Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Biochem ; 86: 277-304, 2017 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-28654323

RESUMEN

Metabolites are the small biological molecules involved in energy conversion and biosynthesis. Studying metabolism is inherently challenging due to metabolites' reactivity, structural diversity, and broad concentration range. Herein, we review the common pitfalls encountered in metabolomics and provide concrete guidelines for obtaining accurate metabolite measurements, focusing on water-soluble primary metabolites. We show how seemingly straightforward sample preparation methods can introduce systematic errors (e.g., owing to interconversion among metabolites) and how proper selection of quenching solvent (e.g., acidic acetonitrile:methanol:water) can mitigate such problems. We discuss the specific strengths, pitfalls, and best practices for each common analytical platform: liquid chromatography-mass spectrometry (LC-MS), gas chromatography-mass spectrometry (GC-MS), nuclear magnetic resonance (NMR), and enzyme assays. Together this information provides a pragmatic knowledge base for carrying out biologically informative metabolite measurements.


Asunto(s)
Cromatografía Liquida/normas , Cromatografía de Gases y Espectrometría de Masas/normas , Espectroscopía de Resonancia Magnética/normas , Espectrometría de Masas/normas , Metabolómica/normas , Adenosina Trifosfato/análisis , Animales , Glutatión/análisis , Guías como Asunto , Humanos , Microextracción en Fase Líquida/métodos , Metabolómica/instrumentación , Metabolómica/métodos , Ratones , NADP/análisis , Solventes
2.
Nature ; 614(7947): 349-357, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36725930

RESUMEN

Tissues derive ATP from two pathways-glycolysis and the tricarboxylic acid (TCA) cycle coupled to the electron transport chain. Most energy in mammals is produced via TCA metabolism1. In tumours, however, the absolute rates of these pathways remain unclear. Here we optimize tracer infusion approaches to measure the rates of glycolysis and the TCA cycle in healthy mouse tissues, Kras-mutant solid tumours, metastases and leukaemia. Then, given the rates of these two pathways, we calculate total ATP synthesis rates. We find that TCA cycle flux is suppressed in all five primary solid tumour models examined and is increased in lung metastases of breast cancer relative to primary orthotopic tumours. As expected, glycolysis flux is increased in tumours compared with healthy tissues (the Warburg effect2,3), but this increase is insufficient to compensate for low TCA flux in terms of ATP production. Thus, instead of being hypermetabolic, as commonly assumed, solid tumours generally produce ATP at a slower than normal rate. In mouse pancreatic cancer, this is accommodated by the downregulation of protein synthesis, one of this tissue's major energy costs. We propose that, as solid tumours develop, cancer cells shed energetically expensive tissue-specific functions, enabling uncontrolled growth despite a limited ability to produce ATP.


Asunto(s)
Adenosina Trifosfato , Neoplasias de la Mama , Ciclo del Ácido Cítrico , Desaceleración , Neoplasias Pulmonares , Metástasis de la Neoplasia , Neoplasias Pancreáticas , Animales , Ratones , Adenosina Trifosfato/biosíntesis , Adenosina Trifosfato/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Ciclo del Ácido Cítrico/fisiología , Metabolismo Energético , Glucólisis , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Especificidad de Órganos , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Biosíntesis de Proteínas
3.
Nature ; 565(7737): E3, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30523330

RESUMEN

In this Letter, 'released' should have been 'regulated' in the sentence starting: 'Deletion of Atg5 in the host similarly regulated circulating arginine and suppressed tumorigenesis...' This has been corrected online.

4.
Nat Methods ; 18(11): 1377-1385, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34711973

RESUMEN

Liquid chromatography-high-resolution mass spectrometry (LC-MS)-based metabolomics aims to identify and quantify all metabolites, but most LC-MS peaks remain unidentified. Here we present a global network optimization approach, NetID, to annotate untargeted LC-MS metabolomics data. The approach aims to generate, for all experimentally observed ion peaks, annotations that match the measured masses, retention times and (when available) tandem mass spectrometry fragmentation patterns. Peaks are connected based on mass differences reflecting adduction, fragmentation, isotopes, or feasible biochemical transformations. Global optimization generates a single network linking most observed ion peaks, enhances peak assignment accuracy, and produces chemically informative peak-peak relationships, including for peaks lacking tandem mass spectrometry spectra. Applying this approach to yeast and mouse data, we identified five previously unrecognized metabolites (thiamine derivatives and N-glucosyl-taurine). Isotope tracer studies indicate active flux through these metabolites. Thus, NetID applies existing metabolomic knowledge and global optimization to substantially improve annotation coverage and accuracy in untargeted metabolomics datasets, facilitating metabolite discovery.


