Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(6)2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38542228

ABSTRACT

Recently, we identified a novel mechanism of enzyme inhibition in N-myristoyltransferases (NMTs), which we have named 'inhibitor trapping'. Inhibitor trapping occurs when the protein captures the small molecule within its structural confines, thereby preventing its free dissociation and resulting in a dramatic increase in inhibitor affinity and potency. Here, we demonstrate that inhibitor trapping also occurs in the kinases. Remarkably, the drug imatinib, which has revolutionized targeted cancer therapy, is entrapped in the structure of the Abl kinase. This effect is also observed in p38α kinase, where inhibitor trapping was found to depend on a 'magic' methyl group, which stabilizes the protein conformation and increases the affinity of the compound dramatically. Altogether, these results suggest that inhibitor trapping is not exclusive to N-myristoyltransferases, as it also occurs in the kinase family. Inhibitor trapping could enhance the binding affinity of an inhibitor by thousands of times and is as a key mechanism that plays a critical role in determining drug affinity and potency.


Subject(s)
Piperazines , Pyrimidines , Pyrimidines/pharmacology , Piperazines/pharmacology , Benzamides/pharmacology , Imatinib Mesylate/pharmacology , Fusion Proteins, bcr-abl/metabolism , src-Family Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use
2.
Molecules ; 29(2)2024 Jan 05.
Article in English | MEDLINE | ID: mdl-38257197

ABSTRACT

Peptide-protein interactions form a cornerstone in molecular biology, governing cellular signaling, structure, and enzymatic activities in living organisms. Improving computational models and experimental techniques to describe and predict these interactions remains an ongoing area of research. Here, we present a computational method for peptide-protein interactions' description and prediction based on leveraged amino acid frequencies within specific binding cores. Utilizing normalized frequencies, we construct quantitative matrices (QMs), termed 'logo models' derived from sequence logos. The method was developed to predict peptide binding to HLA-DQ2.5 and HLA-DQ8.1 proteins associated with susceptibility to celiac disease. The models were validated by more than 17,000 peptides demonstrating their efficacy in discriminating between binding and non-binding peptides. The logo method could be applied to diverse peptide-protein interactions, offering a versatile tool for predictive analysis in molecular binding studies.


Subject(s)
Celiac Disease , Peptides , Humans , Amino Acids , Molecular Biology , Position-Specific Scoring Matrices
3.
Chem Biol Interact ; 386: 110772, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37898285

ABSTRACT

Having continued our recent study on the synthesis and DNase I inhibition of several monosquaramides, two new chloro-substituted pyridine squaramates were synthesized and their structure was identified by X-ray. Their inhibitory properties towards deoxyribonuclease I (DNase I) and xanthine oxidase (XO) were evaluated in vitro. 3-(((6-Chloropyridin-3-yl)methyl)amino)-4-ethoxycyclobut-3-ene-1,2-dione (compound 3a) inhibited DNase I with an IC50 value of 43.82 ± 6.51 µM, thus standing out as one of the most potent small organic DNase I inhibitors tested to date. No cytotoxicity to human tumor cell lines (HL-60, MDA-MB-231 and MCF-7) was observed for the tested compounds. In order to investigate the drug-likeness of the squaramates, the ADME profile and pharmacokinetic properties were evaluated. Molecular docking was performed to reveal the binding mode of the studied compounds on DNase I.


Subject(s)
Deoxyribonuclease I , Pyridines , Humans , Molecular Docking Simulation , Structure-Activity Relationship , Cell Line, Tumor , Pyridines/pharmacology , Deoxyribonuclease I/metabolism , Molecular Structure , Enzyme Inhibitors/chemistry
4.
Int J Mol Sci ; 24(14)2023 Jul 18.
Article in English | MEDLINE | ID: mdl-37511367

ABSTRACT

Predicting inhibitor potency is critical in drug design and development, yet it has remained one of computational biology's biggest unresolved challenges. Here, we show that in the case of the N-myristoyltransferase (NMT), this problem could be traced to the mechanisms by which the NMT enzyme is inhibited. NMT adopts open or closed conformations necessary for orchestrating the different steps of the catalytic process. The results indicate that the potency of the NMT inhibitors is determined by their ability to stabilize the enzyme conformation in the closed state, and that in this state, the small molecules themselves are trapped and locked inside the structure of the enzyme, creating a significant barrier for their dissociation. By using molecular dynamics simulations, we demonstrate that the conformational stabilization of the protein molecule in its closed form is highly correlated with the ligands activity and can be used to predict their potency. Hence, predicting inhibitor potency in silico might depend on modeling the conformational changes of the protein molecule upon binding of the ligand rather than estimating the changes in free binding energy that arise from their interaction.


