Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Methods Mol Biol ; 1613: 101-124, 2017.
Article in English | MEDLINE | ID: mdl-28849560

ABSTRACT

Analysis of NGS and other sequencing data, gene variants, gene expression, proteomics, and other high-throughput (OMICs) data is challenging because of its biological complexity and high level of technical and biological noise. One way to deal with both problems is to perform analysis with a high fidelity annotated knowledgebase of protein interactions, pathways, and functional ontologies. This knowledgebase has to be structured in a computer-readable format and must include software tools for managing experimental data, analysis, and reporting. Here, we present MetaCore™ and Key Pathway Advisor (KPA), an integrated platform for functional data analysis. On the content side, MetaCore and KPA encompass a comprehensive database of molecular interactions of different types, pathways, network models, and ten functional ontologies covering human, mouse, and rat genes. The analytical toolkit includes tools for gene/protein list enrichment analysis, statistical "interactome" tool for the identification of over- and under-connected proteins in the dataset, and a biological network analysis module made up of network generation algorithms and filters. The suite also features Advanced Search, an application for combinatorial search of the database content, as well as a Java-based tool called Pathway Map Creator for drawing and editing custom pathway maps. Applications of MetaCore and KPA include molecular mode of action of disease research, identification of potential biomarkers and drug targets, pathway hypothesis generation, analysis of biological effects for novel small molecule compounds and clinical applications (analysis of large cohorts of patients, and translational and personalized medicine).


Subject(s)
Computational Biology/methods , Gene Regulatory Networks , Protein Interaction Mapping , Algorithms , Animals , Humans , Knowledge Bases , Mice , Rats
2.
Methods Mol Biol ; 563: 177-96, 2009.
Article in English | MEDLINE | ID: mdl-19597786

ABSTRACT

Analysis of microarray, SNPs, proteomics, and other high-throughput (OMICs) data is challenging because of its biological complexity and high level of technical and biological noise. One way to deal with both problems is to perform analysis with a high-fidelity annotated knowledge base of protein interactions, pathways, and functional ontologies. This knowledge base has to be structured in a computer-readable format and must include software tools for managing experimental data, analysis, and reporting. Here we present MetaDiscovery, an integrated platform for functional data analysis which is being developed at GeneGo for the past 8 years. On the content side, MetaDiscovery encompasses a comprehensive database of protein interactions of different types, pathways, network models and 10 functional ontologies covering human, mouse, and rat proteins. The analytical toolkit includes tools for gene/protein list enrichment analysis, statistical "interactome" tool for identification of over- and under-connected proteins in the data set, and a network module made up of network generation algorithms and filters. The suite also features MetaSearch, an application for combinatorial search of the database content, as well as a Java-based tool called MapEditor for drawing and editing custom pathway maps. Applications of MetaDiscovery include identification of potential biomarkers and drug targets, pathway hypothesis generation, analysis of biological effects for novel small molecule compounds, and clinical applications (analysis of large cohorts of patients and translational and personalized medicine).


Subject(s)
Genomics/methods , Knowledge Bases , Protein Interaction Mapping/methods , Proteins/metabolism , Software , Systems Biology/methods , Animals , Database Management Systems , Databases, Genetic , Drug Discovery , Humans , Metabolic Networks and Pathways , Proteins/genetics , Small Molecule Libraries/pharmacology
3.
BMC Biol ; 6: 49, 2008 Nov 12.
Article in English | MEDLINE | ID: mdl-19014478

ABSTRACT

BACKGROUND: In recent years, the maturation of microarray technology has allowed the genome-wide analysis of gene expression patterns to identify tissue-specific and ubiquitously expressed ('housekeeping') genes. We have performed a functional and topological analysis of housekeeping and tissue-specific networks to identify universally necessary biological processes, and those unique to or characteristic of particular tissues. RESULTS: We measured whole genome expression in 31 human tissues, identifying 2374 housekeeping genes expressed in all tissues, and genes uniquely expressed in each tissue. Comprehensive functional analysis showed that the housekeeping set is substantially larger than previously thought, and is enriched with vital processes such as oxidative phosphorylation, ubiquitin-dependent proteolysis, translation and energy metabolism. Network topology of the housekeeping network was characterized by higher connectivity and shorter paths between the proteins than the global network. Ontology enrichment scoring and network topology of tissue-specific genes were consistent with each tissue's function and expression patterns clustered together in accordance with tissue origin. Tissue-specific genes were twice as likely as housekeeping genes to be drug targets, allowing the identification of tissue 'signature networks' that will facilitate the discovery of new therapeutic targets and biomarkers of tissue-targeted diseases. CONCLUSION: A comprehensive functional analysis of housekeeping and tissue-specific genes showed that the biological function of housekeeping and tissue-specific genes was consistent with tissue origin. Network analysis revealed that tissue-specific networks have distinct network properties related to each tissue's function. Tissue 'signature networks' promise to be a rich source of targets and biomarkers for disease treatment and diagnosis.


Subject(s)
Gene Expression Regulation , Genes/genetics , Organ Specificity , Cluster Analysis , Gene Regulatory Networks/genetics , Humans , Oligonucleotide Array Sequence Analysis
4.
Drug Metab Dispos ; 34(3): 495-503, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16381662

ABSTRACT

The challenge of predicting the metabolism or toxicity of a drug in humans has been approached using in vivo animal models, in vitro systems, high throughput genomics and proteomics methods, and, more recently, computational approaches. Understanding the complexity of biological systems requires a broader perspective rather than focusing on just one method in isolation for prediction. Multiple methods may therefore be necessary and combined for a more accurate prediction. In the field of drug metabolism and toxicology, we have seen the growth, in recent years, of computational quantitative structure-activity relationships (QSARs), as well as empirical data from microarrays. In the current study we have further developed a novel computational approach, MetaDrug, that 1) predicts metabolites for molecules based on their chemical structure, 2) predicts the activity of the original compound and its metabolites with various absorption, distribution, metabolism, excretion, and toxicity models, 3) incorporates the predictions with human cell signaling and metabolic pathways and networks, and 4) integrates networks and metabolites, with relevant toxicogenomic or other high throughput data. We have demonstrated the utility of such an approach using recently published data from in vitro metabolism and microarray studies for aprepitant, 2(S)-((3,5-bis(trifluoromethyl)benzyl)-oxy)-3(S)phenyl-4-((3-oxo-1,2,4-triazol-5-yl)methyl)morpholine (L-742694), trovofloxacin, 4-hydroxytamoxifen, and artemisinin and other artemisinin analogs to show the predicted interactions with cytochromes P450, pregnane X receptor, and P-glycoprotein, and the metabolites and the networks of genes that are affected. As a comparison, we used a second computational approach, MetaCore, to generate statistically significant gene networks with the available expression data. These case studies demonstrate the combination of QSARs and systems biology methods.


Subject(s)
Drug-Related Side Effects and Adverse Reactions , Microarray Analysis , Models, Theoretical , Pharmaceutical Preparations , Quantitative Structure-Activity Relationship , Software Design , Morpholines/metabolism , Morpholines/pharmacokinetics , Morpholines/toxicity , Pharmaceutical Preparations/chemistry , Pharmaceutical Preparations/metabolism
5.
J Alzheimers Dis ; 8(3): 227-41, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16340081

ABSTRACT

Increasing evidence suggests that oxidative injury is involved in the pathogenesis of many age-related neurodegenerative disorders, including Alzheimer's disease (AD). Identifying the protein targets of oxidative stress is critical to determine which proteins may be responsible for the neuronal impairments and subsequent cell death that occurs in AD. In this study, we have applied a high-throughput shotgun proteomic approach to identify the targets of protein carbonylation in both aged and PS1 + AbetaPP transgenic mice. However, because of the inherent difficulties associated with proteomic database searching algorithms, several newly developed bioinformatic tools were implemented to ascertain a probability-based discernment between correct protein assignments and false identifications to improve the accuracy of protein identification. Assigning a probability to each identified peptide/protein allows one to objectively monitor the expression and relative abundance of particular proteins from diverse samples, including tissue from transgenic mice of mixed genetic backgrounds. This robust bioinformatic approach also permits the comparison of proteomic data generated by different laboratories since it is instrument- and database-independent. Applying these statistical models to our initial studies, we detected a total of 117 oxidatively modified (carbonylated) proteins, 59 of which were specifically associated with PS1 + AbetaPP mice. Pathways and network component analyses suggest that there are three major protein networks that could be potentially altered in PS1 + AbetaPP mice as a result of oxidative modifications. These pathways are 1) iNOS-integrin signaling pathway, 2) CRE/CBP transcription regulation and 3) rab-lyst vesicular trafficking. We believe the results of these studies will help establish an initial AD database of oxidatively modified proteins and provide a foundation for the design of future hypothesis driven research in the areas of aging and neurodegeneration.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Disease Models, Animal , Membrane Proteins/metabolism , Oxidative Stress/physiology , Proteomics/methods , Synaptic Membranes/metabolism , Activating Transcription Factor 2/metabolism , Aging/physiology , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Cell Death , Integrins/metabolism , Mice , Mice, Transgenic , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Nerve Net/physiology , Nitric Oxide Synthase/metabolism , Phosphoproteins/metabolism , Presenilin-1 , Probability , Protein Carbonylation/physiology , Signal Transduction/physiology , Synaptic Membranes/pathology
6.
Expert Opin Drug Metab Toxicol ; 1(2): 303-24, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16922645

