Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.186
Filter
1.
Molecules ; 29(9)2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38731472

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by the accumulation of amyloid beta (Aß) plaques in the brain. Aß1-42 is the main component of Aß plaque, which is toxic to neuronal cells. Si nanowires (Si NWs) have the advantages of small particle size, high specific surface area, and good biocompatibility, and have potential application prospects in suppressing Aß aggregation. In this study, we employed the vapor-liquid-solid (VLS) growth mechanism to grow Si NWs using Au nanoparticles as catalysts in a plasma-enhanced chemical vapor deposition (PECVD) system. Subsequently, these Si NWs were transferred to a phosphoric acid buffer solution (PBS). We found that Si NWs significantly reduced cell death in PC12 cells (rat adrenal pheochromocytoma cells) induced by Aß1-42 oligomers via double staining with 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and fluorescein diacetate/propyl iodide (FDA/PI). Most importantly, pre-incubated Si NWs largely prevented Aß1-42 oligomer-induced PC12 cell death, suggesting that Si NWs exerts an anti-Aß neuroprotective effect by inhibiting Aß aggregation. The analysis of Fourier Transform Infrared (FTIR) results demonstrates that Si NWs reduce the toxicity of fibrils and oligomers by intervening in the formation of ß-sheet structures, thereby protecting the viability of nerve cells. Our findings suggest that Si NWs may be a potential therapeutic agent for AD by protecting neuronal cells from the toxicity of Aß1-42.


Subject(s)
Amyloid beta-Peptides , Nanowires , Silicon , Amyloid beta-Peptides/toxicity , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Nanowires/chemistry , Animals , PC12 Cells , Rats , Silicon/chemistry , Peptide Fragments/chemistry , Peptide Fragments/toxicity , Peptide Fragments/pharmacology , Cell Survival/drug effects , Neuroprotective Agents/pharmacology , Neuroprotective Agents/chemistry , Protein Aggregates/drug effects , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism
2.
Eur J Med Chem ; 271: 116450, 2024 May 05.
Article in English | MEDLINE | ID: mdl-38701714

ABSTRACT

The complexity and multifaceted nature of Alzheimer's disease (AD) have driven us to further explore quinazoline scaffolds as multi-targeting agents for AD treatment. The lead optimization strategy was utilized in designing of new series of derivatives (AK-1 to AK-14) followed by synthesis, characterization, and pharmacological evaluation against human cholinesterase's (hChE) and ß-secretase (hBACE-1) enzymes. Amongst them, compounds AK-1, AK-2, and AK-3 showed good and significant inhibitory activity against both hAChE and hBACE-1 enzymes with favorable permeation across the blood-brain barrier. The most active compound AK-2 revealed significant propidium iodide (PI) displacement from the AChE-PAS region and was non-neurotoxic against SH-SY5Y cell lines. The lead molecule (AK-2) also showed Aß aggregation inhibition in a self- and AChE-induced Aß aggregation, Thioflavin-T assay. Further, compound AK-2 significantly ameliorated Aß-induced cognitive deficits in the Aß-induced Morris water maze rat model and demonstrated a significant rescue in eye phenotype in the Aꞵ-phenotypic drosophila model of AD. Ex-vivo immunohistochemistry (IHC) analysis on hippocampal rat brains showed reduced Aß and BACE-1 protein levels. Compound AK-2 suggested good oral absorption via pharmacokinetic studies and displayed a good and stable ligand-protein interaction in in-silico molecular modeling analysis. Thus, the compound AK-2 can be regarded as a lead molecule and should be investigated further for the treatment of AD.


Subject(s)
Acetylcholinesterase , Alzheimer Disease , Amyloid Precursor Protein Secretases , Amyloid beta-Peptides , Cholinesterase Inhibitors , Drug Design , Quinazolines , Quinazolines/pharmacology , Quinazolines/chemical synthesis , Quinazolines/chemistry , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Animals , Humans , Cholinesterase Inhibitors/pharmacology , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/chemistry , Acetylcholinesterase/metabolism , Rats , Structure-Activity Relationship , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Molecular Structure , Neuroprotective Agents/pharmacology , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/chemistry , Dose-Response Relationship, Drug , Butyrylcholinesterase/metabolism , Male
3.
Bioorg Med Chem Lett ; 107: 129788, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38740144