Asunto(s)
Algoritmos , Curaduría de Datos/normas , Hígado/metabolismo , Metaboloma , Metabolómica/normas , Saccharomyces cerevisiae/metabolismo , Animales , Cromatografía Liquida/métodos , Curaduría de Datos/métodos , Metabolómica/métodos , Ratones , Espectrometría de Masas en Tándem/métodos
5.
Nature ; 563(7732): 569-573, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30429607

RESUMEN

Autophagy captures intracellular components and delivers them to lysosomes, where they are degraded and recycled to sustain metabolism and to enable survival during starvation1-5. Acute, whole-body deletion of the essential autophagy gene Atg7 in adult mice causes a systemic metabolic defect that manifests as starvation intolerance and gradual loss of white adipose tissue, liver glycogen and muscle mass1. Cancer cells also benefit from autophagy. Deletion of essential autophagy genes impairs the metabolism, proliferation, survival and malignancy of spontaneous tumours in models of autochthonous cancer6,7. Acute, systemic deletion of Atg7 or acute, systemic expression of a dominant-negative ATG4b in mice induces greater regression of KRAS-driven cancers than does tumour-specific autophagy deletion, which suggests that host autophagy promotes tumour growth1,8. Here we show that host-specific deletion of Atg7 impairs the growth of multiple allografted tumours, although not all tumour lines were sensitive to host autophagy status. Loss of autophagy in the host was associated with a reduction in circulating arginine, and the sensitive tumour cell lines were arginine auxotrophs owing to the lack of expression of the enzyme argininosuccinate synthase 1. Serum proteomic analysis identified the arginine-degrading enzyme arginase I (ARG1) in the circulation of Atg7-deficient hosts, and in vivo arginine metabolic tracing demonstrated that serum arginine was degraded to ornithine. ARG1 is predominantly expressed in the liver and can be released from hepatocytes into the circulation. Liver-specific deletion of Atg7 produced circulating ARG1, and reduced both serum arginine and tumour growth. Deletion of Atg5 in the host similarly regulated [corrected] circulating arginine and suppressed tumorigenesis, which demonstrates that this phenotype is specific to autophagy function rather than to deletion of Atg7. Dietary supplementation of Atg7-deficient hosts with arginine partially restored levels of circulating arginine and tumour growth. Thus, defective autophagy in the host leads to the release of ARG1 from the liver and the degradation of circulating arginine, which is essential for tumour growth; this identifies a metabolic vulnerability of cancer.


Asunto(s)
Arginina/sangre , Autofagia , Neoplasias/sangre , Neoplasias/patología , Aloinjertos , Animales , Arginasa/sangre , Arginasa/metabolismo , Arginina/administración & dosificación , Arginina/farmacología , Autofagia/genética , Proteína 5 Relacionada con la Autofagia/deficiencia , Proteína 5 Relacionada con la Autofagia/genética , Proteína 7 Relacionada con la Autofagia/deficiencia , Proteína 7 Relacionada con la Autofagia/genética , Proteína 7 Relacionada con la Autofagia/metabolismo , Carcinogénesis/efectos de los fármacos , Carcinogénesis/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Suplementos Dietéticos , Hepatocitos/enzimología , Hepatocitos/metabolismo , Hígado/enzimología , Masculino , Ratones , Trasplante de Neoplasias , Neoplasias/genética , Ornitina/metabolismo
6.
Anal Chem ; 95(40): 14879-14888, 2023 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-37756255

