Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Nature ; 614(7947): 349-357, 2023 02.
Article in English | MEDLINE | ID: mdl-36725930

ABSTRACT

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.


Subject(s)
Adenosine Triphosphate , Breast Neoplasms , Citric Acid Cycle , Deceleration , Lung Neoplasms , Neoplasm Metastasis , Pancreatic Neoplasms , Animals , Mice , Adenosine Triphosphate/biosynthesis , Adenosine Triphosphate/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Citric Acid Cycle/physiology , Energy Metabolism , Glycolysis , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Organ Specificity , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Protein Biosynthesis
2.
Nat Methods ; 19(2): 223-230, 2022 02.
Article in English | MEDLINE | ID: mdl-35132243

ABSTRACT

Isotope tracing has helped to determine the metabolic activities of organs. Methods to probe metabolic heterogeneity within organs are less developed. We couple stable-isotope-labeled nutrient infusion to matrix-assisted laser desorption ionization imaging mass spectrometry (iso-imaging) to quantitate metabolic activity in mammalian tissues in a spatially resolved manner. In the kidney, we visualize gluconeogenic flux and glycolytic flux in the cortex and medulla, respectively. Tricarboxylic acid cycle substrate usage differs across kidney regions; glutamine and citrate are used preferentially in the cortex and fatty acids are used in the medulla. In the brain, we observe spatial gradations in carbon inputs to the tricarboxylic acid cycle and glutamate under a ketogenic diet. In a carbohydrate-rich diet, glucose predominates throughout but in a ketogenic diet, 3-hydroxybutyrate contributes most strongly in the hippocampus and least in the midbrain. Brain nitrogen sources also vary spatially; branched-chain amino acids contribute most in the midbrain, whereas ammonia contributes in the thalamus. Thus, iso-imaging can reveal the spatial organization of metabolic activity.


Subject(s)
Brain/metabolism , Carbon Isotopes/pharmacokinetics , Kidney/metabolism , Nitrogen Isotopes/pharmacokinetics , Animals , Diet , Enzymes , Gluconeogenesis , Glutamic Acid/biosynthesis , Glycolysis , Male , Mice, Inbred C57BL , Molecular Imaging , Single-Cell Analysis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tandem Mass Spectrometry , Tricarboxylic Acids/metabolism , Workflow
3.
Anal Chem ; 95(40): 14879-14888, 2023 10 10.
Article in English | MEDLINE | ID: mdl-37756255

ABSTRACT

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.


Subject(s)
Glycolysis , Methanol , Mice , Animals , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Glucose , Lipids , Solvents , Isotopes , Phosphates , Lasers
4.
Nat Chem Biol ; 16(7): 731-739, 2020 07.
Article in English | MEDLINE | ID: mdl-32393898

ABSTRACT

Glucose is catabolized by two fundamental pathways, glycolysis to make ATP and the oxidative pentose phosphate pathway to make reduced nicotinamide adenine dinucleotide phosphate (NADPH). The first step of the oxidative pentose phosphate pathway is catalyzed by the enzyme glucose-6-phosphate dehydrogenase (G6PD). Here we develop metabolite reporter and deuterium tracer assays to monitor cellular G6PD activity. Using these, we show that the most widely cited G6PD antagonist, dehydroepiandosterone, does not robustly inhibit G6PD in cells. We then identify a small molecule (G6PDi-1) that more effectively inhibits G6PD. Across a range of cultured cells, G6PDi-1 depletes NADPH most strongly in lymphocytes. In T cells but not macrophages, G6PDi-1 markedly decreases inflammatory cytokine production. In neutrophils, it suppresses respiratory burst. Thus, we provide a cell-active small molecule tool for oxidative pentose phosphate pathway inhibition, and use it to identify G6PD as a pharmacological target for modulating immune response.


