Quantitation of cellular metabolic fluxes of methionine.

Methionine is an essential proteogenic amino acid. In addition, it is a methyl donor for DNA and protein methylation and a propylamine donor for polyamine biosynthesis. Both the methyl and propylamine donation pathways involve metabolic cycles, and methods are needed to quantitate these cycles. Here, we describe an analytical approach for quantifying methionine metabolic fluxes that accounts for the mixing of intracellular and extracellular methionine pools. We observe that such mixing prevents isotope tracing experiments from reaching the steady state due to the large size of the media pools and hence precludes the use of standard stationary metabolic flux analysis. Our approach is based on feeding cells with (13)C methionine and measuring the isotope-labeling kinetics of both intracellular and extracellular methionine by liquid chromatography-mass spectrometry (LC-MS). We apply this method to quantify methionine metabolism in a human fibrosarcoma cell line and study how methionine salvage pathway enzyme methylthioadenosine phosphorylase (MTAP), frequently deleted in cancer, affects methionine metabolism. We find that both transmethylation and propylamine transfer fluxes amount to roughly 15% of the net methionine uptake, with no major changes due to MTAP deletion. Our method further enables the quantification of flux through the pro-tumorigenic enzyme ornithine decarboxylase, and this flux increases 2-fold following MTAP deletion. The analytical approach used to quantify methionine metabolic fluxes is applicable for other metabolic systems affected by mixing of intracellular and extracellular metabolite pools.

[1]  Jamey D. Young,et al.  Isotopically nonstationary 13C metabolic flux analysis. , 2013, Methods in molecular biology.

[2]  J. Testa,et al.  Increasing the therapeutic index of 5-fluorouracil and 6-thioguanine by targeting loss of MTAP in tumor cells , 2012, Cancer biology & therapy.

[3]  Abhishek K. Jha,et al.  Functional genomics reveal that the serine synthesis pathway is essential in breast cancer , 2011, Nature.

[4]  J. Bertino,et al.  Targeting tumors that lack methylthioadenosine phosphorylase (MTAP) activity , 2011, Cancer biology & therapy.

[5]  Uri Alon,et al.  Proteome Half-Life Dynamics in Living Human Cells , 2011, Science.

[6]  Nicola Zamboni,et al.  13C metabolic flux analysis in complex systems. , 2011, Current opinion in biotechnology.

[7]  Bin Wang,et al.  Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of α-ketoglutarate-dependent dioxygenases. , 2011, Cancer cell.

[8]  J. R. Peterson,et al.  Chemical Genetic Screening for Compounds That Preferentially Inhibit Growth of Methylthioadenosine Phosphorylase (MTAP)–Deficient Saccharomyces cerevisiae , 2011, Journal of biomolecular screening.

[9]  G. Evans,et al.  Growth and Metastases of Human Lung Cancer Are Inhibited in Mouse Xenografts by a Transition State Analogue of 5′-Methylthioadenosine Phosphorylase* , 2010, The Journal of Biological Chemistry.

[10]  Joshua D Rabinowitz,et al.  Metabolomic analysis and visualization engine for LC-MS data. , 2010, Analytical chemistry.

[11]  Elizabeth L. Johnson,et al.  Quiescent Fibroblasts Exhibit High Metabolic Activity , 2010, PLoS biology.

[12]  Daniel Amador-Noguez,et al.  Metabolomic analysis via reversed-phase ion-pairing liquid chromatography coupled to a stand alone orbitrap mass spectrometer. , 2010, Analytical chemistry.

[13]  B. Pfeifer,et al.  Metabolic flux analysis and pharmaceutical production. , 2010, Metabolic engineering.

[14]  D. Ruffieux,et al.  Determination of dansylated polyamines in red blood cells by liquid chromatography-tandem mass spectrometry. , 2009, Analytical biochemistry.

[15]  E. Papoutsakis,et al.  Aldehyde–alcohol dehydrogenase and/or thiolase overexpression coupled with CoA transferase downregulation lead to higher alcohol titers and selectivity in Clostridium acetobutylicum fermentations , 2009, Biotechnology and bioengineering.