RESUMEN

Detection of small molecule metabolites (SMM), particularly those involved in energy metabolism using MALDI-mass spectrometry imaging (MSI), is challenging due to factors including ion suppression from other analytes present (e.g., proteins and lipids). One potential solution to enhance SMM detection is to remove analytes that cause ion suppression from tissue sections before matrix deposition through solvent washes. Here, we systematically investigated solvent treatment conditions to improve SMM signal and preserve metabolite localization. Washing with acidic methanol significantly enhances the detection of phosphate-containing metabolites involved in energy metabolism. The improved detection is due to removing lipids and highly polar metabolites that cause ion suppression and denaturing proteins that release bound phosphate-containing metabolites. Stable isotope infusions of [13C6]nicotinamide coupled to MALDI-MSI ("Iso-imaging") in the kidney reveal patterns that indicate blood vessels, medulla, outer stripe, and cortex. We also observed different ATP:ADP raw signals across mouse kidney regions, consistent with regional differences in glucose metabolism favoring either gluconeogenesis or glycolysis. In mouse muscle, Iso-imaging using [13C6]glucose shows high glycolytic flux from infused circulating glucose in type 1 and 2a fibers (soleus) and relatively lower glycolytic flux in type 2b fiber type (gastrocnemius). Thus, improved detection of phosphate-containing metabolites due to acidic methanol treatment combined with isotope tracing provides an improved way to probe energy metabolism with spatial resolution in vivo.


Asunto(s)
Glucólisis , Metanol , Ratones , Animales , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Glucosa , Lípidos , Solventes , Isótopos , Fosfatos , Rayos Láser
7.
PLoS Biol ; 18(8): e3000757, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32833957

RESUMEN

In eukaryotes, conserved mechanisms ensure that cell growth is coordinated with nutrient availability. Overactive growth during nutrient limitation ("nutrient-growth dysregulation") can lead to rapid cell death. Here, we demonstrate that cells can adapt to nutrient-growth dysregulation by evolving major metabolic defects. Specifically, when yeast lysine-auxotrophic mutant lys- encountered lysine limitation, an evolutionarily novel stress, cells suffered nutrient-growth dysregulation. A subpopulation repeatedly evolved to lose the ability to synthesize organosulfurs (lys-orgS-). Organosulfurs, mainly reduced glutathione (GSH) and GSH conjugates, were released by lys- cells during lysine limitation when growth was dysregulated, but not during glucose limitation when growth was regulated. Limiting organosulfurs conferred a frequency-dependent fitness advantage to lys-orgS- by eliciting a proper slow growth program, including autophagy. Thus, nutrient-growth dysregulation is associated with rapid organosulfur release, which enables the selection of organosulfur auxotrophy to better tune cell growth to the metabolic environment. We speculate that evolutionarily novel stresses can trigger atypical release of certain metabolites, setting the stage for the evolution of new ecological interactions.


Asunto(s)
Adaptación Fisiológica/genética , Lisina/farmacología , Redes y Vías Metabólicas/efectos de los fármacos , Nutrientes/farmacología , Saccharomyces cerevisiae/metabolismo , Autofagia/efectos de los fármacos , Autofagia/genética , Evolución Biológica , Glucosa/metabolismo , Glucosa/farmacología , Lisina/deficiencia , Redes y Vías Metabólicas/genética , Nitrógeno/metabolismo , Nitrógeno/farmacología , Nutrientes/metabolismo , Ribosomas/efectos de los fármacos , Ribosomas/metabolismo , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crecimiento & desarrollo , Sirolimus/farmacología , Estrés Fisiológico
8.
Nature ; 551(7678): 115-118, 2017 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-29045397

RESUMEN

Mammalian tissues are fuelled by circulating nutrients, including glucose, amino acids, and various intermediary metabolites. Under aerobic conditions, glucose is generally assumed to be burned fully by tissues via the tricarboxylic acid cycle (TCA cycle) to carbon dioxide. Alternatively, glucose can be catabolized anaerobically via glycolysis to lactate, which is itself also a potential nutrient for tissues and tumours. The quantitative relevance of circulating lactate or other metabolic intermediates as fuels remains unclear. Here we systematically examine the fluxes of circulating metabolites in mice, and find that lactate can be a primary source of carbon for the TCA cycle and thus of energy. Intravenous infusions of 13C-labelled nutrients reveal that, on a molar basis, the circulatory turnover flux of lactate is the highest of all metabolites and exceeds that of glucose by 1.1-fold in fed mice and 2.5-fold in fasting mice; lactate is made primarily from glucose but also from other sources. In both fed and fasted mice, 13C-lactate extensively labels TCA cycle intermediates in all tissues. Quantitative analysis reveals that during the fasted state, the contribution of glucose to tissue TCA metabolism is primarily indirect (via circulating lactate) in all tissues except the brain. In genetically engineered lung and pancreatic cancer tumours in fasted mice, the contribution of circulating lactate to TCA cycle intermediates exceeds that of glucose, with glutamine making a larger contribution than lactate in pancreatic cancer. Thus, glycolysis and the TCA cycle are uncoupled at the level of lactate, which is a primary circulating TCA substrate in most tissues and tumours.