ABSTRACT

Effectively inhibition of amyloid ß (Aß) aggregation is considered an important method for treatment of the Alzheimer's disease. Herein, inspired by the ability of trans-clovamide to effectively inhibit Aß aggregation, we synthesized a series of structurally related catecholamine derivatives and tested them as Aß aggregation inhibitors using the Thioflavin T assay. The results show that they demonstrated a higher inhibitory rate against Aß aggregation. Furthermore, these compounds exhibited high water solubilities and low cytotoxicities. Additionally, transmission electron microscopy images and dynamic light scattering of their Aß aggregations were observed. Docking simulations revealed that the catechol moiety of the synthesized compounds can form hydrogen bonds with the key regions of Aß and thereby inhibit Aß aggregation.


Subject(s)
Amyloid beta-Peptides , Catecholamines , Molecular Docking Simulation , Protein Aggregates , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Catecholamines/metabolism , Humans , Protein Aggregates/drug effects , Structure-Activity Relationship , Molecular Structure , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Dose-Response Relationship, Drug
4.
Inorg Chem ; 63(21): 10001-10010, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38742626

ABSTRACT

There is a growing interest in the search for metal-based therapeutics for protein misfolding disorders such as Alzheimer's disease (AD). A novel and largely unexplored class of metallodrugs is constituted by paddlewheel diruthenium complexes, which exhibit unusual water solubility and stability and unique coordination modes to proteins. Here, we investigate the ability of the complexes [Ru2Cl(DPhF)(O2CCH3)3]·H2O (1), [Ru2Cl(DPhF)2(O2CCH3)2]·H2O (2), and K2[Ru2(DPhF)(CO3)3]·3H2O (3) (DPhF- = N,N'-diphenylformamidinate) to interfere with the amyloid aggregation of the Aß1-42 peptide. These compounds differ in charge and steric hindrance due to the coordination of a different number of bulky ligands. The mechanisms of action of the three complexes were studied by employing a plethora of physicochemical and biophysical techniques as well as cellular assays. All these studies converge on different mechanisms of inhibition of amyloid fibrillation: complexes 1 and 2 show a clear inhibitory effect due to an exchange ligand process in the Ru2 unit aided by aromatic interactions. Complex 3 shows no inhibition of aggregation, probably due to its negative charge in solution. This study demonstrates that slight variations in the ligands surrounding the bimetallic core can modulate the amyloid aggregation inhibition and supports the use of paddlewheel diruthenium complexes as promising therapeutics for Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides , Coordination Complexes , Peptide Fragments , Ruthenium , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/chemistry , Ruthenium/chemistry , Ruthenium/pharmacology , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Coordination Complexes/chemical synthesis , Peptide Fragments/chemistry , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/metabolism , Humans , Protein Aggregates/drug effects , Molecular Structure , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism
5.
Eur J Med Chem ; 272: 116463, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38704944

ABSTRACT

Butyrylcholinesterase (BChE) has attracted wide interest as a promising target in Alzheimer's disease (AD) investigation. BChE is considered to play a compensable role of hydrolyzing acetylcholine (ACh), and its positive correlation with ß-amyloid (Aß) deposition also promotes disease progression. Herein, we uncovered a selective potent BChE inhibitor S21-1011 (eqBChE IC50 = 0.059 ± 0.006 µM, hBChE IC50 = 0.162 ± 0.069 µM), which presented satisfactory druggability and therapeutic efficacy in AD models. In pharmacokinetics (PK) studies, S21-1011 showed excellent blood-brain barrier (BBB) permeability, metabolism stability and high oral-bioavailability. In pharmacodynamic (PD) studies, it protected neural cells from toxicity and inflammation stimulation in vitro. Besides, it also exerted anti-inflammatory effect and alleviated cognitive impairment in mice models induced by lipopolysaccharides (LPS) and Aß. Generally, this compound has been confirmed to function as a neuroprotector and cognition improver in various AD pathology-like models. Therefore, S21-1011, a novel potent BChE inhibitor, could be considered as a potential anti-AD candidate worthy of more profound investigation.


