Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Neurodegener Dis ; 17(2-3): 73-82, 2017.
Article in English | MEDLINE | ID: mdl-27760426

ABSTRACT

Alzheimer's disease (AD) is the most abundant tauopathy and is characterized by Aß-derived plaques and tau-derived tangles, resulting from the unfolding of the corresponding monomeric subunits into ordered ß-sheet oligomers and fibrils. Intervening in the toxic aggregation process is a promising therapeutic approach, but, to date, a disease-modifying therapy is neither available for AD nor for other tauopathies. Along these lines, we have previously demonstrated that a small naphthoquinone-tryptophan hybrid, termed NQTrp, is an effective modulator of tauopathy in vitro and in vivo. However, NQTrp is difficult to synthesize and is not very stable. Therefore, we tested whether a more stable and easier-to-synthesize modified version of NQTrp, containing a Cl ion, namely Cl-NQTrp, is also an effective inhibitor of tau aggregation in vitro and in vivo. Cl-NQTrp was previously shown to efficiently inhibit the aggregation of various amyloidogenic proteins and peptides. We demonstrate that Cl-NQTrp inhibits the in vitro assembly of PHF6, the aggregation-prone fragment of tau, and alleviates tauopathy symptoms in a transgenic Drosophila model through the inhibition of tau aggregation-engendered toxicity. These results suggest that Cl-NQTrp could be a unique potential therapeutic for AD since it targets aggregation of both Aß and tau.


Subject(s)
Naphthalenes/administration & dosage , Neuroprotective Agents/administration & dosage , Tauopathies/metabolism , Tryptophan/analogs & derivatives , tau Proteins/antagonists & inhibitors , tau Proteins/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Animals , Animals, Genetically Modified , Disease Models, Animal , Drosophila melanogaster , Eye/drug effects , Eye/pathology , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/metabolism , Tauopathies/drug therapy , Tryptophan/administration & dosage
2.
J Alzheimers Dis ; 51(1): 165-78, 2016.
Article in English | MEDLINE | ID: mdl-26836184

ABSTRACT

Tauopathies, such as Alzheimer's disease (AD), are a group of disorders characterized neuropathologically by intracellular toxic accumulations of abnormal protein aggregates formed by misfolding of the microtubule-associated protein tau. Since protein self-assembly appears to be an initial key step in the pathology of this group of diseases, intervening in this process can be both a prophylactic measure and a means for modifying the course of the disease for therapeutic purposes. We and others have shown that aromatic small molecules can be effective inhibitors of aggregation of various protein assemblies, by binding to the aromatic core in aggregation-prone motifs and preventing their self-assembly. Specifically, we have designed a series of small aromatic naphthoquinone-tryptophan hybrid molecules as candidate aggregation inhibitors of ß -sheet based assembly and demonstrated their efficacy toward inhibiting aggregation of the amyloid-ß peptide, another culprit of AD, as well as of various other aggregative proteins involved in other protein misfolding diseases. Here we tested whether a leading naphthoquinone-tryptophan hybrid molecule, namely NQTrp, can be repurposed as an inhibitor of the aggregation of the tau protein in vitro and in vivo. We show that the molecule inhibits the in vitro assembly of PHF6, the aggregation-prone fragment of tau protein, reduces hyperphosphorylated tau deposits and ameliorates tauopathy-related behavioral defect in an established transgenic Drosophila model expressing human tau. We suggest that NQTrp, or optimized versions of it, could act as novel disease modifying drugs for AD and other tauopathies.


Subject(s)
Carrier Proteins/metabolism , Naphthoquinones/therapeutic use , Neurotoxicity Syndromes/drug therapy , Oligopeptides/metabolism , Tryptophan/therapeutic use , tau Proteins/metabolism , Animals , Disease Models, Animal , Drosophila , Drosophila Proteins/genetics , Eye/metabolism , Eye/pathology , Eye/ultrastructure , Female , Humans , Immunoprecipitation , In Vitro Techniques , Larva , Locomotion/drug effects , Mice, Transgenic , Microscopy, Electron, Scanning , Naphthoquinones/pharmacology , Neurotoxicity Syndromes/pathology , Oligopeptides/genetics , Protein Aggregates/drug effects , Retinal Pigments/metabolism , Tryptophan/analogs & derivatives , Tryptophan/pharmacology , tau Proteins/genetics
3.
Angew Chem Int Ed Engl ; 54(7): 2062-7, 2015 Feb 09.
Article in English | MEDLINE | ID: mdl-25533189

ABSTRACT

Amyloid deposits are pathological hallmark of a large group of human degenerative disorders of unrelated etiologies. While accumulating evidence suggests that early oligomers may account for tissue degeneration, most detection tools do not allow the monitoring of early association events. Here we exploit bimolecular fluorescence complementation analysis to detect and quantify the dimerization of three major amyloidogenic polypeptides; islet amyloid polypeptide, ß-amyloid and α-synuclein. The constructed systems provided direct visualization of protein-protein interactions in which only assembled dimers display strong fluorescent signal. Potential inhibitors that interfere with the initial intermolecular interactions of islet amyloid polypeptide were further identified using this system. Moreover, the identified compounds were able to inhibit the aggregation and cytotoxicity of islet amyloid polypeptide, demonstrating the importance of targeting amyloid dimer formation for future drug development.


