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.
Brief Bioinform ; 19(6): 1183-1202, 2018 11 27.
Article in English | MEDLINE | ID: mdl-28453640

ABSTRACT

The bipartite network representation of the drug-target interactions (DTIs) in a biosystem enhances understanding of the drugs' multifaceted action modes, suggests therapeutic switching for approved drugs and unveils possible side effects. As experimental testing of DTIs is costly and time-consuming, computational predictors are of great aid. Here, for the first time, state-of-the-art DTI supervised predictors custom-made in network biology were compared-using standard and innovative validation frameworks-with unsupervised pure topological-based models designed for general-purpose link prediction in bipartite networks. Surprisingly, our results show that the bipartite topology alone, if adequately exploited by means of the recently proposed local-community-paradigm (LCP) theory-initially detected in brain-network topological self-organization and afterwards generalized to any complex network-is able to suggest highly reliable predictions, with comparable performance with the state-of-the-art-supervised methods that exploit additional (non-topological, for instance biochemical) DTI knowledge. Furthermore, a detailed analysis of the novel predictions revealed that each class of methods prioritizes distinct true interactions; hence, combining methodologies based on diverse principles represents a promising strategy to improve drug-target discovery. To conclude, this study promotes the power of bio-inspired computing, demonstrating that simple unsupervised rules inspired by principles of topological self-organization and adaptiveness arising during learning in living intelligent systems (like the brain) can efficiently equal perform complicated algorithms based on advanced, supervised and knowledge-based engineering.


Subject(s)
Brain/metabolism , Computational Biology/methods , Drug Delivery Systems , Algorithms , Drug Discovery , Drug Interactions , Reproducibility of Results
2.
Sci Rep ; 7(1): 11401, 2017 09 12.
Article in English | MEDLINE | ID: mdl-28900272

ABSTRACT

Drug repositioning identifies new indications for known drugs. Here we report repositioning of the malaria drug amodiaquine as a potential anti-cancer agent. While most repositioning efforts emerge through serendipity, we have devised a computational approach, which exploits interaction patterns shared between compounds. As a test case, we took the anti-viral drug brivudine (BVDU), which also has anti-cancer activity, and defined ten interaction patterns using our tool PLIP. These patterns characterise BVDU's interaction with its target s. Using PLIP we performed an in silico screen of all structural data currently available and identified the FDA approved malaria drug amodiaquine as a promising repositioning candidate. We validated our prediction by showing that amodiaquine suppresses chemoresistance in a multiple myeloma cancer cell line by inhibiting the chaperone function of the cancer target Hsp27. This work proves that PLIP interaction patterns are viable tools for computational repositioning and can provide search query information from a given drug and its target to identify structurally unrelated candidates, including drugs approved by the FDA, with a known safety and pharmacology profile. This approach has the potential to reduce costs and risks in drug development by predicting novel indications for known drugs and drug candidates.


Subject(s)
Amodiaquine/pharmacology , Antimalarials/pharmacology , Antineoplastic Agents/pharmacology , Computational Biology , Drug Repositioning , Amodiaquine/chemistry , Amodiaquine/therapeutic use , Antimalarials/chemistry , Antimalarials/therapeutic use , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Computational Biology/methods , Drug Repositioning/methods , HSP27 Heat-Shock Proteins/antagonists & inhibitors , Humans , Ligands , Models, Molecular , Molecular Conformation , Protein Binding , Reproducibility of Results , Structure-Activity Relationship
3.
J Med Chem ; 59(24): 11069-11078, 2016 12 22.
Article in English | MEDLINE | ID: mdl-27936766

ABSTRACT

Drug discovery is usually focused on a single protein target; in this process, existing compounds that bind to related proteins are often ignored. We describe ProBiS plugin, extension of our earlier ProBiS-ligands approach, which for a given protein structure allows prediction of its binding sites and, for each binding site, the ligands from similar binding sites in the Protein Data Bank. We developed a new database of precalculated binding site comparisons of about 290000 proteins to allow fast prediction of binding sites in existing proteins. The plugin enables advanced viewing of predicted binding sites, ligands' poses, and their interactions in three-dimensional graphics. Using the InhA query protein, an enoyl reductase enzyme in the Mycobacterium tuberculosis fatty acid biosynthesis pathway, we predicted its possible ligands and assessed their inhibitory activity experimentally. This resulted in three previously unrecognized inhibitors with novel scaffolds, demonstrating the plugin's utility in the early drug discovery process.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Drug Discovery , Mycobacterium tuberculosis/enzymology , Oxidoreductases/antagonists & inhibitors , Bacterial Proteins/metabolism , Binding Sites/drug effects , Dose-Response Relationship, Drug , Fatty Acids/biosynthesis , Ligands , Models, Molecular , Molecular Structure , Mycobacterium tuberculosis/metabolism , Oxidoreductases/metabolism , Structure-Activity Relationship
4.
Curr Pharm Des ; 22(21): 3124-34, 2016.
Article in English | MEDLINE | ID: mdl-26873186

ABSTRACT

BACKGROUND: Drug repositioning aims to identify novel indications for existing drugs. One approach to repositioning exploits shared binding sites between the drug targets and other proteins. Here, we review the principle and algorithms of such target hopping and illustrate them in Chagas disease, an in Latin America widely spread, but neglected disease. CONCLUSION: We demonstrate how target hopping recovers known treatments for Chagas disease and predicts novel drugs, such as the antiviral foscarnet, which we predict to target Farnesyl Pyrophosphate Synthase in Trypanosoma cruzi, the causative agent of Chagas disease.


Subject(s)
Algorithms , Chagas Disease/drug therapy , Drug Repositioning , Trypanocidal Agents/pharmacology , Trypanosoma cruzi/drug effects , Chagas Disease/metabolism , Humans , Models, Molecular , Polyisoprenyl Phosphates/antagonists & inhibitors , Polyisoprenyl Phosphates/metabolism , Sesquiterpenes/antagonists & inhibitors , Sesquiterpenes/metabolism , Trypanocidal Agents/chemistry , Trypanosoma cruzi/enzymology
5.
Prog Biophys Mol Biol ; 116(2-3): 174-86, 2014.
Article in English | MEDLINE | ID: mdl-24923864

ABSTRACT

Detection of remote binding site similarity in proteins plays an important role for drug repositioning and off-target effect prediction. Various non-covalent interactions such as hydrogen bonds and van-der-Waals forces drive ligands' molecular recognition by binding sites in proteins. The increasing amount of available structures of protein-small molecule complexes enabled the development of comparative approaches. Several methods have been developed to characterize and compare protein-ligand interaction patterns. Usually implemented as fingerprints, these are mainly used for post processing docking scores and (off-)target prediction. In the latter application, interaction profiles detect similarities in the bound interactions of different ligands and thus identify essential interactions between a protein and its small molecule ligands. Interaction pattern similarity correlates with binding site similarity and is thus contributing to a higher precision in binding site similarity assessment of proteins with distinct global structure. This renders it valuable for existing drug repositioning approaches in structural bioinformatics. Current methods to characterize and compare structure-based interaction patterns - both for protein-small-molecule and protein-protein interactions - as well as their potential in target prediction will be reviewed in this article. The question of how the set of interaction types, flexibility or water-mediated interactions, influence the comparison of interaction patterns will be discussed. Due to the wealth of protein-ligand structures available today, predicted targets can be ranked by comparing their ligand interaction pattern to patterns of the known target. Such knowledge-based methods offer high precision in comparison to methods comparing whole binding sites based on shape and amino acid physicochemical similarity.


Subject(s)
Polypharmacology , Proteins/chemistry , Proteins/metabolism , Binding Sites , Ligands , Protein Binding , Small Molecule Libraries/metabolism
6.
PLoS One ; 8(6): e65894, 2013.
Article in English | MEDLINE | ID: mdl-23805191

