Publications Search
Explore how scientists all over the world use DrugBank in their research.
Showing 4841 to 4844 of 5186 entries
Published in July 2012
The beta2-adrenoceptor agonist formoterol stimulates mitochondrial biogenesis.
Abstract: Mitochondrial dysfunction is a common mediator of disease and organ injury. Although recent studies show that inducing mitochondrial biogenesis (MB) stimulates cell repair and regeneration, only a limited number of chemicals are known to induce MB. To examine the impact of the beta-adrenoceptor (beta-AR) signaling pathway on MB, primary renal proximal tubule cells (RPTC) and adult feline cardiomyocytes were exposed for 24 h to multiple beta-AR agonists: isoproterenol (nonselective beta-AR agonist), (+/-)-(R*,R*)-[4-[2-[[2-(3-chlorophenyl)-2-hydroxyethyl]amino]propyl]phenoxy] acetic acid sodium hydrate (BRL 37344) (selective beta(3)-AR agonist), and formoterol (selective beta(2)-AR agonist). The Seahorse Biosciences (North Billerica, MA) extracellular flux analyzer was used to quantify carbonylcyanide p-trifluoromethoxyphenylhydrazone (FCCP)-uncoupled oxygen consumption rate (OCR), a marker of maximal electron transport chain activity. Isoproterenol and BRL 37244 did not alter mitochondrial respiration at any of the concentrations examined. Formoterol exposure resulted in increases in both FCCP-uncoupled OCR and mitochondrial DNA (mtDNA) copy number. The effect of formoterol on OCR in RPTC was inhibited by the beta-AR antagonist propranolol and the beta(2)-AR inverse agonist 3-(isopropylamino)-1-[(7-methyl-4-indanyl)oxy]butan-2-ol hydrochloride (ICI-118,551). Mice exposed to formoterol for 24 or 72 h exhibited increases in kidney and heart mtDNA copy number, peroxisome proliferator-activated receptor gamma coactivator 1alpha, and multiple genes involved in the mitochondrial electron transport chain (F0 subunit 6 of transmembrane F-type ATP synthase, NADH dehydrogenase subunit 1, NADH dehydrogenase subunit 6, and NADH dehydrogenase [ubiquinone] 1beta subcomplex subunit 8). Cheminformatic modeling, virtual chemical library screening, and experimental validation identified nisoxetine from the Sigma Library of Pharmacologically Active Compounds and two compounds from the ChemBridge DIVERSet that increased mitochondrial respiratory capacity. These data provide compelling evidence for the use and development of beta(2)-AR ligands for therapeutic MB.
Published in July 2012
Bioinformatics for personal genome interpretation.
Abstract: An international consortium released the first draft sequence of the human genome 10 years ago. Although the analysis of this data has suggested the genetic underpinnings of many diseases, we have not yet been able to fully quantify the relationship between genotype and phenotype. Thus, a major current effort of the scientific community focuses on evaluating individual predispositions to specific phenotypic traits given their genetic backgrounds. Many resources aim to identify and annotate the specific genes responsible for the observed phenotypes. Some of these use intra-species genetic variability as a means for better understanding this relationship. In addition, several online resources are now dedicated to collecting single nucleotide variants and other types of variants, and annotating their functional effects and associations with phenotypic traits. This information has enabled researchers to develop bioinformatics tools to analyze the rapidly increasing amount of newly extracted variation data and to predict the effect of uncharacterized variants. In this work, we review the most important developments in the field--the databases and bioinformatics tools that will be of utmost importance in our concerted effort to interpret the human variome.
Published on July 23, 2012
ZINC: a free tool to discover chemistry for biology.
Abstract: ZINC is a free public resource for ligand discovery. The database contains over twenty million commercially available molecules in biologically relevant representations that may be downloaded in popular ready-to-dock formats and subsets. The Web site also enables searches by structure, biological activity, physical property, vendor, catalog number, name, and CAS number. Small custom subsets may be created, edited, shared, docked, downloaded, and conveyed to a vendor for purchase. The database is maintained and curated for a high purchasing success rate and is freely available at zinc.docking.org.
Published on July 23, 2012
Sets2Networks: network inference from repeated observations of sets.
Abstract: BACKGROUND: The skeleton of complex systems can be represented as networks where vertices represent entities, and edges represent the relations between these entities. Often it is impossible, or expensive, to determine the network structure by experimental validation of the binary interactions between every vertex pair. It is usually more practical to infer the network from surrogate observations. Network inference is the process by which an underlying network of relations between entities is determined from indirect evidence. While many algorithms have been developed to infer networks from quantitative data, less attention has been paid to methods which infer networks from repeated co-occurrence of entities in related sets. This type of data is ubiquitous in the field of systems biology and in other areas of complex systems research. Hence, such methods would be of great utility and value. RESULTS: Here we present a general method for network inference from repeated observations of sets of related entities. Given experimental observations of such sets, we infer the underlying network connecting these entities by generating an ensemble of networks consistent with the data. The frequency of occurrence of a given link throughout this ensemble is interpreted as the probability that the link is present in the underlying real network conditioned on the data. Exponential random graphs are used to generate and sample the ensemble of consistent networks, and we take an algorithmic approach to numerically execute the inference method. The effectiveness of the method is demonstrated on synthetic data before employing this inference approach to problems in systems biology and systems pharmacology, as well as to construct a co-authorship collaboration network. We predict direct protein-protein interactions from high-throughput mass-spectrometry proteomics, integrate data from Chip-seq and loss-of-function/gain-of-function followed by expression data to infer a network of associations between pluripotency regulators, extract a network that connects 53 cancer drugs to each other and to 34 severe adverse events by mining the FDA's Adverse Events Reporting Systems (AERS), and construct a co-authorship network that connects Mount Sinai School of Medicine investigators. The predicted networks and online software to create networks from entity-set libraries are provided online at http://www.maayanlab.net/S2N. CONCLUSIONS: The network inference method presented here can be applied to resolve different types of networks in current systems biology and systems pharmacology as well as in other fields of research.
Published on July 18, 2012
CIDeR: multifactorial interaction networks in human diseases.
Abstract: The pathobiology of common diseases is influenced by heterogeneous factors interacting in complex networks. CIDeR http://mips.helmholtz-muenchen.de/cider/ is a publicly available, manually curated, integrative database of metabolic and neurological disorders. The resource provides structured information on 18,813 experimentally validated interactions between molecules, bioprocesses and environmental factors extracted from the scientific literature. Systematic annotation and interactive graphical representation of disease networks make CIDeR a versatile knowledge base for biologists, analysis of large-scale data and systems biology approaches.
Published on July 17, 2012
INDI: a computational framework for inferring drug interactions and their associated recommendations.
Abstract: Inferring drug-drug interactions (DDIs) is an essential step in drug development and drug administration. Most computational inference methods focus on modeling drug pharmacokinetics, aiming at interactions that result from a common metabolizing enzyme (CYP). Here, we introduce a novel prediction method, INDI (INferring Drug Interactions), allowing the inference of both pharmacokinetic, CYP-related DDIs (along with their associated CYPs) and pharmacodynamic, non-CYP associated ones. On cross validation, it obtains high specificity and sensitivity levels (AUC (area under the receiver-operating characteristic curve) >/=0.93). In application to the FDA adverse event reporting system, 53% of the drug events could potentially be connected to known (41%) or predicted (12%) DDIs. Additionally, INDI predicts the severity level of each DDI upon co-administration of the involved drugs, suggesting that severe interactions are abundant in the clinical practice. Examining regularly taken medications by hospitalized patients, 18% of the patients receive known or predicted severely interacting drugs and are hospitalized more frequently. Access to INDI and its predictions is provided via a web tool at http://www.cs.tau.ac.il/~bnet/software/INDI, facilitating the inference and exploration of drug interactions and providing important leads for physicians and pharmaceutical companies alike.
Published on July 15, 2012
iFad: an integrative factor analysis model for drug-pathway association inference.
Abstract: MOTIVATION: Pathway-based drug discovery considers the therapeutic effects of compounds in the global physiological environment. This approach has been gaining popularity in recent years because the target pathways and mechanism of action for many compounds are still unknown, and there are also some unexpected off-target effects. Therefore, the inference of drug-pathway associations is a crucial step to fully realize the potential of system-based pharmacological research. Transcriptome data offer valuable information on drug-pathway targets because the pathway activities may be reflected through gene expression levels. Hence, it is of great interest to jointly analyze the drug sensitivity and gene expression data from the same set of samples to investigate the gene-pathway-drug-pathway associations. RESULTS: We have developed iFad, a Bayesian sparse factor analysis model to jointly analyze the paired gene expression and drug sensitivity datasets measured across the same panel of samples. The model enables direct incorporation of prior knowledge regarding gene-pathway and/or drug-pathway associations to aid the discovery of new association relationships. We use a collapsed Gibbs sampling algorithm for inference. Satisfactory performance of the proposed model was found for both simulated datasets and real data collected on the NCI-60 cell lines. Our results suggest that iFad is a promising approach for the identification of drug targets. This model also provides a general statistical framework for pathway-based integrative analysis of other types of -omics data. AVAILABILITY: The R package 'iFad' and real NCI-60 dataset used are available at http://bioinformatics.med.yale.edu/group.