Subject(s)
Alzheimer Disease , Butyrylcholinesterase , Cholinesterase Inhibitors , Quinolines , Butyrylcholinesterase/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Animals , Cholinesterase Inhibitors/pharmacology , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/chemical synthesis , Mice , Humans , Structure-Activity Relationship , Quinolines/chemistry , Quinolines/pharmacology , Quinolines/chemical synthesis , Drug Discovery , Molecular Structure , Male , Lipopolysaccharides/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Dose-Response Relationship, Drug , Neuroprotective Agents/pharmacology , Neuroprotective Agents/chemistry , Neuroprotective Agents/chemical synthesis , Piperazines/pharmacology , Piperazines/chemistry , Piperazines/chemical synthesis , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/chemical synthesis , Inflammation/drug therapy , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects
6.
Int J Nanomedicine ; 19: 4299-4317, 2024.
Article in English | MEDLINE | ID: mdl-38766654

ABSTRACT

Background: Inhibition of amyloid ß protein fragment (Aß) aggregation is considered to be one of the most effective strategies for the treatment of Alzheimer's disease. (-)-Epigallocatechin-3-gallate (EGCG) has been found to be effective in this regard; however, owing to its low bioavailability, nanodelivery is recommended for practical applications. Compared to chemical reduction methods, biosynthesis avoids possible biotoxicity and cumbersome preparation processes. Materials and Methods: The interaction between EGCG and Aß42 was simulated by molecular docking, and green tea-conjugated gold nanoparticles (GT-Au NPs) and EGCG-Au NPs were synthesized using EGCG-enriched green tea and EGCG solutions, respectively. Surface active molecules of the particles were identified and analyzed using various liquid chromatography-tandem triple quadrupole mass spectrometry methods. ThT fluorescence assay, circular dichroism, and TEM were used to investigate the effect of synthesized particles on the inhibition of Aß42 aggregation. Results: EGCG as well as apigenin, quercetin, baicalin, and glutathione were identified as capping ligands stabilized on the surface of GT-Au NPs. They more or less inhibited Aß42 aggregation or promoted fibril disaggregation, with EGCG being the most effective, which bound to Aß42 through hydrogen bonding, hydrophobic interactions, etc. resulting in 39.86% and 88.50% inhibition of aggregation and disaggregation effects, respectively. EGCG-Au NPs were not as effective as free EGCG, whereas multiple thiols and polyphenols in green tea accelerated and optimized heavy metal detoxification. The synthesized GT-Au NPs conferred the efficacy of diverse ligands to the particles, with inhibition of aggregation and disaggregation effects of 54.69% and 88.75%, respectively, while increasing the yield, enhancing water solubility, and decreasing cost. Conclusion: Biosynthesis of nanoparticles using green tea is a promising simple and economical drug-carrying approach to confer multiple pharmacophore molecules to Au NPs. This could be used to design new drug candidates to treat Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides , Catechin , Gold , Metal Nanoparticles , Molecular Docking Simulation , Peptide Fragments , Tea , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Catechin/chemistry , Catechin/pharmacology , Catechin/analogs & derivatives , Tea/chemistry , Metal Nanoparticles/chemistry , Metal Nanoparticles/administration & dosage , Gold/chemistry , Ligands , Peptide Fragments/chemistry , Peptide Fragments/antagonists & inhibitors , Humans , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Protein Aggregates/drug effects
8.
Chem Biol Drug Des ; 103(4): e14529, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38670598

ABSTRACT

With the increasing aging population, rational design of drugs for Alzheimer's disease (AD) treatment has become an important research area. Based on the multifunctional design strategy, four diosmetin derivatives (1-4) were designed, synthesized, and characterized by 1H NMR, 13C NMR, and MS. Docking study was firstly applied to substantiate the design strategies and then the biological activities including cholinesterase inhibition, metal chelation, antioxidation and ß-amyloid (Aß) aggregation inhibition in vitro were evaluated. The results showed that 1-4 had good acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE) inhibition, metal chelation (selective chelation of Cu2+ ions), antioxidation, self-induced, Cu2+-induced, and AChE-induced Aß aggregation inhibition activities, and suitable blood-brain barrier (BBB) permeability. Especially, compound 3 had the strongest inhibitory effect on AChE (10-8 M magnitude) and BuChE (10-7 M magnitude) and showed the best inhibition on AChE-induced Aß aggregation with 66.14% inhibition ratio. Furthermore, compound 3 could also reduce intracellular reactive oxygen species (ROS) levels in Caenorhabditis elegans and had lower cytotoxicity. In summary, 3 might be considered as a potential multifunctional anti-AD ligand.