Subject(s)
Amyloid/chemistry , Protein Multimerization , Amyloid/ultrastructure , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/ultrastructure , Drug Discovery , Fluorescence , Humans , Islet Amyloid Polypeptide/chemistry , Islet Amyloid Polypeptide/ultrastructure , Models, Molecular , Protein Aggregates/drug effects , Protein Aggregation, Pathological/drug therapy , Protein Aggregation, Pathological/pathology , Protein Multimerization/drug effects , alpha-Synuclein/chemistry , alpha-Synuclein/ultrastructure
4.
J Phys Chem B ; 117(6): 1780-9, 2013 Feb 14.
Article in English | MEDLINE | ID: mdl-23259849

ABSTRACT

Aggregation of amyloid beta (Aß) is the hallmark of Alzheimer's disease (AD). Small molecules inhibiting Aß can be valuable therapeutics for AD. We have previously reported that 1,4-naphthoquinon-2-yl-l-tryptophan (NQTrp), reduces aggregation and oligomerization of Aß in vitro and in vivo. In silico analysis further showed that certain functional groups of NQTrp, not in the aromatic rings, are also involved in binding and inhibiting Aß. To better understand the exact mode of action and identify the groups crucial for NQTrp inhibitory activity, we conducted structure-activity analysis. Four derivatives of NQTrp were studied in silico: a D-isomer, two single-methylated and one double-methylated derivative. In silico results showed that the NQTrp groups involved in hydrogen bonds are the anilinic NH (i.e., the NH linker between the quinone and tryptophan moieties), the quinonic carbonyls, and the carboxylic acid. These predictions were supported by in vitro results. Our results should aid in designing improved small-molecule inhibitors of Aß aggregation for treating AD.


Subject(s)
Amyloid beta-Peptides/metabolism , Naphthoquinones/chemistry , Tryptophan/analogs & derivatives , Tryptophan/chemistry , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/genetics , Fluorescence Polarization , Humans , Hydrogen Bonding , Molecular Dynamics Simulation , Naphthoquinones/chemical synthesis , Naphthoquinones/therapeutic use , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Structure-Activity Relationship , Tryptophan/chemical synthesis , Tryptophan/therapeutic use
5.
Proteins ; 80(8): 1962-73, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22488522

ABSTRACT

Amyloid formation is associated with several human diseases including Alzheimer's disease (AD), Parkinson's disease, Type 2 Diabetes, and so forth, no disease modifying therapeutics are available for them. Because of the structural similarities between the amyloid species characterizing these diseases, (despite the lack of amino acid homology) it is believed that there might be a common mechanism of toxicity for these conditions. Thus, inhibition of amyloid formation could be a promising disease-modifying therapeutic strategy for them. Aromatic residues have been identified as crucial in formation and stabilization of amyloid structures. This finding was corroborated by high-resolution structural studies, theoretical analysis, and molecular dynamics simulations. Amongst the aromatic entities, tryptophan was found to possess the most amyloidogenic potential. We therefore postulate that targeting aromatic recognition interfaces by tryptophan could be a useful approach for inhibiting the formation of amyloids. Quinones are known as inhibitors of cellular metabolic pathways, to have anti- cancer, anti-viral and anti-bacterial properties and were shown to inhibit aggregation of several amyloidogenic proteins in vitro. We have previously described two quinone-tryptophan hybrids which are capable of inhibiting amyloid-beta, the protein associated with AD pathology, both in vitro and in vivo. Here we tested their generic properties and their ability to inhibit other amyloidogenic proteins including α-synuclein, islet amyloid polypeptide, lysozyme, calcitonin, and insulin. Both compounds showed efficient inhibition of all five proteins examined both by ThT fluorescence analysis and by electron microscope imaging. If verified in vivo, these small molecules could serve as leads for developing generic anti-amyloid drugs.