Subject(s)
Enzyme Inhibitors/pharmacology , Glucosephosphate Dehydrogenase/antagonists & inhibitors , Lymphocytes/drug effects , Macrophages/drug effects , Neutrophils/drug effects , Pentose Phosphate Pathway/drug effects , Animals , Cell Line , Dehydroepiandrosterone/pharmacology , Dose-Response Relationship, Drug , Enzyme Assays , Glucose/metabolism , Glucosephosphate Dehydrogenase/immunology , Glucosephosphate Dehydrogenase/metabolism , Glycolysis/immunology , HCT116 Cells , Hep G2 Cells , Humans , Immunity, Innate , Lymphocyte Activation/drug effects , Lymphocytes/cytology , Lymphocytes/enzymology , Lymphocytes/immunology , Macrophage Activation/drug effects , Macrophages/cytology , Macrophages/enzymology , Macrophages/immunology , NADP/antagonists & inhibitors , NADP/metabolism , Neutrophils/cytology , Neutrophils/enzymology , Neutrophils/immunology , Pentose Phosphate Pathway/immunology
5.
EMBO J ; 34(2): 138-53, 2015 Jan 13.
Article in English | MEDLINE | ID: mdl-25476451

ABSTRACT

Recent studies link changes in energy metabolism with the fate of pluripotent stem cells (PSCs). Safe use of PSC derivatives in regenerative medicine requires an enhanced understanding and control of factors that optimize in vitro reprogramming and differentiation protocols. Relative shifts in metabolism from naïve through "primed" pluripotent states to lineage-directed differentiation place variable demands on mitochondrial biogenesis and function for cell types with distinct energetic and biosynthetic requirements. In this context, mitochondrial respiration, network dynamics, TCA cycle function, and turnover all have the potential to influence reprogramming and differentiation outcomes. Shifts in cellular metabolism affect enzymes that control epigenetic configuration, which impacts chromatin reorganization and gene expression changes during reprogramming and differentiation. Induced PSCs (iPSCs) may have utility for modeling metabolic diseases caused by mutations in mitochondrial DNA, for which few disease models exist. Here, we explore key features of PSC energy metabolism research in mice and man and the impact this work is starting to have on our understanding of early development, disease modeling, and potential therapeutic applications.


Subject(s)
Energy Metabolism/physiology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Animals , Humans , Mice
6.
Semin Cell Dev Biol ; 52: 76-83, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26828436

ABSTRACT

Human pluripotent stem cells (hPSCs) have great potential in regenerative medicine because they can differentiate into any cell type in the body. Genome integrity is vital for human development and for high fidelity passage of genetic information across generations through the germ line. To ensure genome stability, hPSCs maintain a lower rate of mutation than somatic cells and undergo rapid apoptosis in response to DNA damage and additional cell stresses. Furthermore, cellular metabolism and the cell cycle are also differentially regulated between cells in pluripotent and differentiated states and can aid in protecting hPSCs against DNA damage and damaged cell propagation. Despite these safeguards, clinical use of hPSC derivatives could be compromised by tumorigenic potential and possible malignant transformation from failed to differentiate cells. Since hPSCs and mature cells differentially respond to cell stress, it may be possible to specifically target undifferentiated cells for rapid apoptosis in mixed cell populations to enable safer use of hPSC-differentiated cells in patients.


Subject(s)
Apoptosis/physiology , Induced Pluripotent Stem Cells/physiology , Mitochondria/physiology , Animals , Cellular Reprogramming/physiology , Humans , Induced Pluripotent Stem Cells/cytology
7.
EMBO J ; 33(13): 1454-73, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24825347

ABSTRACT

Much of the mechanism by which Wnt signaling drives proliferation during oncogenesis is attributed to its regulation of the cell cycle. Here, we show how Wnt/ß-catenin signaling directs another hallmark of tumorigenesis, namely Warburg metabolism. Using biochemical assays and fluorescence lifetime imaging microscopy (FLIM) to probe metabolism in vitro and in living tumors, we observe that interference with Wnt signaling in colon cancer cells reduces glycolytic metabolism and results in small, poorly perfused tumors. We identify pyruvate dehydrogenase kinase 1 (PDK1) as an important direct target within a larger gene program for metabolism. PDK1 inhibits pyruvate flux to mitochondrial respiration and a rescue of its expression in Wnt-inhibited cancer cells rescues glycolysis as well as vessel growth in the tumor microenvironment. Thus, we identify an important mechanism by which Wnt-driven Warburg metabolism directs the use of glucose for cancer cell proliferation and links it to vessel delivery of oxygen and nutrients.


Subject(s)
Colonic Neoplasms/metabolism , Glucose/metabolism , Glycolysis , Neovascularization, Pathologic/metabolism , Tumor Microenvironment , Wnt Signaling Pathway , Animals , Cell Line, Tumor , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Glucose/genetics , Humans , Mice , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Oxygen Consumption/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase
8.
Proc Natl Acad Sci U S A ; 108(29): 12095-100, 2011 Jul 19.
Article in English | MEDLINE | ID: mdl-21730143

ABSTRACT

Burkholderia pseudomallei and Burkholderia thailandensis are related pathogens that invade a variety of cell types, replicate in the cytoplasm, and spread to nearby cells. We have investigated temporal and spatial requirements for virulence determinants in the intracellular life cycle, using genetic dissection and photothermal nanoblade delivery, which allows efficient placement of bacterium-sized cargo into the cytoplasm of mammalian cells. The conserved Bsa type III secretion system (T3SS(Bsa)) is dispensable for invasion, but is essential for escape from primary endosomes. By nanoblade delivery of B. thailandensis we demonstrate that all subsequent events in intercellular spread occur independently of T3SS(Bsa) activity. Although intracellular movement was essential for cell-cell spread by B. pseudomallei and B. thailandensis, neither BimA-mediated actin polymerization nor the formation of membrane protrusions containing bacteria was required for B. thailandensis. Surprisingly, the cryptic (fla2) flagellar system encoded on chromosome 2 of B. thailandensis supported rapid intracellular motility and efficient cell-cell spread. Plaque formation by both pathogens was dependent on the activity of a type VI secretion system (T6SS-1) that functions downstream from T3SS(Bsa)-mediated endosome escape. A remarkable feature of Burkholderia is their ability to induce the formation of multinucleate giant cells (MNGCs) in multiple cell types. By infection and nanoblade delivery, we observed complete correspondence between mutant phenotypes in assays for cell fusion and plaque formation, and time-course studies showed that plaque formation represents MNGC death. Our data suggest that the primary means for intercellular spread involves cell fusion, as opposed to pseudopod engulfment and bacterial escape from double-membrane vacuoles.


Subject(s)
Bacterial Secretion Systems/physiology , Burkholderia pseudomallei/physiology , Burkholderia pseudomallei/pathogenicity , Cytosol/microbiology , Melioidosis/transmission , Cell Fusion , Cell Line , Cytological Techniques/methods , Humans , Lasers , Microscopy, Fluorescence , Virulence Factors
9.
Cell Metab ; 36(1): 103-115.e4, 2024 01 02.
Article in English | MEDLINE | ID: mdl-38171330

ABSTRACT

The folate-dependent enzyme serine hydroxymethyltransferase (SHMT) reversibly converts serine into glycine and a tetrahydrofolate-bound one-carbon unit. Such one-carbon unit production plays a critical role in development, the immune system, and cancer. Using rodent models, here we show that the whole-body SHMT flux acts to net consume rather than produce glycine. Pharmacological inhibition of whole-body SHMT1/2 and genetic knockout of liver SHMT2 elevated circulating glycine levels up to eight-fold. Stable-isotope tracing revealed that the liver converts glycine to serine, which is then converted by serine dehydratase into pyruvate and burned in the tricarboxylic acid cycle. In response to diets deficient in serine and glycine, de novo biosynthetic flux was unaltered, but SHMT2- and serine-dehydratase-mediated catabolic flux was lower. Thus, glucose-derived serine synthesis is largely insensitive to systemic demand. Instead, circulating serine and glycine homeostasis is maintained through variable consumption, with liver SHMT2 a major glycine-consuming enzyme.