Asunto(s)
Ciclo del Ácido Cítrico , Glucosa/metabolismo , Ácido Láctico/sangre , Ácido Láctico/metabolismo , Animales , Glucemia/metabolismo , Encéfalo/metabolismo , Carbono/sangre , Carbono/metabolismo , Ayuno/sangre , Ayuno/metabolismo , Glutamina/sangre , Glutamina/metabolismo , Glucólisis , Ratones , Músculos/metabolismo , Neoplasias Pancreáticas/sangre , Neoplasias Pancreáticas/metabolismo
9.
J Am Chem Soc ; 144(51): 23633-23641, 2022 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-36525649

RESUMEN

Sialylation, the addition of sialic acid to glycans, is a crucial post-translational modification of proteins, contributing to neurodevelopment, oncogenesis, and immune response. In cancer, sialylation is dramatically upregulated. Yet, the functional biochemical consequences of sialylation remain mysterious. Here, we establish a µMap proximity labeling platform that utilizes metabolically inserted azidosialic acid to introduce iridium-based photocatalysts on sialylated cell-surface glycoproteins as a means to profile local microenvironments across the sialylated proteome. In comparative experiments between primary cervical cells and a cancerous cell line (HeLa), we identify key differences in both the global sialome and proximal proteins, including solute carrier proteins that regulate metabolite and ion transport. In particular, we show that cell-surface interactions between receptors trafficking ethanolamine and zinc are sialylation-dependent and impact intracellular metabolite levels. These results establish a µMap method for interrogating proteoglycan function and support a role for sialylated glycoproteins in regulating cell-surface transporters.


Asunto(s)
Glicoproteínas , Ácido N-Acetilneuramínico , Humanos , Glicoproteínas/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Glicoproteínas de Membrana/metabolismo , Membrana Celular/metabolismo , Transporte Iónico , Polisacáridos/metabolismo
10.
J Immunol ; 204(2): 459-471, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31826940

RESUMEN

Specific deletion of the tumor suppressor TRAF3 from B lymphocytes in mice leads to the prolonged survival of mature B cells and expanded B cell compartments in secondary lymphoid organs. In the current study, we investigated the metabolic basis of TRAF3-mediated regulation of B cell survival by employing metabolomic, lipidomic, and transcriptomic analyses. We compared the polar metabolites, lipids, and metabolic enzymes of resting splenic B cells purified from young adult B cell-specific Traf3 -/- and littermate control mice. We found that multiple metabolites, lipids, and enzymes regulated by TRAF3 in B cells are clustered in the choline metabolic pathway. Using stable isotope labeling, we demonstrated that phosphocholine and phosphatidylcholine biosynthesis was markedly elevated in Traf3 -/- mouse B cells and decreased in TRAF3-reconstituted human multiple myeloma cells. Furthermore, pharmacological inhibition of choline kinase α, an enzyme that catalyzes phosphocholine synthesis and was strikingly increased in Traf3 -/- B cells, substantially reversed the survival phenotype of Traf3 -/- B cells both in vitro and in vivo. Taken together, our results indicate that enhanced phosphocholine and phosphatidylcholine synthesis supports the prolonged survival of Traf3 -/- B lymphocytes. Our findings suggest that TRAF3-regulated choline metabolism has diagnostic and therapeutic value for B cell malignancies with TRAF3 deletions or relevant mutations.


Asunto(s)
Linfocitos B/fisiología , Colina Quinasa/metabolismo , Colina/metabolismo , Mieloma Múltiple/metabolismo , Factor 3 Asociado a Receptor de TNF/metabolismo , Animales , Línea Celular , Supervivencia Celular , Colina Quinasa/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Noqueados , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Mutación/genética , Fosforilcolina/metabolismo , Transducción de Señal , Factor 3 Asociado a Receptor de TNF/genética
11.
Genes Dev ; 27(13): 1447-61, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23824538