Subject(s)
Acetylcholinesterase , Alzheimer Disease , Amyloid beta-Peptides , Blood-Brain Barrier , Butyrylcholinesterase , Caenorhabditis elegans , Cholinesterase Inhibitors , Drug Design , Flavonoids , Molecular Docking Simulation , Reactive Oxygen Species , Cholinesterase Inhibitors/pharmacology , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/metabolism , Acetylcholinesterase/metabolism , Animals , Butyrylcholinesterase/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/metabolism , Ligands , Blood-Brain Barrier/metabolism , Humans , Reactive Oxygen Species/metabolism , Flavonoids/chemistry , Flavonoids/pharmacology , Flavonoids/chemical synthesis , Antioxidants/pharmacology , Antioxidants/chemistry , Antioxidants/chemical synthesis , Structure-Activity Relationship , Protein Aggregates/drug effects
9.
Bioorg Med Chem ; 105: 117715, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38615460

ABSTRACT

Amyloid-ß (Aß) oligomers are a cause of neurodegeneration in Alzheimer's disease (AD). These soluble aggregates of the Aß peptide have proven difficult to study due to their inherent metastability and heterogeneity. Strategies to isolate and stabilize homogenous Aß oligomer populations have emerged such as mutations, covalent cross-linking, and protein fusions. These strategies along with molecular dynamics simulations have provided a variety of proposed structures of Aß oligomers, many of which consist of molecules of Aß in ß-hairpin conformations. ß-Hairpins are intramolecular antiparallel ß-sheets composed of two ß-strands connected by a loop or turn. Three decades of research suggests that Aß peptides form several different ß-hairpin conformations, some of which are building blocks of toxic Aß oligomers. The insights from these studies are currently being used to design anti-Aß antibodies and vaccines to treat AD. Research suggests that antibody therapies designed to target oligomeric Aß may be more successful at treating AD than antibodies designed to target linear epitopes of Aß or fibrillar Aß. Aß ß-hairpins are good epitopes to use in antibody development to selectively target oligomeric Aß. This review summarizes the research on ß-hairpins in Aß peptides and discusses the relevance of this conformation in AD pathogenesis and drug development.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Alzheimer Disease/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/chemistry , Humans
10.
Bioorg Chem ; 147: 107373, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38653149

ABSTRACT

The escalating prevalence of Alzheimer's disease (AD) has prompted extensive research into potential therapeutic interventions, with a specific focus on molecular targets such as amyloid beta (Aß) and tau protein aggregation. In this study, a series of α-ketoamide derivatives was synthesized from ß,γ-unsaturated α-keto thioesters, achieving high purity and good yield. Thioflavin T based Aß aggregation assay identified four promising compounds (BD19, BD23, BD24, and BD27) that demonstrated significant inhibitory effects on Aß aggregation. BD23, selected for its better solubility (0.045 ± 0.0012 mg/ml), was further subjected to in vitro Parallel Artificial Membrane Permeability Assay to determine the Blood-Brain-Barrier permeability and emerged as BBB permeable with permeability rate (Pe) of 10.66 ± 8.11 × 10-6 cm/s. In addition to its Aß inhibitory properties, BD23 exhibited significant inhibition of heparin-induced tau aggregation and demonstrated non-toxicity in SHSY5Y cell lines. Subsequent in vivo assays were conducted, administering compound BD23 to an Aß induced mouse model of AD at various doses (1, 2, & 5 mg/kg). The results revealed a noteworthy enhancement in cognitive functions, particularly when BD23 was administered at a dosage of 5 mg/kg, comparable to the effects observed with the standard dose of Donepezil (DNP). In silico investigations, including molecular docking, molecular dynamics simulations, and Density Functional Theory calculations provided insights into BD23's interactions with the targets and electronic properties. These analyses contribute to the understanding of the therapeutic potential of the lead compounds BD23 which further pave the way for further exploration of its therapeutic potential in the context of AD.