Published on July 10, 2012
Druggability Assessment of Allosteric Proteins by Dynamics Simulations in the Presence of Probe Molecules.
Abstract: Druggability assessment of a target protein has emerged in recent years as an important concept in hit-to-lead optimization. A reliable and physically relevant measure of druggability would allow informed decisions on the risk of investing in a particular target. Here, we define "druggability" as a quantitative estimate of binding sites and affinities for a potential drug acting on a specific protein target. In the present study, we describe a new methodology that successfully predicts the druggability and maximal binding affinity for a series of challenging targets, including those that function through allosteric mechanisms. Two distinguishing features of the methodology are (i) simulation of the binding dynamics of a diversity of probe molecules selected on the basis of an analysis of approved drugs and (ii) identification of druggable sites and estimation of corresponding binding affinities on the basis of an evaluation of the geometry and energetics of bound probe clusters. The use of the methodology for a variety of targets such as murine double mutant-2, protein tyrosine phosphatase 1B (PTP1B), lymphocyte function-associated antigen 1, vertebrate kinesin-5 (Eg5), and p38 mitogen-activated protein kinase provides examples for which the method correctly captures the location and binding affinities of known drugs. It also provides insights into novel druggable sites and the target's structural changes that would accommodate, if not promote and stabilize, drug binding. Notably, the ability to identify high affinity spots even in challenging cases such as PTP1B or Eg5 shows promise as a rational tool for assessing the druggability of protein targets and identifying allosteric or novel sites for drug binding.
Published on July 10, 2012
Identifying mechanism-of-action targets for drugs and probes.
Abstract: Notwithstanding their key roles in therapy and as biological probes, 7% of approved drugs are purported to have no known primary target, and up to 18% lack a well-defined mechanism of action. Using a chemoinformatics approach, we sought to "de-orphanize" drugs that lack primary targets. Surprisingly, targets could be easily predicted for many: Whereas these targets were not known to us nor to the common databases, most could be confirmed by literature search, leaving only 13 Food and Drug Administration-approved drugs with unknown targets; the number of drugs without molecular targets likely is far fewer than reported. The number of worldwide drugs without reasonable molecular targets similarly dropped, from 352 (25%) to 44 (4%). Nevertheless, there remained at least seven drugs for which reasonable mechanism-of-action targets were unknown but could be predicted, including the antitussives clemastine, cloperastine, and nepinalone; the antiemetic benzquinamide; the muscle relaxant cyclobenzaprine; the analgesic nefopam; and the immunomodulator lobenzarit. For each, predicted targets were confirmed experimentally, with affinities within their physiological concentration ranges. Turning this question on its head, we next asked which drugs were specific enough to act as chemical probes. Over 100 drugs met the standard criteria for probes, and 40 did so by more stringent criteria. A chemical information approach to drug-target association can guide therapeutic development and reveal applications to probe biology, a focus of much current interest.
Published on July 7, 2012
Bioinformatics and variability in drug response: a protein structural perspective.
Abstract: Marketed drugs frequently perform worse in clinical practice than in the clinical trials on which their approval is based. Many therapeutic compounds are ineffective for a large subpopulation of patients to whom they are prescribed; worse, a significant fraction of patients experience adverse effects more severe than anticipated. The unacceptable risk-benefit profile for many drugs mandates a paradigm shift towards personalized medicine. However, prior to adoption of patient-specific approaches, it is useful to understand the molecular details underlying variable drug response among diverse patient populations. Over the past decade, progress in structural genomics led to an explosion of available three-dimensional structures of drug target proteins while efforts in pharmacogenetics offered insights into polymorphisms correlated with differential therapeutic outcomes. Together these advances provide the opportunity to examine how altered protein structures arising from genetic differences affect protein-drug interactions and, ultimately, drug response. In this review, we first summarize structural characteristics of protein targets and common mechanisms of drug interactions. Next, we describe the impact of coding mutations on protein structures and drug response. Finally, we highlight tools for analysing protein structures and protein-drug interactions and discuss their application for understanding altered drug responses associated with protein structural variants.
Showing 4841 to 4844 of 5186 entries