Subject(s)
Acyltransferases , Molecular Dynamics Simulation , Acyltransferases/metabolism
5.
Methods Mol Biol ; 2673: 237-249, 2023.
Article in English | MEDLINE | ID: mdl-37258919

ABSTRACT

Major histocompatibility complex (MHC) proteins are the most polymorphic and polygenic proteins in humans. They bind peptides, derived from cleavage of different pathogenic antigens, and are responsible for presenting them to T cells. The peptides recognized by the T cell receptors are denoted as epitopes and they trigger an immune response.In this chapter, we describe a docking protocol for predicting the peptide binding to a given MHC protein using the software tool GOLD. The protocol starts with the construction of a combinatorial peptide library used in the docking and ends with the derivation of a quantitative matrix (QM) accounting for the contribution of each amino acid at each peptide position.


Subject(s)
Peptide Library , Peptides , Humans , Peptides/chemistry , Epitopes/metabolism , Software , Protein Binding
6.
Chem Biol Interact ; 381: 110542, 2023 Aug 25.
Article in English | MEDLINE | ID: mdl-37224992

ABSTRACT

A library of 43 thiazole derivatives, including 31 previously and 12 newly synthesized in the present study, was evaluated in vitro for their inhibitory properties against bovine pancreatic DNase I. Nine compounds (including three newly synthesized) inhibited the enzyme showing improved inhibitory properties compared to that of the reference crystal violet (IC50 = 346.39 µM). Two compounds (5 and 29) stood out as the most potent DNase I inhibitors, with IC50 values below 100 µM. The 5-LO inhibitory properties of the investigated derivatives were also analyzed due to the importance of this enzyme in the development of neurodegenerative diseases. Compounds (12 and 29) proved to be the most prominent new 5-LO inhibitors, with IC50 values of 60 nM and 56 nM, respectively, in cell-free assay. Four compounds, including one previously (41) and three newly (12, 29 and 30) synthesized, have the ability to inhibit DNase I with IC50 values below 200 µM and 5-LO with IC50 values below 150 nM in cell-free assay. Molecular docking and molecular dynamics simulations were used to clarify DNase I and 5-LO inhibitory properties of the most potent representatives at the molecular level. The newly synthesized compound 29 (4-((4-(3-bromo-4-morpholinophenyl)thiazol-2-yl)amino)phenol) represents the most promising dual DNase I and 5-LO inhibitor, as it inhibited 5-LO in the nanomolar and DNase I in the double-digit micromolar concentration ranges. The results obtained in the present study, together with our recently published results for 4-(4-chlorophenyl)thiazol-2-amines, represent a good basis for the development of new neuroprotective therapeutics based on dual inhibition of DNase I and 5-LO.


Subject(s)
Neuroprotective Agents , Thiazoles , Animals , Cattle , Structure-Activity Relationship , Thiazoles/pharmacology , Thiazoles/chemistry , Molecular Docking Simulation , Neuroprotective Agents/pharmacology , Arachidonate 5-Lipoxygenase , Deoxyribonuclease I , Lipoxygenase Inhibitors/pharmacology , Molecular Structure
7.
Molecules ; 28(2)2023 Jan 05.
Article in English | MEDLINE | ID: mdl-36677597

ABSTRACT

Three new monosquaramides (3a-c) were synthesized, characterized by IR, NMR and X-ray, and evaluated for inhibitory activity against deoxyribonuclease I (DNase I) and xanthine oxidase (XO) in vitro. The target compounds inhibited DNase I with IC50 values below 100 µM, being at the same time more potent DNase I inhibitors than crystal violet, used as a positive control. 3-Ethoxy-4-((1-(pyridin-3-yl)propan-2-yl)amino)cyclobut-3-ene-1,2-dione (3c) stood out as the most potent compound, exhibiting a slightly better IC50 value (48.04 ± 7.98 µM) compared to the other two compounds. In order to analyze potential binding sites for the studied compounds with DNase I, a molecular docking study was performed. Compounds 3a-c are among the most potent small organic DNase I inhibitors tested to date.


Subject(s)
Deoxyribonuclease I , Enzyme Inhibitors , Structure-Activity Relationship , Molecular Docking Simulation , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Deoxyribonuclease I/chemistry , Deoxyribonuclease I/metabolism , Xanthine Oxidase
8.
Molecules ; 29(1)2023 Dec 26.
Article in English | MEDLINE | ID: mdl-38202724

ABSTRACT

Human leukocyte antigens (HLAs) are pivotal in antigen processing, presenting to CD4+ T cells, and are linked to autoimmune disease susceptibility. In celiac disease, HLA-DQ2.5 and HLA-DQ8.1 bind gluten peptides on APCs, some recognized by CD4+ T cells, prompting inflammation and tissue damage. While extensively studied experimentally, these alleles lack comprehensive in silico analysis. To explore peptide-HLA preferences, we used molecular docking on peptide libraries, deriving quantitative matrices (QMs) for evaluating amino acids at nine-residue peptide binding cores. Our findings tie specific residue preferences to peptide backbone conformations. Validating QMs on known binders and non-binders showed strong predictive power (89-94% accuracy). These QMs excel in screening protein libraries, even whole proteomes, notably reducing time and costs for celiac disease risk assessment in novel proteins. This computational approach aligns with European Food Safety Authority guidance, promising efficient screening for potential celiac disease triggers.


Subject(s)
Celiac Disease , Humans , Molecular Docking Simulation , Glutens , Amino Acids , Peptides
9.
Int J Mol Sci ; 23(23)2022 Nov 27.
Article in English | MEDLINE | ID: mdl-36499171

ABSTRACT

Oxidative stress is an essential factor in the development and progression of Alzheimer's disease (AD). An excessive amount of reactive oxygen species (ROS) induces the peroxidation of lipid membranes, reduces the activity of antioxidant enzymes and causes neurotoxicity. In this study, we investigated the antioxidant and cholinesterase inhibitory potential of a novel galantamine-curcumin hybrid, named 4b, administered orally in two doses (2.5 mg/kg and 5 mg/kg) in scopolamine (SC)-induced neurotoxicity in mice. To evaluate the effects of 4b, we used galantamine (GAL) (3 mg/kg) and curcumin (CCN) (25 mg/kg) as positive controls. Ex vivo experiments on mouse brains showed that the higher dose of 4b (5 mg/kg) increased reduced glutathione (GSH) levels by 46%, catalase (CAT) and superoxide dismutase (SOD) activity by 57%, and glutathione peroxidase (GPx) activity by 108%, compared with the SC-treated group. At the same time, 4b (5 mg/kg) significantly reduced the brain malondialdehyde (MDA) level by 31% and acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) activities by 40% and 30%, respectively, relative to the SC-impaired group. The results showed that 4b acted as an antioxidant agent and brain protector, making it promising for further experimental research in the field of neurodegenerative diseases.


Subject(s)
Curcumin , Neurotoxicity Syndromes , Animals , Mice , Antioxidants/pharmacology , Antioxidants/metabolism , Butyrylcholinesterase , Scopolamine/pharmacology , Acetylcholinesterase/metabolism , Curcumin/pharmacology , Lipid Peroxidation , Galantamine/pharmacology , Superoxide Dismutase/metabolism , Catalase/metabolism , Oxidative Stress , Glutathione Peroxidase/metabolism , Glutathione/metabolism
10.
Molecules ; 27(17)2022 Aug 26.
Article in English | MEDLINE | ID: mdl-36080246

