Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Article in English | MEDLINE | ID: mdl-31139575

ABSTRACT

Little is known about the metabolic state of Mycobacterium tuberculosis (Mtb) inside the phagosome, a compartment inside phagocytes for killing pathogens and other foreign substances. We have developed a combined model of Mtb and human metabolism, sMtb-RECON and used this model to predict the metabolic state of Mtb during infection of the host. Amino acids are predicted to be used for energy production as well as biomass formation. Subsequently we assessed the effect of increasing dosages of drugs targeting metabolism on the metabolic state of the pathogen and predict resulting metabolic adaptations and flux rerouting through various pathways. In particular, the TCA cycle becomes more important upon drug application, as well as alanine, aspartate, glutamate, proline, arginine and porphyrin metabolism, while glycine, serine, and threonine metabolism become less important. We modeled the effect of 11 metabolically active drugs. Notably, the effect of eight could be recreated and two major profiles of the metabolic state were predicted. The profiles of the metabolic states of Mtb affected by the drugs BTZ043, cycloserine and its derivative terizidone, ethambutol, ethionamide, propionamide, and isoniazid were very similar, while TMC207 is predicted to have quite a different effect on metabolism as it inhibits ATP synthase and therefore indirectly interferes with a multitude of metabolic pathways.


Subject(s)
Antitubercular Agents/pharmacology , Host-Pathogen Interactions/drug effects , Metabolic Networks and Pathways/drug effects , Models, Biological , Mycobacterium tuberculosis/metabolism , Adenosine Triphosphatases/drug effects , Amides/pharmacology , Amino Acids/metabolism , Cycloserine/pharmacology , Diarylquinolines/pharmacology , Drug Tolerance/physiology , Ethambutol/pharmacology , Ethionamide/pharmacology , Gene Expression Profiling , Gene Expression Regulation, Bacterial/drug effects , Humans , Isoniazid/pharmacology , Isoxazoles/pharmacology , Mycobacterium bovis/drug effects , Mycobacterium bovis/genetics , Mycobacterium bovis/growth & development , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/growth & development , Oxazolidinones/pharmacology , Spiro Compounds/pharmacology , Thiazines/pharmacology , Tuberculosis/microbiology
2.
Biochim Biophys Acta Mol Basis Dis ; 1865(2): 360-370, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30385409

ABSTRACT

Flavin adenine dinucleotide (FAD) and its precursor flavin mononucleotide (FMN) are redox cofactors that are required for the activity of more than hundred human enzymes. Mutations in the genes encoding these proteins cause severe phenotypes, including a lack of energy supply and accumulation of toxic intermediates. Ideally, patients should be diagnosed before they show symptoms so that treatment and/or preventive care can start immediately. This can be achieved by standardized newborn screening tests. However, many of the flavin-related diseases lack appropriate biomarker profiles. Genome-scale metabolic models can aid in biomarker research by predicting altered profiles of potential biomarkers. Unfortunately, current models, including the most recent human metabolic reconstructions Recon and HMR, typically treat enzyme-bound flavins incorrectly as free metabolites. This in turn leads to artificial degrees of freedom in pathways that are strictly coupled. Here, we present a reconstruction of human metabolism with a curated and extended flavoproteome. To illustrate the functional consequences, we show that simulations with the curated model - unlike simulations with earlier Recon versions - correctly predict the metabolic impact of multiple-acyl-CoA-dehydrogenase deficiency as well as of systemic flavin-depletion. Moreover, simulations with the new model allowed us to identify a larger number of biomarkers in flavoproteome-related diseases, without loss of accuracy. We conclude that adequate inclusion of cofactors in constraint-based modelling contributes to higher precision in computational predictions.


Subject(s)
Coenzymes/metabolism , Flavoproteins/metabolism , Genome, Human , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/metabolism , Adenosine Triphosphate/metabolism , Biomarkers/metabolism , Flavin-Adenine Dinucleotide/deficiency , Flavin-Adenine Dinucleotide/metabolism , Humans , Models, Biological , Proteome/metabolism
3.
Article in English | MEDLINE | ID: mdl-30123778

ABSTRACT

Genome-scale metabolic models of Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis, have been envisioned as a platform for drug discovery. By systematically probing the networks that underpin such models, the reactions that are essential for Mtb are identified. A majority of these reactions are catalyzed by enzymes and thus represent candidate drug targets to fight an Mtb infection. Nevertheless, this is complicated by the limited knowledge on the environment that Mtb encounters during infection. Modeling the behavior of the bacteria during infection requires knowledge of the so-called biomass reaction that represents bacterial biomass composition. This composition varies in different environments or bacterial growth phases. Accurate modeling of the metabolic state requires a precise biomass reaction for the described condition. In recent years, additional insights in the in-host environment occupied by Mtb have been gained as transcript abundance data of interacting host and pathogen have become available. Therefore, we used transcript abundance data and developed a straightforward and systematic method to obtain a condition-specific biomass reaction for Mtb during in vitro growth and during infection of its host. The method described herein is virtually free of any pre-set assumptions on uptake rates of nutrients, making it suitable for exploring environments with limited accessibility. The condition-specific biomass reaction represents the "metabolic objective" of Mtb in a given environment (in-host growth and growth on defined medium) at a specific time point, and as such allows modeling the bacterial metabolic state in these environments. Five different biomass reactions were used to predict nutrient uptake rates and gene essentiality. Predictions were subsequently compared to available experimental data. Our results show that nutrient uptake can accurately be predicted. Gene essentiality can also be predicted but accurate predictions remain difficult to obtain. In conclusion, a viable strategy to model Mtb metabolism in hard-to-access environments that is virtually free of pre-set assumptions is provided.