Subject(s)
Glycine Hydroxymethyltransferase , Glycine , Glycine Hydroxymethyltransferase/genetics , Homeostasis , Carbon , Serine
10.
bioRxiv ; 2024 Sep 28.
Article in English | MEDLINE | ID: mdl-39386450

ABSTRACT

Malonyl-CoA is the essential building block of fatty acids and regulates cell function through protein malonylation and allosteric regulation of signaling networks. Accordingly, the production and use of malonyl-CoA is finely tuned by the cellular energy status. Most studies of malonyl-CoA dynamics rely on bulk approaches that take only a snapshot of the average metabolic state of a population of cells, missing out on dynamic changes in malonyl-CoA and fatty acid biosynthesis that could be occurring within a single cell. To overcome this limitation, we have developed a genetically encoded fluorescent protein-based biosensor for malonyl-CoA that can be used to capture malonyl-CoA dynamics in single cells. This biosensor, termed Malibu (malonyl-CoA i ntracellular biosensor to understand dynamics), exhibits an excitation-ratiometric change in response to malonyl-CoA binding. We first used Malibu to monitor malonyl-CoA dynamics during inhibition of fatty acid biosynthesis using cerulenin in E. coli , observing an increase in Malibu response in a time- and dose-dependent manner. In HeLa cells, we used Malibu to monitor the impact of fatty acid biosynthesis inhibition on malonyl-CoA dynamics in single cells, finding that two inhibitors of fatty acid biosynthesis, cerulenin and orlistat, which inhibit different steps of fatty acid biosynthesis, increase malonyl-CoA levels. Altogether, we have developed a new genetically encoded biosensor for malonyl-CoA, which can be used to sensitively study malonyl-CoA dynamics in single cells, providing an unparalleled view into fatty acid biosynthesis.

11.
Nano Lett ; 12(11): 5669-72, 2012 Nov 14.
Article in English | MEDLINE | ID: mdl-23094784

ABSTRACT

Quantum dots (QDs) have not been used to label cytoskeleton structure of live cells owing to limitations in delivery strategies, and QDs conjugation methods and issues with nonspecific binding. We conjugated tubulin to QDs and applied the emerging method of photothermal nanoblade to deliver QD-tubulin conjugates into live Hela cells. This method will open new opportunities for cytosolic targeting of QDs in live cells.


Subject(s)
Nanotechnology/methods , Quantum Dots , Tubulin/chemistry , Cytoskeleton/metabolism , Cytosol/metabolism , Drug Delivery Systems , Endosomes/chemistry , HeLa Cells , Humans , Light , Microscopy/methods , Photochemistry/methods
12.
Nat Metab ; 5(8): 1275-1289, 2023 08.
Article in English | MEDLINE | ID: mdl-37612403

ABSTRACT

The pentose phosphate pathway (PPP) is a glucose-oxidizing pathway that runs in parallel to upper glycolysis to produce ribose 5-phosphate and nicotinamide adenine dinucleotide phosphate (NADPH). Ribose 5-phosphate is used for nucleotide synthesis, while NADPH is involved in redox homoeostasis as well as in promoting biosynthetic processes, such as the synthesis of tetrahydrofolate, deoxyribonucleotides, proline, fatty acids and cholesterol. Through NADPH, the PPP plays a critical role in suppressing oxidative stress, including in certain cancers, in which PPP inhibition may be therapeutically useful. Conversely, PPP-derived NADPH also supports purposeful cellular generation of reactive oxygen species (ROS) and reactive nitrogen species (RNS) for signalling and pathogen killing. Genetic deficiencies in the PPP occur relatively commonly in the committed pathway enzyme glucose-6-phosphate dehydrogenase (G6PD). G6PD deficiency typically manifests as haemolytic anaemia due to red cell oxidative damage but, in severe cases, also results in infections due to lack of leucocyte oxidative burst, highlighting the dual redox roles of the pathway in free radical production and detoxification. This Review discusses the PPP in mammals, covering its roles in biochemistry, physiology and disease.


Subject(s)
Oxidative Stress , Pentose Phosphate Pathway , Animals , NADP , Homeostasis , Fatty Acids , Mammals
13.
Med ; 3(2): 119-136, 2022 02 11.
Article in English | MEDLINE | ID: mdl-35425930

ABSTRACT

Background: Ketogenic diet is a potential means of augmenting cancer therapy. Here, we explore ketone body metabolism and its interplay with chemotherapy in pancreatic cancer. Methods: Metabolism and therapeutic responses of murine pancreatic cancer were studied using KPC primary tumors and tumor chunk allografts. Mice on standard high-carbohydrate diet or ketogenic diet were treated with cytotoxic chemotherapy (nab-paclitaxel, gemcitabine, cisplatin). Metabolic activity was monitored with metabolomics and isotope tracing, including 2H- and 13C-tracers, liquid chromatography-mass spectrometry, and imaging mass spectrometry. Findings: Ketone bodies are unidirectionally oxidized to make NADH. This stands in contrast to the carbohydrate-derived carboxylic acids lactate and pyruvate, which rapidly interconvert, buffering NADH/NAD. In murine pancreatic tumors, ketogenic diet decreases glucose's concentration and tricarboxylic acid cycle contribution, enhances 3-hydroxybutyrate's concentration and tricarboxylic acid contribution, and modestly elevates NADH, but does not impact tumor growth. In contrast, the combination of ketogenic diet and cytotoxic chemotherapy substantially raises tumor NADH and synergistically suppresses tumor growth, tripling the survival benefits of chemotherapy alone. Chemotherapy and ketogenic diet also synergize in immune-deficient mice, although long-term growth suppression was only observed in mice with an intact immune system. Conclusions: Ketogenic diet sensitizes murine pancreatic cancer tumors to cytotoxic chemotherapy. Based on these data, we have initiated a randomized clinical trial of chemotherapy with standard versus ketogenic diet for patients with metastatic pancreatic cancer (NCT04631445).


Subject(s)
Diet, Ketogenic , Pancreatic Neoplasms , Animals , Carbohydrates , Diet, Ketogenic/methods , Humans , Mice , NAD , Pancreatic Neoplasms/diet therapy , Pancreatic Neoplasms/drug therapy , Randomized Controlled Trials as Topic , Pancreatic Neoplasms
14.
Anal Chem ; 83(4): 1321-7, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21247066

ABSTRACT

It is difficult to achieve controlled cutting of elastic, mechanically fragile, and rapidly resealing mammalian cell membranes. Here, we report a photothermal nanoblade that utilizes a metallic nanostructure to harvest short laser pulse energy and convert it into a highly localized explosive vapor bubble, which rapidly punctures a lightly contacting cell membrane via high-speed fluidic flows and induced transient shear stress. The cavitation bubble pattern is controlled by the metallic structure configuration and laser pulse duration and energy. Integration of the metallic nanostructure with a micropipet, the nanoblade generates a micrometer-sized membrane access port for delivering highly concentrated cargo (5 × 10(8) live bacteria/mL) with high efficiency (46%) and cell viability (>90%) into mammalian cells. Additional biologic and inanimate cargo over 3-orders of magnitude in size including DNA, RNA, 200 nm polystyrene beads, to 2 µm bacteria have also been delivered into multiple mammalian cell types. Overall, the photothermal nanoblade is a new approach for delivering difficult cargo into mammalian cells.


Subject(s)
Light , Metal Nanoparticles/chemistry , Temperature , Biological Transport/radiation effects , Burkholderia/metabolism , Cell Line , Cell Membrane/metabolism , Cell Membrane/radiation effects , Cell Survival , Fibroblasts/cytology , Fibroblasts/metabolism , Glass/chemistry , Humans , Optical Phenomena , RNA/metabolism , Reproducibility of Results , Time Factors , Titanium/chemistry
15.
Nat Metab ; 3(7): 896-908, 2021 07.
Article in English | MEDLINE | ID: mdl-34211182