RESUMEN

Macroautophagy (autophagy hereafter) degrades and recycles proteins and organelles to support metabolism and survival in starvation. Oncogenic Ras up-regulates autophagy, and Ras-transformed cell lines require autophagy for mitochondrial function, stress survival, and engrafted tumor growth. Here, the essential autophagy gene autophagy-related-7 (atg7) was deleted concurrently with K-ras(G12D) activation in mouse models for non-small-cell lung cancer (NSCLC). atg7-deficient tumors accumulated dysfunctional mitochondria and prematurely induced p53 and proliferative arrest, which reduced tumor burden that was partly relieved by p53 deletion. atg7 loss altered tumor fate from adenomas and carcinomas to oncocytomas-rare, predominantly benign tumors characterized by the accumulation of defective mitochondria. Surprisingly, lipid accumulation occurred in atg7-deficient tumors only when p53 was deleted. atg7- and p53-deficient tumor-derived cell lines (TDCLs) had compromised starvation survival and formed lipidic cysts instead of tumors, suggesting defective utilization of lipid stores. atg7 deficiency reduced fatty acid oxidation (FAO) and increased sensitivity to FAO inhibition, indicating that with p53 loss, Ras-driven tumors require autophagy for mitochondrial function and lipid catabolism. Thus, autophagy is required for carcinoma fate, and autophagy defects may be a molecular basis for the occurrence of oncocytomas. Moreover, cancers require autophagy for distinct roles in metabolism that are oncogene- and tumor suppressor gene-specific.


Asunto(s)
Adenoma Oxifílico/fisiopatología , Autofagia , Carcinoma de Pulmón de Células no Pequeñas/fisiopatología , Genes ras/fisiología , Metabolismo de los Lípidos , Neoplasias Pulmonares/fisiopatología , Animales , Línea Celular Tumoral , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica , Genes p53/genética , Homeostasis , Longevidad/genética , Ratones , Mitocondrias/patología , Células Tumorales Cultivadas
12.
Am J Hum Genet ; 101(4): 489-502, 2017 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-28942964

RESUMEN

Genome-wide association studies have identified a signal at the SLC22A1 locus for serum acylcarnitines, intermediate metabolites of mitochondrial oxidation whose plasma levels associate with metabolic diseases. Here, we refined the association signal, performed conditional analyses, and examined the linkage structure to find coding variants of SLC22A1 that mediate independent association signals at the locus. We also employed allele-specific expression analysis to find potential regulatory variants of SLC22A1 and demonstrated the effect of one variant on the splicing of SLC22A1. SLC22A1 encodes a hepatic plasma membrane transporter whose role in acylcarnitine physiology has not been described. By targeted metabolomics and isotope tracing experiments in loss- and gain-of-function cell and mouse models of Slc22a1, we uncovered a role of SLC22A1 in the efflux of acylcarnitines from the liver to the circulation. We further validated the impacts of human variants on SLC22A1-mediated acylcarnitine efflux in vitro, explaining their association with serum acylcarnitine levels. Our findings provide the detailed molecular mechanisms of the GWAS association for serum acylcarnitines at the SLC22A1 locus by functionally validating the impact of SLC22A1 and its variants on acylcarnitine transport.


Asunto(s)
Carnitina/análogos & derivados , Regulación de la Expresión Génica , Hígado/metabolismo , Enfermedades Metabólicas/genética , Transportador 1 de Catión Orgánico/genética , Polimorfismo de Nucleótido Simple , Alelos , Empalme Alternativo , Animales , Transporte Biológico , Sistemas CRISPR-Cas , Carnitina/sangre , Carnitina/farmacocinética , Células Cultivadas , Estudios de Cohortes , Femenino , Estudio de Asociación del Genoma Completo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Enfermedades Metabólicas/sangre , Enfermedades Metabólicas/metabolismo , Metabolómica , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Transportador 1 de Catión Orgánico/antagonistas & inhibidores , Transportador 1 de Catión Orgánico/metabolismo , Distribución Tisular
13.
Anal Chem ; 92(17): 11573-11581, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32614575

RESUMEN

Annotation of untargeted high-resolution full-scan LC-MS metabolomics data remains challenging due to individual metabolites generating multiple LC-MS peaks arising from isotopes, adducts, and fragments. Adduct annotation is a particular challenge, as the same mass difference between peaks can arise from adduct formation, fragmentation, or different biological species. To address this, here we describe a buffer modification workflow (BMW) in which the same sample is run by LC-MS in both liquid chromatography solvent with 14NH3-acetate buffer and in solvent with the buffer modified with 15NH3-formate. Buffer switching results in characteristic mass and signal intensity changes for adduct peaks, facilitating their annotation. This relatively simple and convenient chromatography modification annotated yeast metabolomics data with similar effectiveness to growing the yeast in isotope-labeled media. Application to mouse liver data annotated both known metabolite and known adduct peaks with 95% accuracy. Overall, it identified 26% of ∼27 000 liver LC-MS features as putative metabolites, of which ∼2600 showed HMDB or KEGG database formula match. This workflow is well suited to biological samples that cannot be readily isotope labeled, including plants, mammalian tissues, and tumors.