ABSTRACT

There is an urgent requirement within the pharmaceutical and biotechnology industries, regulatory authorities and academia to improve the success of molecules that are selected for clinical trials. Although absorption, distribution, metabolism, excretion and toxicity (ADME/Tox) properties are some of the many components that contribute to successful drug discovery and development, they represent factors for which we currently have in vitro and in vivo data that can be modelled computationally. Understanding the possible toxicity and the metabolic fate of xenobiotics in the human body is particularly important in early drug discovery. There is, therefore, a need for computational methodologies for uncovering the relationships between the structure and the biological activity of novel molecules. The convergence of numerous technologies, including high-throughput techniques, databases, ADME/Tox modelling and systems biology modelling, is leading to the foundation of systems-ADME/Tox. Results from experiments can be integrated with predictions to globally simulate and understand the likely complete effects of a molecule in humans. The development and early application of major components of MetaDrug (GeneGo, Inc.) software will be described, which includes rule-based metabolite prediction, quantitative structure-activity relationship models for major drug metabolising enzymes, and an extensive database of human protein-xenobiotic interactions. This represents a combined approach to predicting drug metabolism. MetaDrug can be readily used for visualising Phase I and II metabolic pathways, as well as interpreting high-throughput data derived from microarrays as networks of interacting objects. This will ultimately aid in hypothesis generation and the early triaging of molecules likely to have undesirable predicted properties or measured effects on key proteins and cellular functions.


Subject(s)
Computational Biology/methods , Pharmaceutical Preparations/metabolism , Technology, Pharmaceutical/methods , Computational Biology/trends , Drug Interactions , Drug-Related Side Effects and Adverse Reactions , Humans , Pharmaceutical Preparations/administration & dosage , Software , Technology, Pharmaceutical/trends
7.
Drug Metab Dispos ; 33(3): 474-81, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15608136

ABSTRACT

The increasing generation of biological data represents a challenge to understanding the complexity of systems, resulting in scientists increasingly focused on a relatively narrow area of study, thereby limiting insight that can be gained from a broader perspective. In the field of drug metabolism and toxicology we are witnessing the characterization of many proteins. Most of the key enzymes and transporters are recognized as transcriptionally regulated by the nuclear hormone receptors such as pregnane X receptor, constitutive androstane receptor, vitamin D receptor, glucocorticoid receptor, and others. There is apparent cross talk in regulation, since multiple receptors may modulate expression of a single enzyme or transporter, representing one of many areas of active research interest. We have used published data on nuclear hormone receptors, enzymes, ligands, and other biological information to manually annotate an Oracle database, forming the basis of a platform for querying (MetaDrug). Using algorithms, we have demonstrated how nuclear hormone receptors alone can form a network of direct interactions, and when expanded, this network increases in complexity to describe the interactions with target genes as well as small molecules known to bind a receptor, enzyme, or transporter. We have also described how the database can be used for visualizing high-throughput microarray data derived from a published study of MCF-7 cells treated with 4-hydroxytamoxifen, to highlight potential downstream effects of molecule treatment. The database represents a novel knowledge mining and analytical tool that, to be relevant, requires continual updating to evolve alongside other key storage systems and sources of biological knowledge.


Subject(s)
Databases, Factual , Pharmaceutical Preparations/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Software Design , Drug Design , Enzymes/metabolism , Neural Networks, Computer , Oligonucleotide Array Sequence Analysis , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...