ABSTRACT

Drug repositioning applies established drugs to new disease indications with increasing success. A pre-requisite for drug repurposing is drug promiscuity (polypharmacology) - a drug's ability to bind to several targets. There is a long standing debate on the reasons for drug promiscuity. Based on large compound screens, hydrophobicity and molecular weight have been suggested as key reasons. However, the results are sometimes contradictory and leave space for further analysis. Protein structures offer a structural dimension to explain promiscuity: Can a drug bind multiple targets because the drug is flexible or because the targets are structurally similar or even share similar binding sites? We present a systematic study of drug promiscuity based on structural data of PDB target proteins with a set of 164 promiscuous drugs. We show that there is no correlation between the degree of promiscuity and ligand properties such as hydrophobicity or molecular weight but a weak correlation to conformational flexibility. However, we do find a correlation between promiscuity and structural similarity as well as binding site similarity of protein targets. In particular, 71% of the drugs have at least two targets with similar binding sites. In order to overcome issues in detection of remotely similar binding sites, we employed a score for binding site similarity: LigandRMSD measures the similarity of the aligned ligands and uncovers remote local similarities in proteins. It can be applied to arbitrary structural binding site alignments. Three representative examples, namely the anti-cancer drug methotrexate, the natural product quercetin and the anti-diabetic drug acarbose are discussed in detail. Our findings suggest that global structural and binding site similarity play a more important role to explain the observed drug promiscuity in the PDB than physicochemical drug properties like hydrophobicity or molecular weight. Additionally, we find ligand flexibility to have a minor influence.


Subject(s)
Acarbose/chemistry , Databases, Protein , Methotrexate/chemistry , Proteins/chemistry , Quercetin/chemistry , Binding Sites
7.
Integr Biol (Camb) ; 4(7): 778-88, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22538435

ABSTRACT

Recently, there has been much interest in gene-disease networks and polypharmacology as a basis for drug repositioning. Here, we integrate data from structural and chemical databases to create a drug-target-disease network for 147 promiscuous drugs, their 553 protein targets, and 44 disease indications. Visualizing and analyzing such complex networks is still an open problem. We approach it by mining the network for network motifs of bi-cliques. In our case, a bi-clique is a subnetwork in which every drug is linked to every target and disease. Since the data are incomplete, we identify incomplete bi-cliques, whose completion introduces novel, predicted links from drugs to targets and diseases. We demonstrate the power of this approach by repositioning cardiovascular drugs to parasitic diseases, by predicting the cancer-related kinase PIK3CG as a novel target of resveratrol, and by identifying for five drugs a shared binding site in four serine proteases and novel links to cancer, cardiovascular, and parasitic diseases.


Subject(s)
Chemistry, Pharmaceutical/methods , Computational Biology/methods , Drug Repositioning , Pharmaceutical Preparations/chemistry , Algorithms , Amino Acid Sequence , Binding Sites , Databases, Factual , Drug Delivery Systems , Gene Regulatory Networks , Humans , Molecular Conformation , Molecular Sequence Data , Quercetin/chemistry , Resveratrol , Sequence Homology, Amino Acid , Software , Stilbenes/chemistry
8.
J Chromatogr A ; 1217(33): 5328-36, 2010 Aug 13.
Article in English | MEDLINE | ID: mdl-20621298

ABSTRACT

Since red blood cells (RBCs) lack nuclei and organelles, cell membrane is their main load-bearing component and, according to a dynamic interaction with the cytoskeleton compartment, plays a pivotal role in their functioning. Even if erythrocyte membranes are available in large quantities, the low abundance and the hydrophobic nature of cell membrane proteins complicate their purification and detection by conventional 2D gel-based proteomic approaches. So, in order to increase the efficiency of RBC membrane proteome identification, here we took advantage of a simple and reproducible membrane sub-fractionation method coupled to Multidimensional Protein Identification Technology (MudPIT). In addition, the adoption of a stringent RBC filtration strategy from the whole blood, permitted to remove exhaustively contaminants, such as platelets and white blood cells, and to identify a total of 275 proteins in the three RBC membrane fractions collected and analysed. Finally, by means of software for the elaboration of the great quantity of data obtained and programs for statistical analysis and protein classification, it was possible to determine the validity of the entire system workflow and to assign the proper sub-cellular localization and function for the greatest number of the identified proteins.


Subject(s)
Erythrocyte Membrane/chemistry , Mass Spectrometry/methods , Membrane Proteins , Proteomics/methods , Software , Humans , Membrane Proteins/chemistry , Membrane Proteins/classification , Membrane Proteins/isolation & purification , Multivariate Analysis , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL
...