Asunto(s)
Metabolómica/métodos , Espectrometría de Masas en Tándem/métodos , Acetatos/química , Aminas/química , Animales , Tampones (Química) , Cromatografía Liquida , Bases de Datos Factuales , Femenino , Formiatos/química , Marcaje Isotópico , Hígado/metabolismo , Extractos Hepáticos/química , Ratones Endogámicos C57BL , Saccharomyces cerevisiae/metabolismo , Solventes/química
14.
Anal Chem ; 91(3): 1838-1846, 2019 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-30586294

RESUMEN

Untargeted metabolomics can detect more than 10 000 peaks in a single LC-MS run. The correspondence between these peaks and metabolites, however, remains unclear. Here, we introduce a Peak Annotation and Verification Engine (PAVE) for annotating untargeted microbial metabolomics data. The workflow involves growing cells in 13C and 15N isotope-labeled media to identify peaks from biological compounds and their carbon and nitrogen atom counts. Improved deisotoping and deadducting are enabled by algorithms that integrate positive mode, negative mode, and labeling data. To distinguish metabolites and their fragments, PAVE experimentally measures the response of each peak to weak in-source collision induced dissociation, which increases the peak intensity for fragments while decreasing it for their parent ions. The molecular formulas of the putative metabolites are then assigned based on database searching using both m/ z and C/N atom counts. Application of this procedure to Saccharomyces cerevisiae and Escherichia coli revealed that more than 80% of peaks do not label, i.e., are environmental contaminants. More than 70% of the biological peaks are isotopic variants, adducts, fragments, or mass spectrometry artifacts yielding ∼2000 apparent metabolites across the two organisms. About 650 match to a known metabolite formula based on m/ z and C/N atom counts, with 220 assigned structures based on MS/MS and/or retention time to match to authenticated standards. Thus, PAVE enables systematic annotation of LC-MS metabolomics data with only ∼4% of peaks annotated as apparent metabolites.


Asunto(s)
Escherichia coli/metabolismo , Metabolómica , Saccharomyces cerevisiae/metabolismo , Algoritmos , Cromatografía Liquida , Bases de Datos Factuales , Marcaje Isotópico , Espectrometría de Masas en Tándem
15.
Circ Res ; 121(12): 1370-1378, 2017 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-28928113

RESUMEN

RATIONALE: Pregnancy profoundly alters maternal physiology. The heart hypertrophies during pregnancy, but its metabolic adaptations, are not well understood. OBJECTIVE: To determine the mechanisms underlying cardiac substrate use during pregnancy. METHODS AND RESULTS: We use here 13C glucose, 13C lactate, and 13C fatty acid tracing analyses to show that hearts in late pregnant mice increase fatty acid uptake and oxidation into the tricarboxylic acid cycle, while reducing glucose and lactate oxidation. Mitochondrial quantity, morphology, and function do not seem altered. Insulin signaling seems intact, and the abundance and localization of the major fatty acid and glucose transporters, CD36 (cluster of differentiation 36) and GLUT4 (glucose transporter type 4), are also unchanged. Rather, we find that the pregnancy hormone progesterone induces PDK4 (pyruvate dehydrogenase kinase 4) in cardiomyocytes and that elevated PDK4 levels in late pregnancy lead to inhibition of PDH (pyruvate dehydrogenase) and pyruvate flux into the tricarboxylic acid cycle. Blocking PDK4 reverses the metabolic changes seen in hearts in late pregnancy. CONCLUSIONS: Taken together, these data indicate that the hormonal environment of late pregnancy promotes metabolic remodeling in the heart at the level of PDH, rather than at the level of insulin signaling.