[16]  Xiao-Jiang Feng,et al.  Systems-level metabolic flux profiling identifies fatty acid synthesis as a target for antiviral therapy , 2008, Nature Biotechnology.

[17]  J. Rabinowitz,et al.  Analytical strategies for LC-MS-based targeted metabolomics. , 2008, Journal of chromatography. B, Analytical technologies in the biomedical and life sciences.

[18]  J. Rabinowitz,et al.  Kinetic flux profiling for quantitation of cellular metabolic fluxes , 2008, Nature Protocols.

[19]  J. Rabinowitz,et al.  Absolute quantitation of intracellular metabolite concentrations by an isotope ratio-based approach , 2008, Nature Protocols.

[20]  G. Stephanopoulos,et al.  Metabolic flux analysis in a nonstationary system: fed-batch fermentation of a high yielding strain of E. coli producing 1,3-propanediol. , 2007, Metabolic engineering.

[21]  G. Stephanopoulos,et al.  Elementary metabolite units (EMU): a novel framework for modeling isotopic distributions. , 2007, Metabolic engineering.

[22]  U. Sauer,et al.  Article number: 62 REVIEW Metabolic networks in motion: 13 C-based flux analysis , 2022 .

[23]  Wolfgang Wiechert,et al.  Experimental design principles for isotopically instationary 13C labeling experiments , 2006, Biotechnology and bioengineering.

[24]  O. Mcconnell,et al.  Enhanced chromatographic resolution of amine enantiomers as carbobenzyloxy derivatives in high-performance liquid chromatography and supercritical fluid chromatography. , 2005, Journal of chromatography. A.

[25]  S. Baylin,et al.  DNA methylation and gene silencing in cancer , 2005, Nature Clinical Practice Oncology.

[26]  Joachim Kopka,et al.  Metabolome analysis: the potential of in vivo labeling with stable isotopes for metabolite profiling. , 2005, Trends in biotechnology.

[27]  E. Gerner,et al.  Polyamines and cancer: old molecules, new understanding , 2004, Nature Reviews Cancer.

[28]  S. Burgess,et al.  Glucose production, gluconeogenesis, and hepatic tricarboxylic acid cycle fluxes measured by nuclear magnetic resonance analysis of a single glucose derivative. , 2004, Analytical biochemistry.

[29]  J J Heijnen,et al.  MIRACLE: mass isotopomer ratio analysis of U‐13C‐labeled extracts. A new method for accurate quantification of changes in concentrations of intracellular metabolites , 2004, Biotechnology and bioengineering.

[30]  P. Diegelman,et al.  Methylthioadenosine Phosphorylase Regulates Ornithine Decarboxylase by Production of Downstream Metabolites* , 2003, Journal of Biological Chemistry.

[31]  P. Diegelman,et al.  Methylthioadenosine phosphorylase, a gene frequently codeleted with p16(cdkN2a/ARF), acts as a tumor suppressor in a breast cancer cell line. , 2002, Cancer research.

[32]  B. Tang,et al.  Defects in methylthioadenosine phosphorylase are associated with but not responsible for methionine-dependent tumor cell growth. , 2000, Cancer research.

[33]  W. Wiechert,et al.  Bidirectional reaction steps in metabolic networks: III. Explicit solution and analysis of isotopomer labeling systems. , 1999, Biotechnology and bioengineering.

[34]  Peter W. Laird,et al.  THE ROLE OF DNA METHYLATION IN CANCER GENETICS AND EPIGENETICS , 1996 .

[35]  J L Cleveland,et al.  The ornithine decarboxylase gene is a transcriptional target of c-Myc. , 1993, Proceedings of the National Academy of Sciences of the United States of America.

[36]  F. Della Ragione,et al.  Effect of analogues of 5'-methylthioadenosine on cellular metabolism. Inactivation of S-adenosylhomocysteine hydrolase by 5'-isobutylthioadenosine. , 1983, The Biochemical journal.

[37]  A. Vandenbark,et al.  Inactivation of S-adenosylhomocysteine hydrolase by 5'-deoxy-5'-methylthioadenosine. , 1981, Biochemical and biophysical research communications.

[38]  R. Pajula,et al.  Methylthioadenosine, a potent inhibitor of spermine synthase from bovine brain , 1979, FEBS letters.