Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Nat Commun ; 6: 7101, 2015 Jun 09.
Article in English | MEDLINE | ID: mdl-26055627

ABSTRACT

Drug side effects cause a significant clinical and economic burden. However, mechanisms of drug action underlying side effect pathogenesis remain largely unknown. Here, we integrate pharmacogenomic and clinical data with a human metabolic network and find that non-pharmacokinetic metabolic pathways dysregulated by drugs are linked to the development of side effects. We show such dysregulated metabolic pathways contain genes with sequence variants affecting side effect incidence, play established roles in pathophysiology, have significantly altered activity in corresponding diseases, are susceptible to metabolic inhibitors and are effective targets for therapeutic nutrient supplementation. Our results indicate that metabolic dysregulation represents a common mechanism underlying side effect pathogenesis that is distinct from the role of metabolism in drug clearance. We suggest that elucidating the relationships between the cellular response to drugs, genetic variation of patients and cell metabolism may help managing side effects by personalizing drug prescriptions and nutritional intervention strategies.


Subject(s)
Drug-Related Side Effects and Adverse Reactions , Pharmacogenetics , Databases, Factual , Humans
2.
Nat Biotechnol ; 31(5): 419-25, 2013 May.
Article in English | MEDLINE | ID: mdl-23455439

ABSTRACT

Multiple models of human metabolism have been reconstructed, but each represents only a subset of our knowledge. Here we describe Recon 2, a community-driven, consensus 'metabolic reconstruction', which is the most comprehensive representation of human metabolism that is applicable to computational modeling. Compared with its predecessors, the reconstruction has improved topological and functional features, including ∼2× more reactions and ∼1.7× more unique metabolites. Using Recon 2 we predicted changes in metabolite biomarkers for 49 inborn errors of metabolism with 77% accuracy when compared to experimental data. Mapping metabolomic data and drug information onto Recon 2 demonstrates its potential for integrating and analyzing diverse data types. Using protein expression data, we automatically generated a compendium of 65 cell type-specific models, providing a basis for manual curation or investigation of cell-specific metabolic properties. Recon 2 will facilitate many future biomedical studies and is freely available at http://humanmetabolism.org/.


Subject(s)
Databases, Protein , Metabolome/physiology , Models, Biological , Proteome/metabolism , Computer Simulation , Humans
3.
Mol Syst Biol ; 8: 558, 2012 Jun 26.
Article in English | MEDLINE | ID: mdl-22735334

ABSTRACT

Macrophages are central players in immune response, manifesting divergent phenotypes to control inflammation and innate immunity through release of cytokines and other signaling factors. Recently, the focus on metabolism has been reemphasized as critical signaling and regulatory pathways of human pathophysiology, ranging from cancer to aging, often converge on metabolic responses. Here, we used genome-scale modeling and multi-omics (transcriptomics, proteomics, and metabolomics) analysis to assess metabolic features that are critical for macrophage activation. We constructed a genome-scale metabolic network for the RAW 264.7 cell line to determine metabolic modulators of activation. Metabolites well-known to be associated with immunoactivation (glucose and arginine) and immunosuppression (tryptophan and vitamin D3) were among the most critical effectors. Intracellular metabolic mechanisms were assessed, identifying a suppressive role for de-novo nucleotide synthesis. Finally, underlying metabolic mechanisms of macrophage activation are identified by analyzing multi-omic data obtained from LPS-stimulated RAW cells in the context of our flux-based predictions. Our study demonstrates metabolism's role in regulating activation may be greater than previously anticipated and elucidates underlying connections between activation and metabolic effectors.


Subject(s)
Immunologic Factors/metabolism , Macrophage Activation/physiology , Metabolic Networks and Pathways/genetics , Adenosine Triphosphate/metabolism , Animals , Cell Line, Tumor , Glutamine/metabolism , Leukemia/pathology , Metabolic Networks and Pathways/immunology , Metabolomics , Mice , Models, Biological , Nitric Oxide/metabolism , Proteomics , Transcriptome
4.
J Biol Chem ; 286(36): 31522-31, 2011 Sep 09.
Article in English | MEDLINE | ID: mdl-21757732

ABSTRACT

The main kidney transporter of many commonly prescribed drugs (e.g. penicillins, diuretics, antivirals, methotrexate, and non-steroidal anti-inflammatory drugs) is organic anion transporter-1 (OAT1), originally identified as NKT (Lopez-Nieto, C. E., You, G., Bush, K. T., Barros, E. J., Beier, D. R., and Nigam, S. K. (1997) J. Biol. Chem. 272, 6471-6478). Targeted metabolomics in knockouts have shown that OAT1 mediates the secretion or reabsorption of many important metabolites, including intermediates in carbohydrate, fatty acid, and amino acid metabolism. This observation raises the possibility that OAT1 helps regulate broader metabolic activities. We therefore examined the potential roles of OAT1 in metabolic pathways using Recon 1, a functionally tested genome-scale reconstruction of human metabolism. A computational approach was used to analyze in vivo metabolomic as well as transcriptomic data from wild-type and OAT1 knock-out animals, resulting in the implication of several metabolic pathways, including the citric acid cycle, polyamine, and fatty acid metabolism. Validation by in vitro and ex vivo analysis using Xenopus oocyte, cell culture, and kidney tissue assays demonstrated interactions between OAT1 and key intermediates in these metabolic pathways, including previously unknown substrates, such as polyamines (e.g. spermine and spermidine). A genome-scale metabolic network reconstruction generated some experimentally supported predictions for metabolic pathways linked to OAT1-related transport. The data support the possibility that the SLC22 and other families of transporters, known to be expressed in many tissues and primarily known for drug and toxin clearance, are integral to a number of endogenous pathways and may be involved in a larger remote sensing and signaling system (Ahn, S. Y., and Nigam, S. K. (2009) Mol. Pharmacol. 76, 481-490, and Wu, W., Dnyanmote, A. V., and Nigam, S. K. (2011) Mol. Pharmacol. 79, 795-805). Drugs may alter metabolism by competing for OAT1 binding of metabolites.


Subject(s)
Metabolic Networks and Pathways , Metabolomics/methods , Organic Anion Transport Protein 1/metabolism , Animals , Cells, Cultured , Genome, Human , Genomics , Humans , Mice , Mice, Knockout , Organic Anion Transport Protein 1/deficiency , Pharmaceutical Preparations
5.
BMC Syst Biol ; 5: 8, 2011 Jan 18.
Article in English | MEDLINE | ID: mdl-21244678

ABSTRACT

BACKGROUND: Metabolic reconstructions (MRs) are common denominators in systems biology and represent biochemical, genetic, and genomic (BiGG) knowledge-bases for target organisms by capturing currently available information in a consistent, structured manner. Salmonella enterica subspecies I serovar Typhimurium is a human pathogen, causes various diseases and its increasing antibiotic resistance poses a public health problem. RESULTS: Here, we describe a community-driven effort, in which more than 20 experts in S. Typhimurium biology and systems biology collaborated to reconcile and expand the S. Typhimurium BiGG knowledge-base. The consensus MR was obtained starting from two independently developed MRs for S. Typhimurium. Key results of this reconstruction jamboree include i) development and implementation of a community-based workflow for MR annotation and reconciliation; ii) incorporation of thermodynamic information; and iii) use of the consensus MR to identify potential multi-target drug therapy approaches. CONCLUSION: Taken together, with the growing number of parallel MRs a structured, community-driven approach will be necessary to maximize quality while increasing adoption of MRs in experimental design and interpretation.


Subject(s)
Cooperative Behavior , Models, Biological , Salmonella typhimurium , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Databases, Factual , Genes, Bacterial/genetics , Humans , Metabolic Networks and Pathways , Salmonella typhimurium/drug effects , Salmonella typhimurium/genetics , Salmonella typhimurium/metabolism , Systems Biology
6.
BMC Syst Biol ; 3: 37, 2009 Mar 25.
Article in English | MEDLINE | ID: mdl-19321003

ABSTRACT

BACKGROUND: Metabolomics has emerged as a powerful tool in the quantitative identification of physiological and disease-induced biological states. Extracellular metabolome or metabolic profiling data, in particular, can provide an insightful view of intracellular physiological states in a noninvasive manner. RESULTS: We used an updated genome-scale metabolic network model of Saccharomyces cerevisiae, iMM904, to investigate how changes in the extracellular metabolome can be used to study systemic changes in intracellular metabolic states. The iMM904 metabolic network was reconstructed based on an existing genome-scale network, iND750, and includes 904 genes and 1,412 reactions. The network model was first validated by comparing 2,888 in silico single-gene deletion strain growth phenotype predictions to published experimental data. Extracellular metabolome data measured in response to environmental and genetic perturbations of ammonium assimilation pathways was then integrated with the iMM904 network in the form of relative overflow secretion constraints and a flux sampling approach was used to characterize candidate flux distributions allowed by these constraints. Predicted intracellular flux changes were consistent with published measurements on intracellular metabolite levels and fluxes. Patterns of predicted intracellular flux changes could also be used to correctly identify the regions of the metabolic network that were perturbed. CONCLUSION: Our results indicate that integrating quantitative extracellular metabolomic profiles in a constraint-based framework enables inferring changes in intracellular metabolic flux states. Similar methods could potentially be applied towards analyzing biofluid metabolome variations related to human physiological and disease states.


Subject(s)
Extracellular Space/metabolism , Intracellular Space/metabolism , Metabolomics/methods , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Amino Acids/metabolism , Gene Deletion , Genes, Fungal , Glutamate Dehydrogenase/metabolism , Metabolic Networks and Pathways , Models, Biological , Potassium/pharmacology , Quaternary Ammonium Compounds/pharmacology , Reproducibility of Results , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Systems Biology
7.
Trends Biotechnol ; 27(1): 37-44, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19010556

ABSTRACT

The intricate nature of human physiology renders its study a difficult undertaking, and a systems biology approach is necessary to understand the complex interactions involved. Network reconstruction is a key step in systems biology and represents a common denominator because all systems biology research on a target organism relies on such a representation. With the recent development of genome-scale human metabolic networks, metabolic systems analysis is now possible and has initiated a shift towards human systems biology. Here, we review the important aspects of reconstructing a bottom-up human metabolic network, the network's role in modeling human physiology and the necessity for a community-based consensus reconstruction of human metabolism to be established.


Subject(s)
Chromosome Mapping/methods , Gene Expression Regulation/physiology , Genome/physiology , Models, Biological , Proteome/metabolism , Signal Transduction/physiology , Systems Biology/methods , Computer Simulation , Humans
8.
Nat Biotechnol ; 26(10): 1155-60, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18846089

ABSTRACT

Genomic data allow the large-scale manual or semi-automated assembly of metabolic network reconstructions, which provide highly curated organism-specific knowledge bases. Although several genome-scale network reconstructions describe Saccharomyces cerevisiae metabolism, they differ in scope and content, and use different terminologies to describe the same chemical entities. This makes comparisons between them difficult and underscores the desirability of a consolidated metabolic network that collects and formalizes the 'community knowledge' of yeast metabolism. We describe how we have produced a consensus metabolic network reconstruction for S. cerevisiae. In drafting it, we placed special emphasis on referencing molecules to persistent databases or using database-independent forms, such as SMILES or InChI strings, as this permits their chemical structure to be represented unambiguously and in a manner that permits automated reasoning. The reconstruction is readily available via a publicly accessible database and in the Systems Biology Markup Language (http://www.comp-sys-bio.org/yeastnet). It can be maintained as a resource that serves as a common denominator for studying the systems biology of yeast. Similar strategies should benefit communities studying genome-scale metabolic networks of other organisms.