Asunto(s)
Miocardio/metabolismo , Embarazo/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Ácido Pirúvico/metabolismo , Animales , Ciclo del Ácido Cítrico , Ácidos Grasos/metabolismo , Femenino , Glucosa/metabolismo , Ácido Láctico/metabolismo , Ratones , Ratones Endogámicos C57BL , Progesterona/metabolismo , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora
16.
Am J Physiol Regul Integr Comp Physiol ; 314(1): R22-R33, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-28978515

RESUMEN

Perinatal exposures are associated with altered risks of childhood allergy. Human studies and our previous work suggest that restricted growth in utero (IUGR) is protective against allergic disease. The mechanisms are not clearly defined, but reduced fetal abundance and altered metabolism of methyl donors are hypothesized as possible underlying mechanisms. Therefore, we examined whether late-gestation maternal dietary methyl donor and cofactor supplementation of the placentally restricted (PR) sheep pregnancy would reverse allergic protection in progeny. Allergic outcomes were compared between progeny from control pregnancies (CON; n = 49), from PR pregnancies without intervention (PR; n = 28), and from PR pregnancies where the dam was fed a methyl donor plus cofactor supplement from day 120 of pregnancy until delivery (PR + Methyl; n = 25). Both PR and PR + Methyl progeny were smaller than CON; supplementation did not alter birth size. PR was protective against cutaneous hypersensitivity responses to ovalbumin (OVA; P < 0.01 in singletons). Cutaneous hypersensitivity responses to OVA in PR + Methyl progeny were intermediate to and not different from the responses of CON and PR sheep. Cutaneous hypersensitivity responses to house dust mites did not differ between treatments. In singleton progeny, upper dermal mast cell density was greater in PR + Methyl than in PR or CON (each P < 0.05). The differences in the cutaneous allergic response were not explained by treatment effects on circulating immune cells or antibodies. Our results suggest that mechanisms underlying in utero programming of allergic susceptibility by IUGR and methyl donor availability may differ and imply that late-gestation methyl donor supplementation may increase allergy risk.


Asunto(s)
Cobalto/administración & dosificación , Dermatitis/prevención & control , Suplementos Dietéticos , Retardo del Crecimiento Fetal/inmunología , Ácido Fólico/administración & dosificación , Hipersensibilidad/prevención & control , Metionina/administración & dosificación , Efectos Tardíos de la Exposición Prenatal , Azufre/administración & dosificación , Animales , Metilación de ADN , Dermatitis/inmunología , Modelos Animales de Enfermedad , Femenino , Edad Gestacional , Hipersensibilidad/inmunología , Inmunoglobulina E/inmunología , Mastocitos/inmunología , Ovalbúmina/inmunología , Placenta/inmunología , Embarazo , Pyroglyphidae/inmunología , Oveja Doméstica , Piel/inmunología
17.
Anal Chem ; 89(11): 5940-5948, 2017 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-28471646

RESUMEN

Orbitraps are high-resolution ion-trap mass spectrometers that are widely used in metabolomics. While the mass accuracy and resolving power of orbitraps have been extensively documented, their spectral accuracy, i.e., accuracy in measuring the abundances of isotopic peaks, remains less studied. In analyzing spectra of unlabeled metabolites, we discovered a systematic under representation of heavier natural isotopic species, especially for high molecular weight metabolites (∼20% under-measurement of [M + 1]/[M + 0] ratio at m/z 600). We hypothesize that these discrepancies arise for metabolites far from the lower limit of the mass scan range, due to the weaker containment in the C-trap that results in suboptimal trajectories inside the Orbitrap analyzer. Consistent with this, spectral fidelity was restored by dividing the mass scan range (initially 75 m/z to 1000 m/z) into two scan events, one for lower molecular weight and the other for higher molecular weight metabolites. Having thus obtained accurate mass spectra at high resolution, we found that natural isotope correction for high-resolution labeling data requires more sophisticated algorithms than typically employed: the correction algorithm must take into account whether isotopologues with the same nominal mass are resolved. We present an algorithm and associated open-source code, named AccuCor, for this purpose. Together, these improvements in instrument parameters and natural isotope correction enable more accurate measurement of metabolite labeling and thus metabolic flux.


Asunto(s)
Algoritmos , Espectrometría de Masas/métodos , Metabolómica/métodos , Marcaje Isotópico/normas , Isótopos , Espectrometría de Masas/normas , Metabolómica/normas , Peso Molecular
18.
Anal Bioanal Chem ; 409(25): 5955-5964, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28799108