ABSTRACT

Altered metabolic activity contributes to the pathogenesis of a number of diseases, including diabetes, heart failure, cancer, fibrosis and neurodegeneration. These diseases, and organismal metabolism more generally, are only partially recapitulated by cell culture models. Accordingly, it is important to measure metabolism in vivo. Over the past century, researchers studying glucose homeostasis have developed strategies for the measurement of tissue-specific and whole-body metabolic activity (pathway fluxes). The power of these strategies has been augmented by recent advances in metabolomics technologies. Here, we review techniques for measuring metabolic fluxes in intact mammals and discuss how to analyse and interpret the results. In tandem, we describe important findings from these techniques, and suggest promising avenues for their future application. Given the broad importance of metabolism to health and disease, more widespread application of these methods holds the potential to accelerate biomedical progress.


Subject(s)
Metabolome , Metabolomics/methods , Animals , Disease Susceptibility , Energy Metabolism , Homeostasis , Humans , Mammals , Metabolic Networks and Pathways , Organ Specificity
16.
Nat Metab ; 3(12): 1608-1620, 2021 12.
Article in English | MEDLINE | ID: mdl-34845393

ABSTRACT

Carbohydrate can be converted into fat by de novo lipogenesis, a process upregulated in fatty liver disease. Chemically, de novo lipogenesis involves polymerization and reduction of acetyl-CoA, using NADPH as the electron donor. The feedstocks used to generate acetyl-CoA and NADPH in lipogenic tissues remain, however, unclear. Here we show using stable isotope tracing in mice that de novo lipogenesis in adipose is supported by glucose and its catabolism via the pentose phosphate pathway to make NADPH. The liver, in contrast, derives acetyl-CoA for lipogenesis from acetate and lactate, and NADPH from folate-mediated serine catabolism. Such NADPH generation involves the cytosolic serine pathway in liver running in the opposite direction to that observed in most tissues and tumours, with NADPH made by the SHMT1-MTHFD1-ALDH1L1 reaction sequence. SHMT inhibition decreases hepatic lipogenesis. Thus, liver folate metabolism is distinctively wired to support cytosolic NADPH production and lipogenesis. More generally, while the same enzymes are involved in fat synthesis in liver and adipose, different substrates are used, opening the door to tissue-specific pharmacological interventions.


Subject(s)
Lipogenesis , Liver/metabolism , NADP/metabolism , Serine/metabolism , Acetyl Coenzyme A/metabolism , Adipose Tissue/metabolism , Aminohydrolases/metabolism , Animals , Fatty Acids/metabolism , Female , Folic Acid/metabolism , Formate-Tetrahydrofolate Ligase/metabolism , Glutamine/metabolism , Glycine Hydroxymethyltransferase/metabolism , Hepatocytes/metabolism , Lipid Metabolism , Male , Metabolic Networks and Pathways , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Mice , Multienzyme Complexes/metabolism , Oxidative Phosphorylation , Oxidoreductases Acting on CH-NH Group Donors/metabolism
17.
Cell Metab ; 33(2): 367-378.e5, 2021 02 02.
Article in English | MEDLINE | ID: mdl-33472024

ABSTRACT

Glycolysis plays a central role in organismal metabolism, but its quantitative inputs across mammalian tissues remain unclear. Here we use 13C-tracing in mice to quantify glycolytic intermediate sources: circulating glucose, intra-tissue glycogen, and circulating gluconeogenic precursors. Circulating glucose is the main source of circulating lactate, the primary end product of tissue glycolysis. Yet circulating glucose highly labels glycolytic intermediates in only a few tissues: blood, spleen, diaphragm, and soleus muscle. Most glycolytic intermediates in the bulk of body tissue, including liver and quadriceps muscle, come instead from glycogen. Gluconeogenesis contributes less but also broadly to glycolytic intermediates, and its flux persists with physiologic feeding (but not hyperinsulinemic clamp). Instead of suppressing gluconeogenesis, feeding activates oxidation of circulating glucose and lactate to maintain glucose homeostasis. Thus, the bulk of the body slowly breaks down internally stored glycogen while select tissues rapidly catabolize circulating glucose to lactate for oxidation throughout the body.