Subject(s)
Alzheimer Disease , Amides , Amyloid beta-Peptides , Dose-Response Relationship, Drug , Protein Aggregates , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Humans , Animals , Mice , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Amides/chemistry , Amides/pharmacology , Amides/chemical synthesis , Protein Aggregates/drug effects , Structure-Activity Relationship , Molecular Structure , Molecular Docking Simulation , tau Proteins/metabolism , tau Proteins/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/chemistry , Male
11.
Phytochemistry ; 222: 114098, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38648960

ABSTRACT

Nine undescribed compounds, along with eight known compounds, were isolated from the stipes of Lentinus edodes. Their structures were established by extensive spectroscopic and circular dichroism analyses. The protective effects against Aß25-35-induced N9 microglia cells injury of these compounds were tested by MTT method, and the levels of apoptosis and ROS were detected by flow cytometry. In addition, the binding sites and interactions of compound with amyloid precursor protein were revealed using molecular docking simulations. These findings further establish the structural diversity and bioactivity of stipes of L. edodes, and provide an experimental basis for targeting Alzheimer's disease as a potential strategy.


Subject(s)
Amyloid beta-Peptides , Apoptosis , Microglia , Molecular Docking Simulation , Peptide Fragments , Microglia/drug effects , Microglia/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Peptide Fragments/pharmacology , Animals , Apoptosis/drug effects , Mice , Molecular Structure , Neuroprotective Agents/pharmacology , Neuroprotective Agents/chemistry , Neuroprotective Agents/isolation & purification , Reactive Oxygen Species/metabolism , Structure-Activity Relationship , Dose-Response Relationship, Drug , Lentinula/chemistry , Cell Line
12.
Colloids Surf B Biointerfaces ; 238: 113907, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38608464

ABSTRACT

Multifunctional agents with therapeutic and diagnostic capabilities are imperative to the prevention of Alzheimer's disease (AD), which is considered due to abnormal aggregation and deposition of ß-amyloid protein (Aß) as well as oxidative stress. Herein, quercetin (Que)- and p-phenylenediamine (p-PD)-derived red emission carbon dots (CDs) synthesized via a one-step hydrothermal method were designed as a novel theranostic nano-agent for the multi-target treatment of AD. R-CD-75 with an optimized composition exhibited significant inhibition of Aß aggregation and rapid depolymerization of mature Aß fibrils (<4 h) at micromolar concentrations (2 and 5 µg/mL, respectively). Moreover, R-CD-75 potently scavenged reactive oxygen species and showed turned-on red fluorescence imaging of Aß plaques both in vitro and in vivo. In vitro assays proved that R-CD-75 significantly mitigated the Aß-induced cytotoxicity and enhanced the cultured cell viability from 74.9 % to 98.0 %, while in vivo studies demonstrated that R-CD-75 prolonged the lifespan of AD nematodes by over 50 % (from 13 to 20 d). Compared to the precursors Que and p-PD, R-CD-75 inherited some of their structures and functional groups, such as aromatic structures, phenolic hydroxyl and amino groups, which were considered to interact with Aß species through hydrogen bonding, electrostatic interactions, hydrophobic interactions, and π-π stacking, thus contributing to its effectiveness in its theranostic functions. This research has opened a new avenue to the development of potent theranostic agents by designing novel carbon dots.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Carbon , Quantum Dots , Quercetin , Theranostic Nanomedicine , Quercetin/chemistry , Quercetin/pharmacology , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/chemistry , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , Carbon/chemistry , Carbon/pharmacology , Quantum Dots/chemistry , Animals , Humans , Cell Survival/drug effects , Reactive Oxygen Species/metabolism , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/metabolism , Particle Size
13.
Eur J Med Chem ; 271: 116409, 2024 May 05.
Article in English | MEDLINE | ID: mdl-38663285

ABSTRACT

Inspite of established symptomatic relief drug targets, a multi targeting approach is highly in demand to cure Alzheimer's disease (AD). Simultaneous inhibition of cholinesterase (ChE), ß secretase-1 (BACE-1) and Dyrk1A could be promising in complete cure of AD. A series of 18 diaryl triazine based molecular hybrids were successfully designed, synthesized, and tested for their hChE, hBACE-1, Dyrk1A and Aß aggregation inhibitory potentials. Compounds S-11 and S-12 were the representative molecules amongst the series with multi-targeted inhibitory effects. Compound S-12 showed hAChE inhibition (IC50 value = 0.486 ± 0.047 µM), BACE-1 inhibition (IC50 value = 0.542 ± 0.099 µM) along with good anti-Aß aggregation effects in thioflavin-T assay. Only compound S-02 of the series has shown Dyrk1A inhibition (IC50 value = 2.000 ± 0.360 µM). Compound S-12 has also demonstrated no neurotoxic liabilities against SH-SY5Y as compared to donepezil. The in vivo behavioral studies of the compound S-12 in the scopolamine- and Aß-induced animal models also demonstrated attanuation of learning and memory functions in rats models having AD-like characteristics. The ex vivo studies, on the rat hippocampal brain demonstrated reduction in certain biochemical markers of the AD brain with a significant increase in ACh level. The Western blot and Immunohistochemistry further revealed lower tau, APP and BACE-1 molecular levels. The drosophilla AD model also revealed improved eyephenotype after treatment with compound S-12. The molecular docking studies of the compounds suggested that compound S-12 was interacting with the ChE-PAS & CAS residues and catalytic dyad residues of the BACE-1 enzymes. The 100 ns molecular dynamics simulation studies of the ligand-protein complexed with hAChE and hBACE-1 also suggested stable ligand-protein confirmation throughout the simulation run.


Subject(s)
Acetylcholinesterase , Alzheimer Disease , Amyloid Precursor Protein Secretases , Cholinesterase Inhibitors , Drug Design , Triazines , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Animals , Humans , Cholinesterase Inhibitors/pharmacology , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/chemistry , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Rats , Structure-Activity Relationship , Acetylcholinesterase/metabolism , Triazines/chemistry , Triazines/pharmacology , Triazines/chemical synthesis , Aspartic Acid Endopeptidases/antagonists & inhibitors , Aspartic Acid Endopeptidases/metabolism , Molecular Structure , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Molecular Docking Simulation , Dyrk Kinases , Dose-Response Relationship, Drug , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Male , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Butyrylcholinesterase/metabolism
14.
Arch Pharm (Weinheim) ; 357(5): e2300693, 2024 May.
Article in English | MEDLINE | ID: mdl-38332316

ABSTRACT

Aß1-42 and acetylcholinesterase (AChE) are two key therapeutic targets for Alzheimer's disease (AD). The purpose of this study is to develop a dual-target inhibitor that inhibits both of these targets by fusing the chemical structure of baicalein and donepezil. Among them, we modified the structure of baicalein to arylcoumarin, synthesized three kinds of structural compounds, and evaluated their biological activities. The results showed that compound 3b had the strongest inhibitory effect on AChE (IC50 = 0.05 ± 0.02 µM), which was better than those of donepezil and baicalein. In addition, compound 3b has a strong ability to inhibit the aggregation of Aß1-42 and protect nerve cells, and it can also penetrate the blood-brain barrier well. Using a zebrafish behavioral analyzer test, it was found that compound 3b can alleviate the behavioral effects of AlCl3-induced zebrafish larval movement retardation, which has a certain guiding significance for simulating the movement disorders of AD patients. In summary, compound 3b is expected to become a multifunctional agent for treating and alleviating the symptoms of AD patients.


Subject(s)
Acetylcholinesterase , Alzheimer Disease , Amyloid beta-Peptides , Cholinesterase Inhibitors , Drug Design , Zebrafish , Alzheimer Disease/drug therapy , Animals , Cholinesterase Inhibitors/pharmacology , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/chemistry , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Structure-Activity Relationship , Acetylcholinesterase/metabolism , Humans , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/pharmacology , Donepezil/pharmacology , Donepezil/chemical synthesis , Donepezil/chemistry , Blood-Brain Barrier/metabolism , Molecular Structure , Flavanones/pharmacology , Flavanones/chemical synthesis , Flavanones/chemistry , Dose-Response Relationship, Drug , Behavior, Animal/drug effects
15.
Cent Nerv Syst Agents Med Chem ; 24(1): 45-56, 2024.
Article in English | MEDLINE | ID: mdl-38305393

ABSTRACT

Alzheimer's disease (AD) is an onset and incurable neurodegenerative disorder that has been linked to various genetic, environmental, and lifestyle factors. Recent research has revealed several potential targets for drug development, such as the prevention of Aß production and removal, prevention of tau hyperphosphorylation, and keeping neurons alive. Drugs that target numerous ADrelated variables have been developed, and early results are encouraging. This review provides a concise map of the different receptor signaling pathways associated with Alzheimer's Disease, as well as insight into drug design based on these pathways. It discusses the molecular mechanisms of AD pathogenesis, such as oxidative stress, aging, Aß turnover, thiol groups, and mitochondrial activities, and their role in the disease. It also reviews the potential drug targets, in vivo active agents, and docking studies done in AD and provides prospects for future drug development. This review intends to provide more clarity on the molecular processes that occur in Alzheimer's patient's brains, which can be of use in diagnosing and preventing the condition.


Subject(s)
Alzheimer Disease , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Humans , Animals , Oxidative Stress/drug effects , Oxidative Stress/physiology , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Molecular Targeted Therapy/methods , tau Proteins/metabolism , tau Proteins/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/physiology
16.
Arch Pharm (Weinheim) ; 357(5): e2300603, 2024 May.
Article in English | MEDLINE | ID: mdl-38290060

ABSTRACT

Alzheimer's disease (AD) is a multifactorial neurological disease, and the multitarget directed ligand (MTDL) strategy may be an effective approach to delay its progression. Based on this strategy, 27 derivatives of l-tryptophan, 3a-1-3d-1, were designed, synthesized, and evaluated for their biological activity. Among them, IC50 (inhibitor concentration resulting in 50% inhibitory activity) values of compounds 3a-18 and 3b-1 were 0.58 and 0.44 µM for human serum butyrylcholinesterase (hBuChE), respectively, and both of them exhibited more than 30-fold selectivity for human serum acetylcholinesterase. Enzyme kinetics studies showed that these two compounds were mixed inhibitors of hBuChE. In addition, these two derivatives possessed extraordinary antioxidant activity in OH radical scavenging and oxygen radical absorption capacity fluorescein assays. Meanwhile, these compounds could also prevent ß-amyloid (Aß) self-aggregation and possessed low toxicity on PC12 and AML12 cells. Molecular modeling studies revealed that these two compounds could interact with the choline binding site, acetyl binding site, and peripheral anionic site to exert submicromolar BuChE inhibitory activity. In the vitro blood-brain barrier permeation assay, compounds 3a-18 and 3b-1 showed enough blood-brain barrier permeability. In drug-likeness prediction, compounds 3a-18 and 3b-1 showed good gastrointestinal absorption and a low risk of human ether-a-go-go-related gene toxicity. Therefore, compounds 3a-18 and 3b-1 are potential multitarget anti-AD lead compounds, which could work as powerful antioxidants with submicromolar selective inhibitory activity for hBuChE as well as prevent Aß self-aggregation.


Subject(s)
Acetylcholinesterase , Alzheimer Disease , Amyloid beta-Peptides , Antioxidants , Blood-Brain Barrier , Butyrylcholinesterase , Cholinesterase Inhibitors , Drug Design , Tryptophan , Alzheimer Disease/drug therapy , Humans , Antioxidants/pharmacology , Antioxidants/chemical synthesis , Antioxidants/chemistry , Cholinesterase Inhibitors/pharmacology , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/chemistry , Structure-Activity Relationship , Blood-Brain Barrier/metabolism , Butyrylcholinesterase/metabolism , Animals , Tryptophan/pharmacology , Tryptophan/chemistry , Tryptophan/analogs & derivatives , Tryptophan/chemical synthesis , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Rats , Acetylcholinesterase/metabolism , Molecular Structure , PC12 Cells , Dose-Response Relationship, Drug , Models, Molecular
17.
Int J Mol Sci ; 24(14)2023 Jul 11.
Article in English | MEDLINE | ID: mdl-37511086

ABSTRACT

AD is the most common neurodegenerative disorder characterized by progressive memory impairment and cognitive deficits. The pathology of AD is still unclear; however, several studies have shown that the aggregation of the Aß peptide in the CNS is an exclusively pathological process involved in AD. Currently, there is no proven medication to cure or prevent the disease progression. Nevertheless, various therapeutic approaches for AD show only relief of symptoms and mostly work on cognitive recovery. However, one of the promising approaches for therapeutic intervention is to use inhibitors for blocking the Aß peptide aggregation process. Recently, herbal phenolic compounds have been shown to have a therapeutic property for treatment of AD due to their multifaceted action. In this study, we investigated the effectiveness of SA, Gn Rb1, and DMyr on inhibiting the aggregation and toxicity of Aß40 and Aß42 using different biochemical and cell-based assays. Our results showed that SA and DMyr inhibit Aß40 and Aß42 fibrillation, seeded aggregation, and toxicity. Gn Rb1 did not have any effect on the aggregation or toxicity induced by Aß40 and Aß42. Moreover, SA and DMyr were able to disaggregate the preformed fibrils. Overall, these compounds may be used alone or synergistically and could be considered as a lead for designing new compounds that could be used as effective treatment of AD and related disorders.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Cognitive Dysfunction , Peptide Fragments , Humans , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Cognitive Dysfunction/drug therapy , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/metabolism
19.
Biomed Pharmacother ; 165: 115026, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37336148