Subject(s)
Mycobacterium tuberculosis/growth & development , Mycobacterium tuberculosis/metabolism , Tuberculosis/microbiology , Computer Simulation , Gene Expression Profiling , Models, Biological
4.
BMC Genomics ; 16: 34, 2015 Feb 05.
Article in English | MEDLINE | ID: mdl-25649146

ABSTRACT

BACKGROUND: The human pathogen Mycobacterium tuberculosis has the capacity to escape eradication by professional phagocytes. During infection, M. tuberculosis resists the harsh environment of phagosomes and actively manipulates macrophages and dendritic cells to ensure prolonged intracellular survival. In contrast to other intracellular pathogens, it has remained difficult to capture the transcriptome of mycobacteria during infection due to an unfavorable host-to-pathogen ratio. RESULTS: We infected the human macrophage-like cell line THP-1 with the attenuated M. tuberculosis surrogate M. bovis Bacillus Calmette-Guérin (M. bovis BCG). Mycobacterial RNA was up to 1000-fold underrepresented in total RNA preparations of infected host cells. We employed microbial enrichment combined with specific ribosomal RNA depletion to simultaneously analyze the transcriptional responses of host and pathogen during infection by dual RNA sequencing. Our results confirm that mycobacterial pathways for cholesterol degradation and iron acquisition are upregulated during infection. In addition, genes involved in the methylcitrate cycle, aspartate metabolism and recycling of mycolic acids were induced. In response to M. bovis BCG infection, host cells upregulated de novo cholesterol biosynthesis presumably to compensate for the loss of this metabolite by bacterial catabolism. CONCLUSIONS: Dual RNA sequencing allows simultaneous capture of the global transcriptome of host and pathogen, during infection. However, mycobacteria remained problematic due to their relatively low number per host cell resulting in an unfavorable bacterium-to-host RNA ratio. Here, we use a strategy that combines enrichment for bacterial transcripts and dual RNA sequencing to provide the most comprehensive transcriptome of intracellular mycobacteria to date. The knowledge acquired into the pathogen and host pathways regulated during infection may contribute to a solid basis for the deployment of novel intervention strategies to tackle infection.


Subject(s)
Cholesterol/biosynthesis , Host-Pathogen Interactions/genetics , Mycobacterium tuberculosis/genetics , Tuberculosis/genetics , Animals , Cattle , Cholesterol/genetics , Dendritic Cells/metabolism , Dendritic Cells/microbiology , Gene Expression Regulation, Bacterial/drug effects , High-Throughput Nucleotide Sequencing , Humans , Macrophages/microbiology , Mycobacterium bovis/pathogenicity , Mycobacterium tuberculosis/pathogenicity , Phagocytes/metabolism , Phagocytes/microbiology , Transcriptome/drug effects , Tuberculosis/microbiology
5.
Semin Immunol ; 26(6): 610-22, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25453232

ABSTRACT

Systems-level metabolic network reconstructions and the derived constraint-based (CB) mathematical models are efficient tools to explore bacterial metabolism. Approximately one-fourth of the Mycobacterium tuberculosis (Mtb) genome contains genes that encode proteins directly involved in its metabolism. These represent potential drug targets that can be systematically probed with CB models through the prediction of genes essential (or the combination thereof) for the pathogen to grow. However, gene essentiality depends on the growth conditions and, so far, no in vitro model precisely mimics the host at the different stages of mycobacterial infection, limiting model predictions. These limitations can be circumvented by combining expression data from in vivo samples with a validated CB model, creating an accurate description of pathogen metabolism in the host. To this end, we present here a thoroughly curated and extended genome-scale CB metabolic model of Mtb quantitatively validated using 13C measurements. We describe some of the efforts made in integrating CB models and high-throughput data to generate condition specific models, and we will discuss challenges ahead. This knowledge and the framework herein presented will enable to identify potential new drug targets, and will foster the development of optimal therapeutic strategies.


Subject(s)
Gene Expression Regulation, Bacterial , Genome, Bacterial , Metabolic Networks and Pathways/genetics , Models, Statistical , Mycobacterium tuberculosis/metabolism , Antitubercular Agents/therapeutic use , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Carbon Isotopes , Drug Resistance, Multiple, Bacterial/genetics , Gene Regulatory Networks , Host-Pathogen Interactions , Humans , Molecular Targeted Therapy , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/genetics , Systems Biology , Tuberculosis, Multidrug-Resistant/drug therapy , Tuberculosis, Multidrug-Resistant/metabolism , Tuberculosis, Multidrug-Resistant/microbiology , Tuberculosis, Multidrug-Resistant/pathology , Tuberculosis, Pulmonary/drug therapy , Tuberculosis, Pulmonary/metabolism , Tuberculosis, Pulmonary/microbiology , Tuberculosis, Pulmonary/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...