Subject(s)
Diaphragm/metabolism , Muscle, Skeletal/metabolism , Spleen/metabolism , Animals , Blood Glucose/metabolism , Carbon Isotopes , Gluconeogenesis , Glycogen/blood , Glycogen/metabolism , Glycolysis , Male , Mice , Mice, Inbred C57BL
18.
Opt Express ; 18(22): 23153-60, 2010 Oct 25.
Article in English | MEDLINE | ID: mdl-21164656

ABSTRACT

We report a photothermal nanoblade that utilizes a metallic nanostructure to harvest short laser pulse energy and convert it into a highly localized and specifically shaped explosive vapor bubble. Rapid bubble expansion and collapse punctures a lightly-contacting cell membrane via high-speed fluidic flows and induced transient shear stress. The membrane cutting pattern is controlled by the metallic nanostructure configuration, laser pulse polarization, and energy. Highly controllable, sub-micron sized circular hole pairs to half moon-like, or cat-door shaped, membrane cuts were realized in glutaraldehyde treated HeLa cells.


Subject(s)
Cell Membrane/chemistry , Light , Nanostructures/chemistry , Nanotechnology/methods , Pattern Recognition, Automated/methods , Temperature , Absorption , Computer Simulation , HeLa Cells , Humans , Lasers , Reproducibility of Results
19.
Cell Metab ; 32(4): 676-688.e4, 2020 10 06.
Article in English | MEDLINE | ID: mdl-32791100

ABSTRACT

Mammalian organs are nourished by nutrients carried by the blood circulation. These nutrients originate from diet and internal stores, and can undergo various interconversions before their eventual use as tissue fuel. Here we develop isotope tracing, mass spectrometry, and mathematical analysis methods to determine the direct sources of circulating nutrients, their interconversion rates, and eventual tissue-specific contributions to TCA cycle metabolism. Experiments with fifteen nutrient tracers enabled extensive accounting for both circulatory metabolic cycles and tissue TCA inputs, across fed and fasted mice on either high-carbohydrate or ketogenic diet. We find that a majority of circulating carbon flux is carried by two major cycles: glucose-lactate and triglyceride-glycerol-fatty acid. Futile cycling through these pathways is prominent when dietary content of the associated nutrients is low, rendering internal metabolic activity robust to food choice. The presented in vivo flux quantification methods are broadly applicable to different physiological and disease states.


Subject(s)
Fatty Acids/metabolism , Glucose/metabolism , Glycerol/metabolism , Lactic Acid/metabolism , Triglycerides/metabolism , Animals , Citric Acid Cycle , Mice , Mice, Inbred C57BL
20.
Cell Rep ; 33(11): 108500, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33326785

ABSTRACT

Immune cell function is influenced by metabolic conditions. Low-glucose, high-lactate environments, such as the placenta, gastrointestinal tract, and the tumor microenvironment, are immunosuppressive, especially for glycolysis-dependent effector T cells. We report that nicotinamide adenine dinucleotide (NAD+), which is reduced to NADH by lactate dehydrogenase in lactate-rich conditions, is a key point of metabolic control in T cells. Reduced NADH is not available for NAD+-dependent enzymatic reactions involving glyceraldehyde 3-phosphate dehydrogenase (GAPDH) and 3-phosphoglycerate dehydrogenase (PGDH). We show that increased lactate leads to a block at GAPDH and PGDH, leading to the depletion of post-GAPDH glycolytic intermediates, as well as the 3-phosphoglycerate derivative serine that is known to be important for T cell proliferation. Supplementing serine rescues the ability of T cells to proliferate in the presence of lactate-induced reductive stress. Directly targeting the redox state may be a useful approach for developing novel immunotherapies in cancer and therapeutic immunosuppression.


Subject(s)
Lactic Acid/metabolism , NAD/metabolism , Cell Proliferation , Humans , Oxidation-Reduction
SELECTION OF CITATIONS
SEARCH DETAIL