupt microtubule to form neurofibrillary tangles, calcium imbalance, enhanced oxidative stress, impaired mitochondrial function, apoptotic neuronal death, and deterioration of synaptic transmission, particularly at cholinergic neurons. Approximately 100 compounds are presently been investigated directed to single targets, namely inhibitors of ß and γ secretase, vaccines or antibodies that clear Aß, metal chelators to inhibit Aß aggregation, blockers of glycogen synthase kinase 3ß, enhancers of mitochondrial function, antioxidants, modulators of calcium-permeable channels such as voltage-dependent calcium channels, N-methyl-D-aspartate receptors for glutamate, or enhancers of cholinergic neurotransmission such as inhibitors of acetylcholinesterase or butyrylcholinesterase. In view of this complex pathogenic mechanisms, and the successful treatment of chronic diseases such as HIV or cancer, with multiple drugs having complementary mechanisms of action, the concern is growing that AD could better be treated with a single compound targeting two or more of the pathogenic mechanisms leading to neuronal death. This review summarizes the current therapeutic strategies based on the paradigm one-compound-various targets to treat AD. A treatment that delays disease onset and/or progression by 5 years could halve the number of people requiring institutionalization and/or dying from AD.


Subject(s)
Alzheimer Disease/drug therapy , Acetylcholinesterase/metabolism , Alzheimer Disease/enzymology , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Binding Sites , Butyrylcholinesterase/metabolism , Calcium Channels/metabolism , Disease Progression , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , Ligands , Receptors, N-Methyl-D-Aspartate , tau Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL