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.
PLoS Comput Biol ; 20(2): e1011919, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38422168

ABSTRACT

Improvements in the diagnosis and treatment of cancer have revealed long-term side effects of chemotherapeutics, particularly cardiotoxicity. Here, we present paired transcriptomics and metabolomics data characterizing in vitro cardiotoxicity to three compounds: 5-fluorouracil, acetaminophen, and doxorubicin. Standard gene enrichment and metabolomics approaches identify some commonly affected pathways and metabolites but are not able to readily identify metabolic adaptations in response to cardiotoxicity. The paired data was integrated with a genome-scale metabolic network reconstruction of the heart to identify shifted metabolic functions, unique metabolic reactions, and changes in flux in metabolic reactions in response to these compounds. Using this approach, we confirm previously seen changes in the p53 pathway by doxorubicin and RNA synthesis by 5-fluorouracil, we find evidence for an increase in phospholipid metabolism in response to acetaminophen, and we see a shift in central carbon metabolism suggesting an increase in metabolic demand after treatment with doxorubicin and 5-fluorouracil.


Subject(s)
Acetaminophen , Cardiotoxicity , Humans , Cardiotoxicity/metabolism , Metabolomics , Doxorubicin/pharmacology , Gene Expression Profiling , Fluorouracil/pharmacology
2.
Sci Rep ; 11(1): 5535, 2021 03 10.
Article in English | MEDLINE | ID: mdl-33692370

ABSTRACT

Lung cancer rates are rising globally and non-small cell lung cancer (NSCLC) has a five year survival rate of only 24%. Unfortunately, the development of drugs to treat cancer is severely hampered by the inefficiency of translating pre-clinical studies into clinical benefit. Thus, we sought to apply a tumor microenvironment system (TMES) to NSCLC. Using microvascular endothelial cells, lung cancer derived fibroblasts, and NSCLC tumor cells in the presence of in vivo tumor-derived hemodynamic flow and transport, we demonstrate that the TMES generates an in-vivo like biological state and predicts drug response to EGFR inhibitors. Transcriptomic and proteomic profiling indicate that the TMES recapitulates the in vivo and patient molecular biological state providing a mechanistic rationale for the predictive nature of the TMES. This work further validates the TMES for modeling patient tumor biology and drug response indicating utility of the TMES as a predictive tool for drug discovery and development and potential for use as a system for patient avatars.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Endothelial Cells/metabolism , Lung Neoplasms/metabolism , Models, Biological , Tumor Microenvironment , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Endothelial Cells/pathology , Humans , Lung Neoplasms/pathology , Mice , Mice, Nude , Mice, SCID
3.
Cell Rep ; 34(10): 108836, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33691118

ABSTRACT

In diseased states, the heart can shift to use different carbon substrates, measured through changes in uptake of metabolites by imaging methods or blood metabolomics. However, it is not known whether these measured changes are a result of transcriptional changes or external factors. Here, we explore transcriptional changes in late-stage heart failure using publicly available data integrated with a model of heart metabolism. First, we present a heart-specific genome-scale metabolic network reconstruction (GENRE), iCardio. Next, we demonstrate the utility of iCardio in interpreting heart failure gene expression data by identifying tasks inferred from differential expression (TIDEs), which represent metabolic functions associated with changes in gene expression. We identify decreased gene expression for nitric oxide (NO) and N-acetylneuraminic acid (Neu5Ac) synthesis as common metabolic markers of heart failure. The methods presented here for constructing a tissue-specific model and identifying TIDEs can be extended to multiple tissues and diseases of interest.


Subject(s)
Heart Failure/genetics , Metabolic Networks and Pathways/genetics , Models, Biological , Myocardium/metabolism , Databases, Protein , Heart Failure/pathology , Humans , Metabolomics/methods , N-Acetylneuraminic Acid/metabolism , Nitric Oxide/metabolism , Severity of Illness Index
4.
Toxicol Appl Pharmacol ; 412: 115390, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33387578

ABSTRACT

The kidneys are metabolically active organs with importance in several physiological tasks such as the secretion of soluble wastes into the urine and synthesizing glucose and oxidizing fatty acids for energy in fasting (non-fed) conditions. Once damaged, the metabolic capability of the kidneys becomes altered. Here, we define metabolic tasks in a computational modeling framework to capture kidney function in an update to the iRno network reconstruction of rat metabolism using literature-based evidence. To demonstrate the utility of iRno for predicting kidney function, we exposed primary rat renal proximal tubule epithelial cells to four compounds with varying levels of nephrotoxicity (acetaminophen, gentamicin, 2,3,7,8-tetrachlorodibenzodioxin, and trichloroethylene) for six and twenty-four hours, and collected transcriptomics and metabolomics data to measure the metabolic effects of compound exposure. For the transcriptomics data, we observed changes in fatty acid metabolism and amino acid metabolism, as well as changes in existing markers of kidney function such as Clu (clusterin). The iRno metabolic network reconstruction was used to predict alterations in these same pathways after integrating transcriptomics data and was able to distinguish between select compound-specific effects on the proximal tubule epithelial cells. Genome-scale metabolic network reconstructions with coupled omics data can be used to predict changes in metabolism as a step towards identifying novel metabolic biomarkers of kidney function and dysfunction.


Subject(s)
Energy Metabolism/drug effects , Epithelial Cells/drug effects , Kidney Diseases/chemically induced , Kidney Tubules, Proximal/drug effects , Metabolome/drug effects , Transcriptome/drug effects , Acetaminophen/toxicity , Animals , Cells, Cultured , Databases, Genetic , Energy Metabolism/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Gene Expression Profiling , Gene Regulatory Networks , Gentamicins/toxicity , Kidney Diseases/genetics , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Metabolome/genetics , Metabolomics , Polychlorinated Dibenzodioxins/toxicity , Rats, Sprague-Dawley , Trichloroethylene/toxicity
5.
Toxicol Sci ; 172(2): 279-291, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31501904

ABSTRACT

Context-specific GEnome-scale metabolic Network REconstructions (GENREs) provide a means to understand cellular metabolism at a deeper level of physiological detail. Here, we use transcriptomics data from chemically-exposed rat hepatocytes to constrain a GENRE of rat hepatocyte metabolism and predict biomarkers of liver toxicity using the Transcriptionally Inferred Metabolic Biomarker Response algorithm. We profiled alterations in cellular hepatocyte metabolism following in vitro exposure to four toxicants (acetaminophen, carbon tetrachloride, 2,3,7,8-tetrachlorodibenzodioxin, and trichloroethylene) for six hour. TIMBR predictions were compared with paired fresh and spent media metabolomics data from the same exposure conditions. Agreement between computational model predictions and experimental data led to the identification of specific metabolites and thus metabolic pathways associated with toxicant exposure. Here, we identified changes in the TCA metabolites citrate and alpha-ketoglutarate along with changes in carbohydrate metabolism and interruptions in ATP production and the TCA Cycle. Where predictions and experimental data disagreed, we identified testable hypotheses to reconcile differences between the model predictions and experimental data. The presented pipeline for using paired transcriptomics and metabolomics data provides a framework for interrogating multiple omics datasets to generate mechanistic insight of metabolic changes associated with toxicological responses.


Subject(s)
Activation, Metabolic/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Metabolic Networks and Pathways/drug effects , Transcriptome/drug effects , Acetaminophen/toxicity , Activation, Metabolic/genetics , Animals , Biomarkers/metabolism , Carbon Tetrachloride/toxicity , Cells, Cultured , Computational Biology , Gene Expression Profiling , Male , Metabolic Networks and Pathways/genetics , Metabolomics , Polychlorinated Dibenzodioxins/toxicity , Primary Cell Culture , Rats, Sprague-Dawley , Trichloroethylene/toxicity
6.
Toxicol Appl Pharmacol ; 372: 19-32, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30974156

ABSTRACT

Acetaminophen (APAP) is the most commonly used analgesic and antipyretic drug in the world. Yet, it poses a major risk of liver injury when taken in excess of the therapeutic dose. Current clinical markers do not detect the early onset of liver injury associated with excess APAP-information that is vital to reverse injury progression through available therapeutic interventions. Hence, several studies have used transcriptomics, proteomics, and metabolomics technologies, both independently and in combination, in an attempt to discover potential early markers of liver injury. However, the casual relationship between these observations and their relation to the APAP mechanism of liver toxicity are not clearly understood. Here, we used Sprague-Dawley rats orally gavaged with a single dose of 2 g/kg of APAP to collect tissue samples from the liver and kidney for transcriptomic analysis and plasma and urine samples for metabolomic analysis. We developed and used a multi-tissue, metabolism-based modeling approach to integrate these data, characterize the effect of excess APAP levels on liver metabolism, and identify a panel of plasma and urine metabolites that are associated with APAP-induced liver toxicity. Our analyses, which indicated that pathways involved in nucleotide-, lipid-, and amino acid-related metabolism in the liver were most strongly affected within 10 h following APAP treatment, identified a list of potential metabolites in these pathways that could serve as plausible markers of APAP-induced liver injury. Our approach identifies toxicant-induced changes in endogenous metabolism, is applicable to other toxicants based on transcriptomic data, and provides a mechanistic framework for interpreting metabolite alterations.


Subject(s)
Acetaminophen , Chemical and Drug Induced Liver Injury/diagnosis , Liver/metabolism , Metabolomics , Animals , Biomarkers/blood , Biomarkers/urine , Chemical and Drug Induced Liver Injury/blood , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/urine , Disease Models, Animal , Early Diagnosis , Male , Predictive Value of Tests , Rats, Sprague-Dawley , Time Factors
7.
Comput Biol Med ; 105: 64-71, 2019 02.
Article in English | MEDLINE | ID: mdl-30584952

ABSTRACT

GEnome-scale Network REconstructions (GENREs) mathematically describe metabolic reactions of an organism or a specific cell type. GENREs can be used with a number of constraint-based reconstruction and analysis (COBRA) methods to make computational predictions on how a system changes in different environments. We created a simplified GENRE (referred to as iSIM) that captures central energy metabolism with nine metabolic reactions to illustrate the use of and promote the understanding of GENREs and constraint-based methods. We demonstrate the simulation of single and double gene deletions, flux variability analysis (FVA), and test a number of metabolic tasks with the GENRE. Code to perform these analyses is provided in Python, R, and MATLAB. Finally, with iSIM as a guide, we demonstrate how inaccuracies in GENREs can limit their use in the interrogation of energy metabolism.


Subject(s)
Metabolic Flux Analysis , Metabolic Networks and Pathways/physiology , Models, Biological , Animals , Humans
8.
Nat Commun ; 8: 14250, 2017 02 08.
Article in English | MEDLINE | ID: mdl-28176778

ABSTRACT

The laboratory rat has been used as a surrogate to study human biology for more than a century. Here we present the first genome-scale network reconstruction of Rattus norvegicus metabolism, iRno, and a significantly improved reconstruction of human metabolism, iHsa. These curated models comprehensively capture metabolic features known to distinguish rats from humans including vitamin C and bile acid synthesis pathways. After reconciling network differences between iRno and iHsa, we integrate toxicogenomics data from rat and human hepatocytes, to generate biomarker predictions in response to 76 drugs. We validate comparative predictions for xanthine derivatives with new experimental data and literature-based evidence delineating metabolite biomarkers unique to humans. Our results provide mechanistic insights into species-specific metabolism and facilitate the selection of biomarkers consistent with rat and human biology. These models can serve as powerful computational platforms for contextualizing experimental data and making functional predictions for clinical and basic science applications.


Subject(s)
Computational Biology/methods , Metabolic Networks and Pathways/genetics , Models, Biological , Species Specificity , Toxicogenetics/methods , Animals , Biomarkers/analysis , Biomarkers/metabolism , Datasets as Topic , Gene Expression Profiling , Hepatocytes/metabolism , Humans , Metabolomics/methods , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...