Your browser doesn't support javascript.
loading
Common biochemical properties of metabolic genes recurrently dysregulated in tumors.
Oruganty, Krishnadev; Campit, Scott Edward; Mamde, Sainath; Lyssiotis, Costas A; Chandrasekaran, Sriram.
Afiliação
  • Oruganty K; 1Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105 USA.
  • Campit SE; Present Address: Genpact, New York, NY 10036 USA.
  • Mamde S; 3Program in Chemical Biology, University of Michigan, Ann Arbor, MI 48105 USA.
  • Lyssiotis CA; 1Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105 USA.
  • Chandrasekaran S; 3Program in Chemical Biology, University of Michigan, Ann Arbor, MI 48105 USA.
Cancer Metab ; 8: 5, 2020.
Article em En | MEDLINE | ID: mdl-32411371
ABSTRACT

BACKGROUND:

Tumor initiation and progression are associated with numerous metabolic alterations. However, the biochemical drivers and constraints that contribute to metabolic gene dysregulation are unclear.

METHODS:

Here, we present MetOncoFit, a computational model that integrates 142 metabolic features that can impact tumor fitness, including enzyme catalytic activity, pathway association, network topology, and reaction flux. MetOncoFit uses genome-scale metabolic modeling and machine-learning to quantify the relative importance of various metabolic features in predicting cancer metabolic gene expression, copy number variation, and survival data.

RESULTS:

Using MetOncoFit, we performed a meta-analysis of 9 cancer types and over 4500 samples from TCGA, Prognoscan, and COSMIC tumor databases. MetOncoFit accurately predicted enzyme differential expression and its impact on patient survival using the 142 attributes of metabolic enzymes. Our analysis revealed that enzymes with high catalytic activity were frequently upregulated in many tumors and associated with poor survival. Topological analysis also identified specific metabolites that were hot spots of dysregulation.

CONCLUSIONS:

MetOncoFit integrates a broad range of datasets to understand how biochemical and topological features influence metabolic gene dysregulation across various cancer types. MetOncoFit was able to achieve significantly higher accuracy in predicting differential expression, copy number variation, and patient survival than traditional modeling approaches. Overall, MetOncoFit illuminates how enzyme activity and metabolic network architecture influences tumorigenesis.
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Idioma: En Ano de publicação: 2020 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Idioma: En Ano de publicação: 2020 Tipo de documento: Article