Subject(s)
Amyloidogenic Proteins/antagonists & inhibitors , Amyloidogenic Proteins/chemistry , Naphthoquinones/chemistry , Tryptophan/chemistry , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Calcitonin/chemistry , Humans , Insulin/chemistry , Islet Amyloid Polypeptide/chemistry , Muramidase/chemistry , alpha-Synuclein/chemistry
6.
PLoS One ; 6(1): e16564, 2011 Jan 28.
Article in English | MEDLINE | ID: mdl-21305046

ABSTRACT

An increasing body of evidence indicates that accumulation of soluble oligomeric assemblies of ß-amyloid polypeptide (Aß) play a key role in Alzheimer's disease (AD) pathology. Specifically, 56 kDa oligomeric species were shown to be correlated with impaired cognitive function in AD model mice. Several reports have documented the inhibition of Aß plaque formation by compounds from natural sources. Yet, evidence for the ability of common edible elements to modulate Aß oligomerization remains an unmet challenge. Here we identify a natural substance, based on cinnamon extract (CEppt), which markedly inhibits the formation of toxic Aß oligomers and prevents the toxicity of Aß on neuronal PC12 cells. When administered to an AD fly model, CEppt rectified their reduced longevity, fully recovered their locomotion defects and totally abolished tetrameric species of Aß in their brain. Furthermore, oral administration of CEppt to an aggressive AD transgenic mice model led to marked decrease in 56 kDa Aß oligomers, reduction of plaques and improvement in cognitive behavior. Our results present a novel prophylactic approach for inhibition of toxic oligomeric Aß species formation in AD through the utilization of a compound that is currently in use in human diet.


Subject(s)
Alzheimer Disease/prevention & control , Amyloid beta-Peptides/drug effects , Cinnamomum zeylanicum , Cognition Disorders/drug therapy , Administration, Oral , Alzheimer Disease/drug therapy , Animals , Cognition Disorders/prevention & control , Diptera , Disease Models, Animal , Mice , Mice, Transgenic , Protein Multimerization/drug effects , Treatment Outcome
7.
J Comput Aided Mol Des ; 25(2): 135-44, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21165759

ABSTRACT

Inhibiting the aggregation process of the ß-amyloid peptide is a promising strategy in treating Alzheimer's disease. In this work, we have collected a dataset of 80 small molecules with known inhibition levels and utilized them to develop two comprehensive quantitative structure-activity relationship models: a Bayesian model and a decision tree model. These models have exhibited high predictive accuracy: 87% of the training and test sets using the Bayesian model and 89 and 93% of the training and test sets, respectively, by the decision tree model. Subsequently these models were used to predict the activities of several new potential ß-amyloid aggregation inhibitors and these predictions were indeed validated by in vitro experiments. Key chemical features correlated with the inhibition ability were identified. These include the electro-topological state of carbonyl groups, AlogP and the number of hydrogen bond donor groups. The results demonstrate the feasibility of the developed models as tools for rapid screening, which could help in the design of novel potential drug candidates for Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/chemistry , Bayes Theorem , Computational Biology/methods , Quantitative Structure-Activity Relationship , Decision Support Techniques , Drug Design , Hydrogen Bonding , Models, Molecular , Molecular Structure , Small Molecule Libraries/pharmacology , Software
8.
PLoS One ; 5(6): e11101, 2010 Jun 14.
Article in English | MEDLINE | ID: mdl-20559435

ABSTRACT

The rational design of amyloid oligomer inhibitors is yet an unmet drug development need. Previous studies have identified the role of tryptophan in amyloid recognition, association and inhibition. Furthermore, tryptophan was ranked as the residue with highest amyloidogenic propensity. Other studies have demonstrated that quinones, specifically anthraquinones, can serve as aggregation inhibitors probably due to the dipole interaction of the quinonic ring with aromatic recognition sites within the amyloidogenic proteins. Here, using in vitro, in vivo and in silico tools we describe the synthesis and functional characterization of a rationally designed inhibitor of the Alzheimer's disease-associated beta-amyloid. This compound, 1,4-naphthoquinon-2-yl-L-tryptophan (NQTrp), combines the recognition capacities of both quinone and tryptophan moieties and completely inhibited Abeta oligomerization and fibrillization, as well as the cytotoxic effect of Abeta oligomers towards cultured neuronal cell line. Furthermore, when fed to transgenic Alzheimer's disease Drosophila model it prolonged their life span and completely abolished their defective locomotion. Analysis of the brains of these flies showed a significant reduction in oligomeric species of Abeta while immuno-staining of the 3(rd) instar larval brains showed a significant reduction in Abeta accumulation. Computational studies, as well as NMR and CD spectroscopy provide mechanistic insight into the activity of the compound which is most likely mediated by clamping of the aromatic recognition interface in the central segment of Abeta. Our results demonstrate that interfering with the aromatic core of amyloidogenic peptides is a promising approach for inhibiting various pathogenic species associated with amyloidogenic diseases. The compound NQTrp can serve as a lead for developing a new class of disease modifying drugs for Alzheimer's disease.


Subject(s)
Alzheimer Disease/drug therapy , Models, Biological , Naphthoquinones/pharmacology , Quinones/pharmacology , Tryptophan/analogs & derivatives , Tryptophan/pharmacology , Circular Dichroism , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Naphthoquinones/chemistry , Phenotype , Quinones/chemistry , Tryptophan/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...