ABSTRACT

This study sought to investigate the anti-amyloid ß (Aß) and anti-neuroinflammatory effects of catalpol in an Alzheimer's disease (AD) mouse model. METHODS: The effects of catalpol on Aß formation were investigated by thioflavin T assay. The effect of catalpol on generating inflammatory cytokines from microglial cells and the cytotoxicity of microglial cells on HT22 hippocampal cells were assessed by real-time quantitative PCR, ELISA, redox reactions, and cell viability. APPswe/PS1ΔE9 mice were treated with catalpol, and their cognitive ability was investigated using the water maze and novel object recognition tests. Immunohistochemistry and immunofluorescence were used to probe for protein markers of microglia and astrocyte, Aß deposits, and NF-κB pathway activity. Aß peptides, neuroinflammation, and nitric oxide production were examined using ELISA and redox reactions. RESULTS: Catalpol potently inhibited Aß fibril and oligomer formation. In microglial cells stimulated by Aß, catalpol alleviated the expression of the proinflammatory cytokines tumor necrosis factor-α (TNF-α), interleukin (IL)-6, and inducible nitric oxide synthase (iNOS) but promoted the expression of the anti-inflammatory cytokine IL-10. Catalpol alleviated the cytotoxic effects of Aß-exposed microglia on HT22 cells. Treatment with catalpol in APPswe/PS1ΔE9 mice downregulated neuroinflammation production, decreased Aß deposits in the brains and alleviated cognitive impairment. Catalpol treatment decreased the number of IBA-positive microglia and GFAP-positive astrocytes and their activities of the NF-κB pathway in the hippocampus of APPswe/PS1ΔE9 mice. CONCLUSION: The administration of catalpol protected neurons by preventing neuroinflammation and Aß deposits in an AD mouse model. Therefore, catalpol may be a promising strategy for treating AD.


Subject(s)
Amyloid beta-Peptides , Cognitive Dysfunction , Drugs, Chinese Herbal , Iridoid Glucosides , Neuroinflammatory Diseases , Neuroprotective Agents , Plaque, Amyloid , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Iridoid Glucosides/pharmacology , Iridoid Glucosides/therapeutic use , Cognitive Dysfunction/drug therapy , Plaque, Amyloid/drug therapy , Neuroinflammatory Diseases/drug therapy , Animals , Mice , Disease Models, Animal , Cytokines/metabolism , Cell Line , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Amyloid beta-Peptides/antagonists & inhibitors , Mice, Inbred C57BL , Male , Female , Mice, Transgenic
20.
J Nat Med ; 77(3): 508-515, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36933089

ABSTRACT

Amyloid ß (Aß) is thought to be involved in the pathogenesis of Alzheimer's disease (AD). Aß aggregation in the brain is considered the cause of AD. Therefore, inhibiting Aß aggregation and degrading existing Aß aggregates is a promising approach for the treatment and prevention of the disease. In searching for inhibitors of Aß42 aggregation, we found that meroterpenoids isolated from Sargassum macrocarpum possess potent inhibitory activities. Therefore, we searched for active compounds from this brown alga and isolated 16 meroterpenoids, which contain three new compounds. The structures of these new compounds were elucidated using two-dimensional nuclear magnetic resonance techniques. Thioflavin-T assay and transmission electron microscopy were used to reveal the inhibitory activity of these compounds against Aß42 aggregation. All the isolated meroterpenoids were found to be active, and compounds with a hydroquinone structure tended to have stronger activity than those with a quinone structure.


Subject(s)
Alzheimer Disease , Sargassum , Terpenes , Humans , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/antagonists & inhibitors , Sargassum/chemistry , Terpenes/chemistry , Terpenes/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...