ABSTRACT

N-myristoyltransferase (NMT) inhibitors that were initially developed for treatment of parasitic protozoan infections, including sleeping sickness, malaria, and leismaniasis, have also shown great promise as treatment for oncological diseases. The successful transition of NMT inhibitors, which are currently at preclinical to early clinical stages, toward clinical approval and utilization may depend on the development and design of a diverse set of drug molecules with particular selectivity or pharmacological properties. In our study, we report that a common feature in the inhibitory mechanism of NMT is the formation of a salt bridge between a positively charged chemical group of the small molecule and the negatively charged C-terminus of an enzyme. Based on this observation, we designed a virtual screening protocol to identify novel ligands that mimic this mode of interaction. By screening over 1.1 million structures downloaded from the ZINC database, several hits were identified that displayed NMT inhibitory activity. The stability of the inhibitor-NMT complexes was evaluated by molecular dynamics simulations. The ligands from the stable complexes were tested in vitro and some of them appear to be promising leads for further optimization.


Subject(s)
Acyltransferases , Enzyme Inhibitors , Acyltransferases/antagonists & inhibitors , Acyltransferases/chemistry , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Ligands , Molecular Docking Simulation
11.
Molecules ; 27(10)2022 May 13.
Article in English | MEDLINE | ID: mdl-35630613

ABSTRACT

Acetylcholinesterase (AChE) is one of the classical targets in the treatment of Alzheimer's disease (AD). Inhibition of AChE slows down the hydrolysis of acetycholine and increases choline levels, improving the cognitive function. The achieved success of plant-based natural drugs acting as AChE inhibitors, such as galantamine (GAL) from Galanthus genus and huperzine A from Huperzia serrate (approved drug in China), in the treatment of AD, and the fact that natural compounds (NCs) are considered as safer and less toxic compared to synthetic drugs, led us to screen the available NCs (almost 150,000) in the ZINC12 database for AChE inhibitory activity. The compounds were screened virtually by molecular docking, filtered for suitable ADME properties, and 32 ligands from 23 structural groups were selected. The stability of the complexes was estimated via 1 µs molecular dynamics simulation. Ten compounds formed stable complexes with the enzyme and had a vendor and a reasonable price per mg. They were tested for AChE inhibitory and antioxidant activity. Five compounds showed weak AChE inhibition and three of them exhibited high antioxidant activity.


Subject(s)
Alzheimer Disease , Cholinesterase Inhibitors , Acetylcholinesterase/chemistry , Alzheimer Disease/drug therapy , Antioxidants/pharmacology , Antioxidants/therapeutic use , Cholinesterase Inhibitors/chemistry , Galantamine/pharmacology , Humans , Molecular Docking Simulation
12.
Front Mol Biosci ; 9: 1066029, 2022.
Article in English | MEDLINE | ID: mdl-36703920

ABSTRACT

The salt bridge is the strongest non-covalent interaction in nature and is known to participate in protein folding, protein-protein interactions, and molecular recognition. However, the role of salt bridges in the context of drug design has remained not well understood. Here, we report that a common feature in the mechanism of inhibition of the N-myristoyltransferases (NMT), promising targets for the treatment of protozoan infections and cancer, is the formation of a salt bridge between a positively charged chemical group of the small molecule and the negatively charged C-terminus of the enzyme. Substituting the inhibitor positively charged amine group with a neutral methylene group prevents the formation of the salt bridge and leads to a dramatic activity loss. Molecular dynamics simulations have revealed that salt bridges stabilize the NMT-ligand complexes by functioning as molecular clips that stabilize the conformation of the protein structure. As such, the creation of salt bridges between the ligands and their protein targets may find an application as a valuable tool in rational drug design.

13.
Acta Pharm ; 72(2): 289-302, 2022 Jun 01.
Article in English | MEDLINE | ID: mdl-36651509

ABSTRACT

Sophora japonica is a source of several flavonol, flavone and isoflavone glycosides that are reported to positively affect menopausal symptoms including osteoporotic complications. In the present study fructus Sophorae extract (FSE) was administered orally for three months at a dose of 200 mg kg-1 in ovariectomized (OVX) New Zealand rabbits. 3D computed tomography scans and histopathological images revealed microstructural disturbances in the bones of the castrated animals. FSE recovered most of the affected parameters in bones in a manner similar to zoledronic acid (ZA) used as a positive control. The aglycones of the main active compounds of FSE, daidzin, and genistin, were docked into the alpha and beta estrogen receptors and stable complexes were found. The findings of this study provide an insight into the effects of FSE on bone tissue loss and suggest that it could be further developed as a potential candidate for the prevention of postmenopausal osteoporotic complications.


Subject(s)
Osteoporosis , Rabbits , Animals , Osteoporosis/chemically induced , Osteoporosis/drug therapy , Osteoporosis/prevention & control , Zoledronic Acid/therapeutic use , Bone and Bones , Plant Extracts/pharmacology
14.
Int J Mol Sci ; 22(14)2021 Jul 15.
Article in English | MEDLINE | ID: mdl-34299209

ABSTRACT

Misfolded amyloid beta (Aß) peptides aggregate and form neurotoxic oligomers. Membrane and mitochondrial damages, calcium dysregulation, oxidative stress, and fibril deposits are among the possible mechanisms of Aß cytotoxicity. Galantamine (GAL) prevents apoptosis induced by Aß mainly through the ability to stimulate allosterically the α7 nAChRs and to regulate the calcium cytosolic concentration. Here, we examined the cytoprotective effects of two GAL derivatives, namely compounds 4b and 8, against Aß cytotoxicity on the human neuroblastoma cell line SH-SY5Y. The protective effects were tested at simultaneous administration, pre-incubation and post-incubation, with Aß. GAL and curcumin (CU) were used in the study as reference compounds. It was found that 4b protects cells in a similar mode as GAL, while compound 8 and CU potentiate the toxic effects of Aß. Allosteric stimulation of α7 nAChRs is suggested as a possible mechanism of the cytoprotectivity of 4b. These and previous findings characterize 4b as a prospective non-toxic multi-target agent against neurodegenerative disorders with inhibitory activity on acetylcholinesterase, antioxidant, and cytoprotective properties.


Subject(s)
Amyloid beta-Peptides/metabolism , Antioxidants/pharmacology , Cholinesterase Inhibitors/pharmacology , Curcumin/chemistry , Galantamine/chemistry , Neuroblastoma/drug therapy , Protective Agents/pharmacology , Acetylcholinesterase/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antioxidants/chemistry , Cholinesterase Inhibitors/chemistry , Curcumin/pharmacology , Cytoprotection , Galantamine/pharmacology , Humans , Neuroblastoma/metabolism , Neuroblastoma/pathology , Protective Agents/chemistry , Tumor Cells, Cultured
15.
Molecules ; 26(9)2021 May 10.
Article in English | MEDLINE | ID: mdl-34068636

ABSTRACT

The polyphenols curcumin (CU) and ferulic acid (FA) are able to inhibit the aggregation of amyloid-ß (Aß) peptide with different strengths. CU is a strong inhibitor while FA is a weaker one. In the present study, we examine the effects of CU and FA on the folding process of an Aß monomer by 1 µs molecular dynamics (MD) simulations. We found that both inhibitors increase the helical propensity and decrease the non-helical propensity of Aß peptide. They prevent the formation of a dense bulk core and shorten the average lifetime of intramolecular hydrogen bonds in Aß. CU makes more and longer-lived hydrogen bonds, hydrophobic, π-π, and cation-π interactions with Aß peptide than FA does, which is in a good agreement with the observed stronger inhibitory activity of CU on Aß aggregation.


Subject(s)
Amyloid beta-Peptides/chemistry , Coumaric Acids/pharmacology , Curcumin/pharmacology , Protein Folding , Coumaric Acids/chemistry , Curcumin/chemistry , Hydrogen Bonding , Ligands , Models, Molecular , Protein Folding/drug effects , Protein Stability/drug effects , Protein Structure, Secondary , Solvents , Static Electricity
16.
Molecules ; 26(7)2021 Mar 25.
Article in English | MEDLINE | ID: mdl-33806197

ABSTRACT

The acetylcholinesterase (AChE) inhibitors are the main drugs for symptomatic treatment of neurodegenerative disorders like Alzheimer's disease. A recently designed, synthesized and tested hybrid compound between the AChE inhibitor galantamine (GAL) and the antioxidant polyphenol curcumin (CU) showed high AChE inhibition in vitro. Here, we describe tests for acute and short-term toxicity in mice as well as antioxidant tests on brain homogenates measured the levels of malondialdehide (MDA) and glutathione (GSH) and in vitro DPPH, ABTS, FRAP and LPO inhibition assays. Hematological and serum biochemical analyses were also performed. In the acute toxicity tests, the novel AChE inhibitor given orally in mice showed LD50 of 49 mg/kg. The short-term administration of 2.5 and 5 mg/kg did not show toxicity. In the ex vivo tests, the GAL-CU hybrid performed better than GAL and CU themselves; in a dose of 5 mg/kg, it demonstrates 25% reduction in AChE activity, as well as a 28% and 73% increase in the levels of MDA and GSH, respectively. No significant changes in blood biochemical data were observed. The antioxidant activity of 4b measured ex vivo was proven in the in vitro tests. In the ABTS assay, 4b showed radical scavenging activity 10 times higher than the positive control butylhydroxy toluol (BHT). The GAL-CU hybrid is a novel non-toxic AChE inhibitor with high antioxidant activity which makes it a prospective multitarget drug candidate for treatment of neurodegenerative disorders.


Subject(s)
Brain/metabolism , Cholinesterase Inhibitors , Curcumin , Galantamine , Neurodegenerative Diseases/drug therapy , Animals , Brain/pathology , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/pharmacology , Curcumin/analogs & derivatives , Curcumin/chemistry , Curcumin/pharmacology , Disease Models, Animal , Female , Galantamine/analogs & derivatives , Galantamine/chemistry , Galantamine/pharmacology , Male , Mice , Mice, Inbred ICR , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology
17.
Molecules ; 26(7)2021 Apr 03.
Article in English | MEDLINE | ID: mdl-33916760

ABSTRACT

Despite extensive and intensive research efforts in recent decades, there is still no effective treatment for neurodegenerative diseases. On this background, the use of drugs inhibiting the enzyme acetylcholinesterase (AChE) remains an eternal evergreen in the symptomatic treatment of mild to moderate cognitive impairments. Even more, the cholinergic hypothesis, somewhat forgotten in recent years due to the shift in focus on amyloid cascade, is back to life, and the search for new, more effective AChE inhibitors continues. We generated a fragment-based library containing aromatic moieties and linkers originating from a set of novel AChE inhibitors. We used this library to design 1220 galantamine (GAL) derivatives following the model GAL (binding core) - linker (L) - aromatic fragment (Ar). The newly designed compounds were screened virtually for blood-brain barrier (BBB) permeability and binding to AChE. Among the top 10 best-scored compounds, a representative lead molecule was selected and tested for anti-AChE activity and neurotoxicity. It was found that the selected compound was a powerful non-toxic AChE inhibitor, 68 times more active than GAL, and could serve as a lead molecule for further optimization and development.


Subject(s)
Cholinesterase Inhibitors/analysis , Drug Design , Drug Discovery , Drug Evaluation, Preclinical , User-Computer Interface , Acetylcholinesterase/chemistry , Acetylcholinesterase/metabolism , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Cell Line , Cholinesterase Inhibitors/chemistry , Galantamine/chemistry , Galantamine/pharmacology , Mice , Molecular Docking Simulation , Molecular Dynamics Simulation , Neurotoxins/toxicity , Small Molecule Libraries
18.
Biomolecules ; 10(9)2020 09 15.
Article in English | MEDLINE | ID: mdl-32942739

ABSTRACT

The amyloid plaques are a key hallmark of neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease. Amyloidogenesis is a complex long-lasting multiphase process starting with the formation of nuclei of amyloid peptides: a process assigned as a primary nucleation. Curcumin (CU) is a well-known inhibitor of the aggregation of amyloid-beta (Aß) peptides. Even more, CU is able to disintegrate preformed Aß firbils and amyloid plaques. Here, we simulate by molecular dynamics the primary nucleation process of 12 Aß peptides and investigate the effects of CU on the process. We found that CU molecules intercalate among the Aß chains and bind tightly to them by hydrogen bonds, hydrophobic, π-π, and cation-π interactions. In the presence of CU, the Aß peptides form a primary nucleus of a bigger size. The peptide chains in the nucleus become less flexible and more disordered, and the number of non-native contacts and hydrogen bonds between them decreases. For comparison, the effects of the weaker Aß inhibitor ferulic acid (FA) on the primary nucleation are also examined. Our study is in good agreement with the observation that taken regularly, CU is able to prevent or at least delay the onset of neurodegenerative disorders.


Subject(s)
Amyloid beta-Peptides/chemistry , Curcumin/pharmacology , Neuroprotective Agents/pharmacology , Protein Aggregates/drug effects , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/prevention & control , Amyloid/antagonists & inhibitors , Amyloid/chemistry , Amyloid/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Binding Sites , Coumaric Acids/chemistry , Coumaric Acids/pharmacology , Curcumin/chemistry , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Molecular Dynamics Simulation , Neuroprotective Agents/chemistry , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs
19.
Molecules ; 25(15)2020 Jul 23.
Article in English | MEDLINE | ID: mdl-32717861

ABSTRACT

Galantamine (GAL) and curcumin (CU) are alkaloids used to improve symptomatically neurodegenerative conditions like Alzheimer's disease (AD). GAL acts mainly as an inhibitor of the enzyme acetylcholinesterase (AChE). CU binds to amyloid-beta (Aß) oligomers and inhibits the formation of Aß plaques. Here, we combine GAL core with CU fragments and design a combinatorial library of GAL-CU hybrids as dual-site binding AChE inhibitors. The designed hybrids are screened for optimal ADME properties and BBB permeability and docked on AChE. The 14 best performing compounds are synthesized and tested in vitro for neurotoxicity and anti-AChE activity. Five of them are less toxic than GAL and CU and show activities between 41 and 186 times higher than GAL.


Subject(s)
Acetylcholinesterase/metabolism , Alzheimer Disease/metabolism , Cholinesterase Inhibitors/chemical synthesis , Curcumin/chemistry , Galantamine/chemical synthesis , Acetylcholinesterase/chemistry , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Animals , Binding Sites , Blood-Brain Barrier/metabolism , Cell Line , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/pharmacology , Combinatorial Chemistry Techniques , Galantamine/chemistry , Galantamine/pharmacology , Humans , Mice , Molecular Docking Simulation , Molecular Structure , Structure-Activity Relationship
20.
Sci Rep ; 10(1): 10098, 2020 06 22.
Article in English | MEDLINE | ID: mdl-32572101

ABSTRACT

Many gaps in our understanding of Alzheimer's disease remain despite intense research efforts. One such prominent gap is the mechanism of Tau condensation and fibrillization. One viewpoint is that positively charged Tau is condensed by cytosolic polyanions. However, this hypothesis is likely based on an overestimation of the abundance and stability of cytosolic polyanions and an underestimation of crucial intracellular constituents - the cationic polyamines. Here, we propose an alternative mechanism grounded in cellular biology. We describe extensive molecular dynamics simulations and analysis on physiologically relevant model systems, which suggest that it is not positively charged, unmodified Tau that is condensed by cytosolic polyanions but negatively charged, hyperphosphorylated Tau that is condensed by cytosolic polycations. Our work has broad implications for anti-Alzheimer's research and drug development and the broader field of tauopathies in general, potentially paving the way to future etiologic therapies.


Subject(s)
Alzheimer Disease/metabolism , Biogenic Polyamines/adverse effects , tau Proteins/metabolism , Biogenic Polyamines/chemistry , Cytosol/metabolism , Humans , Models, Biological , Molecular Dynamics Simulation , Phosphorylation , Polyamines/metabolism , Polyelectrolytes/metabolism , Protein Aggregation, Pathological/etiology , Protein Aggregation, Pathological/metabolism , Tauopathies , tau Proteins/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...