Subject(s)
Databases, Protein , Models, Biological , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Signal Transduction/physiology , Systems Biology/methods , Computer Simulation , Information Storage and Retrieval/methods , Systems Integration
9.
Mol Biosyst ; 3(9): 598-603, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17700859

ABSTRACT

The recent sequencing and annotation of the human genome enables a new era in biomedicine that will be based on an interdisciplinary, systemic approach to the elucidation and treatment of human disease. Reconstruction of genome-scale metabolic networks is an important part of this approach since networks represent the integration of diverse biological data such as genome annotations, high-throughput data, and legacy biochemical knowledge. This article will describe Homo sapiens Recon 1, a functionally tested, genome-scale reconstruction of human cellular metabolism, and its capabilities for facilitating the understanding of physiological and disease metabolic states.


Subject(s)
Computational Biology/methods , Genome, Human/genetics , Metabolic Networks and Pathways/genetics , Systems Biology/methods , Humans , Software
10.
Nat Protoc ; 2(3): 727-38, 2007.
Article in English | MEDLINE | ID: mdl-17406635

ABSTRACT

The manner in which microorganisms utilize their metabolic processes can be predicted using constraint-based analysis of genome-scale metabolic networks. Herein, we present the constraint-based reconstruction and analysis toolbox, a software package running in the Matlab environment, which allows for quantitative prediction of cellular behavior using a constraint-based approach. Specifically, this software allows predictive computations of both steady-state and dynamic optimal growth behavior, the effects of gene deletions, comprehensive robustness analyses, sampling the range of possible cellular metabolic states and the determination of network modules. Functions enabling these calculations are included in the toolbox, allowing a user to input a genome-scale metabolic model distributed in Systems Biology Markup Language format and perform these calculations with just a few lines of code. The results are predictions of cellular behavior that have been verified as accurate in a growing body of research. After software installation, calculation time is minimal, allowing the user to focus on the interpretation of the computational results.


Subject(s)
Cells/metabolism , Computational Biology/methods , Metabolic Networks and Pathways/physiology , Models, Biological , Software , Systems Biology/methods , Computer Simulation
11.
Proc Natl Acad Sci U S A ; 104(6): 1777-82, 2007 Feb 06.
Article in English | MEDLINE | ID: mdl-17267599

ABSTRACT

Metabolism is a vital cellular process, and its malfunction is a major contributor to human disease. Metabolic networks are complex and highly interconnected, and thus systems-level computational approaches are required to elucidate and understand metabolic genotype-phenotype relationships. We have manually reconstructed the global human metabolic network based on Build 35 of the genome annotation and a comprehensive evaluation of >50 years of legacy data (i.e., bibliomic data). Herein we describe the reconstruction process and demonstrate how the resulting genome-scale (or global) network can be used (i) for the discovery of missing information, (ii) for the formulation of an in silico model, and (iii) as a structured context for analyzing high-throughput biological data sets. Our comprehensive evaluation of the literature revealed many gaps in the current understanding of human metabolism that require future experimental investigation. Mathematical analysis of network structure elucidated the implications of intracellular compartmentalization and the potential use of correlated reaction sets for alternative drug target identification. Integrated analysis of high-throughput data sets within the context of the reconstruction enabled a global assessment of functional metabolic states. These results highlight some of the applications enabled by the reconstructed human metabolic network. The establishment of this network represents an important step toward genome-scale human systems biology.


Subject(s)
Computer Simulation , Gene Expression Profiling , Genome, Human/physiology , Metabolism/genetics , Systems Biology , Computational Biology , Gastric Bypass , Humans/metabolism , Metabolism/physiology , Muscle, Skeletal/metabolism , Muscle, Skeletal/surgery
SELECTION OF CITATIONS
SEARCH DETAIL
...