RESUMEN

The cofactor tetrahydrofolate (THF) is used to reduce, oxidize, and transfer one-carbon (1C) units required for the synthesis of nucleotides, glycine, and methionine. Measurement of intracellular THF species is complicated by their chemical instability, signal dilution caused by variable polyglutamation, and the potential for interconversion among these species. Here, we describe a method using negative mode liquid chromatography-mass spectrometry (LC-MS) to measure intracellular folate species from mammalian cells. Application of this method with isotope-labeled substrates revealed abiotic interconversion of THF and methylene-THF, which renders their separate quantitation particularly challenging. Chemical reduction of methylene-THF using deuterated sodium cyanoborohydride traps methylene-THF, which is unstable, as deuterated 5-methyl-THF, which is stable. Together with proper sample handling and LC-MS, this enables effective measurements of five active folate pools (THF, 5-methyl-THF, methylene-THF, methenyl-THF/10-formyl-THF, and 5-formyl-THF) representing the biologically important 1C oxidation states of THF in mammalian cells. Graphical abstract Chemical derivatization with deuterated cyanoborohydride traps unstable methylene-THF as isotope-labeled 5-methyl-THF, enabling accurate quantification by LC-MS.


Asunto(s)
Cromatografía Liquida/métodos , Leucovorina/análisis , Espectrometría de Masas/métodos , Tetrahidrofolatos/análisis , Técnicas de Cultivo de Célula , Antagonistas del Ácido Fólico/farmacología , Células HEK293 , Humanos , Leucovorina/metabolismo , Metotrexato/farmacología , Tetrahidrofolatos/metabolismo
19.
Anal Chem ; 87(4): 2273-81, 2015 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-25591916

RESUMEN

Liquid chromatography-mass spectrometry (LC-MS) technology allows for rapid quantitation of cellular metabolites, with metabolites identified by mass spectrometry and chromatographic retention time. Recently, with the development of rapid scanning high-resolution high accuracy mass spectrometers and the desire for high throughput screening, minimal or no chromatographic separation has become increasingly popular. When analyzing complex cellular extracts, however, the lack of chromatographic separation could potentially result in misannotation of structurally related metabolites. Here, we show that, even using electrospray ionization, a soft ionization method, in-source fragmentation generates unwanted byproducts of identical mass to common metabolites. For example, nucleotide-triphosphates generate nucleotide-diphosphates, and hexose-phosphates generate triose-phosphates. We evaluated yeast intracellular metabolite extracts and found more than 20 cases of in-source fragments that mimic common metabolites. Accordingly, chromatographic separation is required for accurate quantitation of many common cellular metabolites.


Asunto(s)
Hexosas/análisis , Metabolómica , Nucleótidos/análisis , Fosfatos/análisis , Cromatografía Liquida , Hexosas/metabolismo , Espectrometría de Masas , Nucleótidos/metabolismo , Fosfatos/metabolismo
20.
Mol Syst Biol ; 9: 665, 2013 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-23670538

RESUMEN

Nucleotide degradation is a universal metabolic capability. Here we combine metabolomics, genetics and biochemistry to characterize the yeast pathway. Nutrient starvation, via PKA, AMPK/SNF1, and TOR, triggers autophagic breakdown of ribosomes into nucleotides. A protein not previously associated with nucleotide degradation, Phm8, converts nucleotide monophosphates into nucleosides. Downstream steps, which involve the purine nucleoside phosphorylase, Pnp1, and pyrimidine nucleoside hydrolase, Urh1, funnel ribose into the nonoxidative pentose phosphate pathway. During carbon starvation, the ribose-derived carbon accumulates as sedoheptulose-7-phosphate, whose consumption by transaldolase is impaired due to depletion of transaldolase's other substrate, glyceraldehyde-3-phosphate. Oxidative stress increases glyceraldehyde-3-phosphate, resulting in rapid consumption of sedoheptulose-7-phosphate to make NADPH for antioxidant defense. Ablation of Phm8 or double deletion of Pnp1 and Urh1 prevent effective nucleotide salvage, resulting in metabolite depletion and impaired survival of starving yeast. Thus, ribose salvage provides means of surviving nutrient starvation and oxidative stress.


Asunto(s)
Regulación Fúngica de la Expresión Génica , N-Glicosil Hidrolasas/genética , Nucleótidos/metabolismo , Purina-Nucleósido Fosforilasa/genética , Ribosa/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Gliceraldehído 3-Fosfato/metabolismo , N-Glicosil Hidrolasas/deficiencia , NADP/metabolismo , Vía de Pentosa Fosfato/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Purina-Nucleósido Fosforilasa/deficiencia , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Transducción de Señal , Estrés Fisiológico/genética , Fosfatos de Azúcar , Transaldolasa/genética , Transaldolasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA