1
|
Lin P, Lane AN, Fan TWM. NMR-Based Stable Isotope Tracing of Cancer Metabolism. Methods Mol Biol 2025; 2855:457-504. [PMID: 39354323 DOI: 10.1007/978-1-0716-4116-3_26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2024]
Abstract
NMR is widely used for metabolite profiling (metabolomics, metabonomics) particularly of various readily obtainable biofluids such as plasma and urine. It is especially valuable for stable isotope tracer studies to track metabolic pathways under control or perturbed conditions in a wide range of cell models as well as animal models and human subjects. NMR has unique properties for utilizing stable isotopes to edit or simplify otherwise complex spectra acquired in vitro and in vivo, while quantifying the level of enrichment at specific atomic positions in various metabolites (i.e., isotopomer distribution analysis).In this protocol, we give an overview with specific protocols for NMR-based stable isotope-resolved metabolomics, or SIRM, with a workflow from administration of isotope-enriched precursors, via sample preparation through to NMR data collection and reduction. We focus on indirect detection of common NMR-active stable isotopes including 13C, 15N, 31P, and 2H, using a variety of 1H-based two-dimensional experiments. We also include the application and analyses of multiplex tracer experiments.
Collapse
Affiliation(s)
- Penghui Lin
- Center for Environmental and Systems Biochemistry, Department of Toxicology and Cancer Biology, Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Andrew N Lane
- Center for Environmental and Systems Biochemistry, Department of Toxicology and Cancer Biology, Markey Cancer Center, University of Kentucky, Lexington, KY, USA.
| | - Teresa W-M Fan
- Center for Environmental and Systems Biochemistry, Department of Toxicology and Cancer Biology, Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
2
|
Deja S, Fletcher JA, Kim CW, Kucejova B, Fu X, Mizerska M, Villegas M, Pudelko-Malik N, Browder N, Inigo-Vollmer M, Menezes CJ, Mishra P, Berglund ED, Browning JD, Thyfault JP, Young JD, Horton JD, Burgess SC. Hepatic malonyl-CoA synthesis restrains gluconeogenesis by suppressing fat oxidation, pyruvate carboxylation, and amino acid availability. Cell Metab 2024; 36:1088-1104.e12. [PMID: 38447582 PMCID: PMC11081827 DOI: 10.1016/j.cmet.2024.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 12/10/2023] [Accepted: 02/09/2024] [Indexed: 03/08/2024]
Abstract
Acetyl-CoA carboxylase (ACC) promotes prandial liver metabolism by producing malonyl-CoA, a substrate for de novo lipogenesis and an inhibitor of CPT-1-mediated fat oxidation. We report that inhibition of ACC also produces unexpected secondary effects on metabolism. Liver-specific double ACC1/2 knockout (LDKO) or pharmacologic inhibition of ACC increased anaplerosis, tricarboxylic acid (TCA) cycle intermediates, and gluconeogenesis by activating hepatic CPT-1 and pyruvate carboxylase flux in the fed state. Fasting should have marginalized the role of ACC, but LDKO mice maintained elevated TCA cycle intermediates and preserved glycemia during fasting. These effects were accompanied by a compensatory induction of proteolysis and increased amino acid supply for gluconeogenesis, which was offset by increased protein synthesis during feeding. Such adaptations may be related to Nrf2 activity, which was induced by ACC inhibition and correlated with fasting amino acids. The findings reveal unexpected roles for malonyl-CoA synthesis in liver and provide insight into the broader effects of pharmacologic ACC inhibition.
Collapse
Affiliation(s)
- Stanislaw Deja
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA; Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Justin A Fletcher
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA; Department of Clinical Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Chai-Wan Kim
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Blanka Kucejova
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Xiaorong Fu
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA; Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Monika Mizerska
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Morgan Villegas
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Natalia Pudelko-Malik
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA; Department of Biochemistry, Molecular Biology and Biotechnology, Faculty of Chemistry, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Nicholas Browder
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Melissa Inigo-Vollmer
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Cameron J Menezes
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Prashant Mishra
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Eric D Berglund
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Jeffrey D Browning
- Department of Clinical Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - John P Thyfault
- Departments of Cell Biology and Physiology, Internal Medicine and KU Diabetes Institute, Kansas Medical Center, Kansas City, KS, USA
| | - Jamey D Young
- Department of Chemical and Biomolecular Engineering, Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37235, USA
| | - Jay D Horton
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA; Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA.
| | - Shawn C Burgess
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA; Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA.
| |
Collapse
|
3
|
Malloy CR, Sherry AD, Alger JR, Jin ES. Recent progress in analysis of intermediary metabolism by ex vivo 13 C NMR. NMR IN BIOMEDICINE 2023; 36:e4817. [PMID: 35997012 DOI: 10.1002/nbm.4817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 08/03/2022] [Accepted: 08/16/2022] [Indexed: 06/15/2023]
Abstract
Advanced imaging technologies, large-scale metabolomics, and the measurement of gene transcripts or enzyme expression all enable investigations of intermediary metabolism in human patients. Complementary information about fluxes in individual metabolic pathways may be obtained by ex vivo 13 C NMR of blood or tissue biopsies. Simple molecules such as 13 C-labeled glucose are readily administered to patients prior to surgical biopsies, and 13 C-labeled glycerol is easily administered orally to outpatients. Here, we review recent progress in practical applications of 13 C NMR to study cancer biology, the response to oxidative stress, gluconeogenesis, triglyceride synthesis in patients, as well as new insights into compartmentation of metabolism in the cytosol. The technical aspects of obtaining the sample, preparing material for analysis, and acquiring the spectra are relatively simple. This approach enables convenient, valuable, and quantitative insights into intermediary metabolism in patients.
Collapse
Affiliation(s)
- Craig R Malloy
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Veterans Affairs North Texas Healthcare System, Dallas, Texas, USA
| | - A Dean Sherry
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Chemistry, University of Texas at Dallas, Richardson, Texas, USA
| | - Jeffry R Alger
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Neurology, Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Eunsook S Jin
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
4
|
Hubbard BT, LaMoia TE, Goedeke L, Gaspar RC, Galsgaard KD, Kahn M, Mason GF, Shulman GI. Q-Flux: A method to assess hepatic mitochondrial succinate dehydrogenase, methylmalonyl-CoA mutase, and glutaminase fluxes in vivo. Cell Metab 2023; 35:212-226.e4. [PMID: 36516861 PMCID: PMC9887731 DOI: 10.1016/j.cmet.2022.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 09/14/2022] [Accepted: 11/21/2022] [Indexed: 12/15/2022]
Abstract
The mammalian succinate dehydrogenase (SDH) complex has recently been shown as capable of operating bidirectionally. Here, we develop a method (Q-Flux) capable of measuring absolute rates of both forward (VSDH(F)) and reverse (VSDH(R)) flux through SDH in vivo while also deconvoluting the amount of glucose derived from four discreet carbon sources in the liver. In validation studies, a mitochondrial uncoupler increased net SDH flux by >100% in awake rodents but also increased SDH cycling. During hyperglucagonemia, attenuated pyruvate cycling enhances phosphoenolpyruvate carboxykinase efficiency to drive increased gluconeogenesis, which is complemented by increased glutaminase (GLS) flux, methylmalonyl-CoA mutase (MUT) flux, and glycerol conversion to glucose. During hyperinsulinemic-euglycemic clamp, both pyruvate carboxylase and GLS are suppressed, while VSDH(R) is increased. Unstimulated MUT is a minor anaplerotic reaction but is readily induced by small amounts of propionate, which elicits glucagon-like metabolic rewiring. Taken together, Q-Flux yields a comprehensive picture of hepatic mitochondrial metabolism and should be broadly useful to researchers.
Collapse
Affiliation(s)
- Brandon T Hubbard
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA; Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Traci E LaMoia
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA; Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Leigh Goedeke
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Rafael C Gaspar
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Katrine D Galsgaard
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Mario Kahn
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Graeme F Mason
- Department of Radiology & Biomedical Imaging, Yale School of Medicine, New Haven, CT 06510, USA; Departments of Psychiatry & Biomedical Engineering, Yale School of Medicine, New Haven, CT 06510, USA
| | - Gerald I Shulman
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA; Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
5
|
Repetitive Transcranial Magnetic Stimulation-Associated Changes in Neocortical Metabolites in Major Depression: A Systematic Review. Neuroimage Clin 2022; 35:103049. [PMID: 35738081 PMCID: PMC9233277 DOI: 10.1016/j.nicl.2022.103049] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 04/01/2022] [Accepted: 05/12/2022] [Indexed: 11/23/2022]
Abstract
We reviewed 12 studies that measured metabolites pre and post rTMS in MDD. Frontal lobe Glu, Gln, NAA, and GABA increased after rTMS. Increases in metabolites were often associated with MDD symptom improvement. We propose novel intracellular mechanisms by which metabolites are altered by rTMS.
Introduction Repetitive Transcranial magnetic stimulation (rTMS) is an FDA approved treatment for major depressive disorder (MDD). However, neural mechanisms contributing to rTMS effects on depressive symptoms, cognition, and behavior are unclear. Proton magnetic resonance spectroscopy (MRS), a noninvasive neuroimaging technique measuring concentrations of biochemical compounds within the brain in vivo, may provide mechanistic insights. Methods This systematic review summarized published MRS findings from rTMS treatment trials to address potential neurometabolic mechanisms of its antidepressant action. Using PubMed, Google Scholar, Web of Science, and JSTOR, we identified twelve empirical studies that evaluated changes in MRS metabolites in a within-subjects, pre- vs. post-rTMS treatment design in patients with MDD. Results rTMS protocols ranged from four days to eight weeks duration, were applied at high frequency to the left dorsolateral prefrontal cortex (DLPFC) in most studies, and were conducted in patients aged 13-to-70. Most studies utilized MRS point resolved spectroscopy acquisitions at 3 Tesla in the bilateral anterior cingulate cortex and DLPFC. Symptom improvements were correlated with rTMS-related increases in the concentration of glutamatergic compounds (glutamate, Glu, and glutamine, Gln), GABA, and N-acetylated compounds (NAA), with some results trend-level. Conclusions This is the first in-depth systematic review of metabolic effects of rTMS in individuals with MDD. The extant literature suggests rTMS stimulation does not produce changes in neurometabolites independent of clinical response; increases in frontal lobe glutamatergic compounds, N-acetylated compounds and GABA following high frequency left DLPFC rTMS therapy were generally associated with clinical improvement. Glu, Gln, GABA, and NAA may mediate rTMS treatment effects on MDD symptomatology through intracellular mechanisms.
Collapse
|
6
|
Can E, Bastiaansen JAM, Couturier DL, Gruetter R, Yoshihara HAI, Comment A. [ 13C]bicarbonate labelled from hyperpolarized [1- 13C]pyruvate is an in vivo marker of hepatic gluconeogenesis in fasted state. Commun Biol 2022; 5:10. [PMID: 35013537 PMCID: PMC8748681 DOI: 10.1038/s42003-021-02978-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 12/07/2021] [Indexed: 01/07/2023] Open
Abstract
Hyperpolarized [1-13C]pyruvate enables direct in vivo assessment of real-time liver enzymatic activities by 13C magnetic resonance. However, the technique usually requires the injection of a highly supraphysiological dose of pyruvate. We herein demonstrate that liver metabolism can be measured in vivo with hyperpolarized [1-13C]pyruvate administered at two- to three-fold the basal plasma concentration. The flux through pyruvate dehydrogenase, assessed by 13C-labeling of bicarbonate in the fed condition, was found to be saturated or partially inhibited by supraphysiological doses of hyperpolarized [1-13C]pyruvate. The [13C]bicarbonate signal detected in the liver of fasted rats nearly vanished after treatment with a phosphoenolpyruvate carboxykinase (PEPCK) inhibitor, indicating that the signal originates from the flux through PEPCK. In addition, the normalized [13C]bicarbonate signal in fasted untreated animals is dose independent across a 10-fold range, highlighting that PEPCK and pyruvate carboxylase are not saturated and that hepatic gluconeogenesis can be directly probed in vivo with hyperpolarized [1-13C]pyruvate. Can et al. demonstrate the ability to use hyperpolarized [1-13C]pyruvate at nearphysiological concentrations to directly assess liver enzymatic activities by 13C magnetic resonance. While in the fed state, the normalized [13C]bicarbonate signal produced from hyperpolarized [1-13C]pyruvate derives from PDH activity, which is saturated at supraphysiological doses, it results from PEPCK in the fasted state and is dose-independent, allowing non-invasive in vivo detection of hepatic gluconeogenesis.”
Collapse
Affiliation(s)
- Emine Can
- Institute of Physics, Ecole Polytechnique Fédérale de Lausanne, CH-1015, Lausanne, Switzerland
| | - Jessica A M Bastiaansen
- Institute of Physics, Ecole Polytechnique Fédérale de Lausanne, CH-1015, Lausanne, Switzerland.,Department of Diagnostic and Interventional Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | | | - Rolf Gruetter
- Institute of Physics, Ecole Polytechnique Fédérale de Lausanne, CH-1015, Lausanne, Switzerland
| | - Hikari A I Yoshihara
- Institute of Physics, Ecole Polytechnique Fédérale de Lausanne, CH-1015, Lausanne, Switzerland
| | - Arnaud Comment
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, Cambridgeshire, CB2 0RE, UK. .,General Electric Healthcare, Chalfont St Giles, Buckinghamshire, HP8 4SP, UK.
| |
Collapse
|
7
|
Shannon CE, Ragavan M, Palavicini JP, Fourcaudot M, Bakewell TM, Valdez IA, Ayala I, Jin ES, Madesh M, Han X, Merritt ME, Norton L. Insulin resistance is mechanistically linked to hepatic mitochondrial remodeling in non-alcoholic fatty liver disease. Mol Metab 2021; 45:101154. [PMID: 33359401 PMCID: PMC7811046 DOI: 10.1016/j.molmet.2020.101154] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 12/18/2020] [Accepted: 12/20/2020] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVE Insulin resistance and altered hepatic mitochondrial function are central features of type 2 diabetes (T2D) and non-alcoholic fatty liver disease (NAFLD), but the etiological role of these processes in disease progression remains unclear. Here we investigated the molecular links between insulin resistance, mitochondrial remodeling, and hepatic lipid accumulation. METHODS Hepatic insulin sensitivity, endogenous glucose production, and mitochondrial metabolic fluxes were determined in wild-type, obese (ob/ob) and pioglitazone-treatment obese mice using a combination of radiolabeled tracer and stable isotope NMR approaches. Mechanistic studies of pioglitazone action were performed in isolated primary hepatocytes, whilst molecular hepatic lipid species were profiled using shotgun lipidomics. RESULTS Livers from obese, insulin-resistant mice displayed augmented mitochondrial content and increased tricarboxylic acid cycle (TCA) cycle and pyruvate dehydrogenase (PDH) activities. Insulin sensitization with pioglitazone mitigated pyruvate-driven TCA cycle activity and PDH activation via both allosteric (intracellular pyruvate availability) and covalent (PDK4 and PDP2) mechanisms that were dependent on PPARγ activity in isolated primary hepatocytes. Improved mitochondrial function following pioglitazone treatment was entirely dissociated from changes in hepatic triglycerides, diacylglycerides, or fatty acids. Instead, we highlight a role for the mitochondrial phospholipid cardiolipin, which underwent pathological remodeling in livers from obese mice that was reversed by insulin sensitization. CONCLUSION Our findings identify targetable mitochondrial features of T2D and NAFLD and highlight the benefit of insulin sensitization in managing the clinical burden of obesity-associated disease.
Collapse
Affiliation(s)
- Chris E Shannon
- Division of Diabetes, University of Texas Health Science Center and Texas Diabetes Institute, San Antonio, TX, USA
| | - Mukundan Ragavan
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Juan Pablo Palavicini
- Division of Diabetes, University of Texas Health Science Center and Texas Diabetes Institute, San Antonio, TX, USA; Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Marcel Fourcaudot
- Division of Diabetes, University of Texas Health Science Center and Texas Diabetes Institute, San Antonio, TX, USA
| | - Terry M Bakewell
- Division of Diabetes, University of Texas Health Science Center and Texas Diabetes Institute, San Antonio, TX, USA
| | - Ivan A Valdez
- Division of Diabetes, University of Texas Health Science Center and Texas Diabetes Institute, San Antonio, TX, USA
| | - Iriscilla Ayala
- Division of Diabetes, University of Texas Health Science Center and Texas Diabetes Institute, San Antonio, TX, USA
| | - Eunsook S Jin
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Muniswamy Madesh
- Division of Nephrology, University of Texas Health Science Center and Texas Diabetes Institute, San Antonio, TX, USA
| | - Xianlin Han
- Division of Diabetes, University of Texas Health Science Center and Texas Diabetes Institute, San Antonio, TX, USA; Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA; Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Matthew E Merritt
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Luke Norton
- Division of Diabetes, University of Texas Health Science Center and Texas Diabetes Institute, San Antonio, TX, USA.
| |
Collapse
|
8
|
Neeland IJ, Rocha NDA, Hughes C, Ayers CR, Malloy CR, Jin ES. Effects of Empagliflozin Treatment on Glycerol-Derived Hepatic Gluconeogenesis in Adults with Obesity: A Randomized Clinical Trial. Obesity (Silver Spring) 2020; 28:1254-1262. [PMID: 32568464 PMCID: PMC7316140 DOI: 10.1002/oby.22854] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 04/03/2020] [Accepted: 04/15/2020] [Indexed: 01/18/2023]
Abstract
OBJECTIVE The aim of this study was to determine the effects of empagliflozin on glycerol-derived hepatic gluconeogenesis in adults with obesity without type 2 diabetes mellitus (T2DM) using oral carbon 13 (13 C)-labeled glycerol. METHODS A randomized, double-blind, placebo-controlled trial was performed in participants with magnetic resonance imaging assessment of body fat and measurement of glycerol-derived 13 C enrichment in plasma glucose by nuclear magnetic resonance spectroscopy following ingestion of [U-13 C3 ]glycerol. Participants were randomized to oral empagliflozin 10 mg once daily or placebo for 3 months. Glycerol-derived 13 C enrichment studies were repeated, and treatment differences in the mean percentage of 13 C glycerol enrichment in glucose were compared using mixed linear models. RESULTS Thirty-five participants completed the study. Empagliflozin increased glycerol-derived 13 C enrichment between baseline and follow-up by 6.5% (P = 0.005), consistent with less glycerol from visceral adipose tissue (VAT). No difference was found with placebo. Glycerol-derived 13 C enrichment was lower in participants with high VAT compared with low VAT by 12.6% (P = 0.04), but there was no heterogeneity of the treatment effect by baseline VAT. Glycerol-derived 13 C enrichment was inversely correlated with VAT but was not correlated with weight loss. CONCLUSIONS VAT is associated with endogenous glycerol-derived hepatic gluconeogenesis, and empagliflozin reduces endogenous glycerol gluconeogenesis in adults with obesity without T2DM. These findings suggest a mechanism by which sodium-glucose cotransporter 2 inhibitors may prevent T2DM in obesity.
Collapse
Affiliation(s)
- Ian J. Neeland
- Department of Internal Medicine, Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Natalia de Albuquerque Rocha
- Department of Internal Medicine, Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Connor Hughes
- Department of Internal Medicine, Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Colby R. Ayers
- Department of Internal Medicine, Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Craig R. Malloy
- Advanced Imaging Research Center and Departments of Internal Medicine and Radiology, University of Texas Southwestern Medical Center and VA North Texas Healthcare System, Dallas, TX, USA
| | - Eunsook S. Jin
- Advanced Imaging Research Center and Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
9
|
Deja S, Fu X, Fletcher JA, Kucejova B, Browning JD, Young JD, Burgess SC. Simultaneous tracers and a unified model of positional and mass isotopomers for quantification of metabolic flux in liver. Metab Eng 2019; 59:1-14. [PMID: 31891762 DOI: 10.1016/j.ymben.2019.12.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 12/12/2019] [Accepted: 12/21/2019] [Indexed: 12/23/2022]
Abstract
Computational models based on the metabolism of stable isotope tracers can yield valuable insight into the metabolic basis of disease. The complexity of these models is limited by the number of tracers and the ability to characterize tracer labeling in downstream metabolites. NMR spectroscopy is ideal for multiple tracer experiments since it precisely detects the position of tracer nuclei in molecules, but it lacks sensitivity for detecting low-concentration metabolites. GC-MS detects stable isotope mass enrichment in low-concentration metabolites, but lacks nuclei and positional specificity. We performed liver perfusions and in vivo infusions of 2H and 13C tracers, yielding complex glucose isotopomers that were assigned by NMR and fit to a newly developed metabolic model. Fluxes regressed from 2H and 13C NMR positional isotopomer enrichments served to validate GC-MS-based flux estimates obtained from the same experimental samples. NMR-derived fluxes were largely recapitulated by modeling the mass isotopomer distributions of six glucose fragment ions measured by GC-MS. Modest differences related to limited fragmentation coverage of glucose C1-C3 were identified, but fluxes such as gluconeogenesis, glycogenolysis, cataplerosis and TCA cycle flux were tightly correlated between the methods. Most importantly, modeling of GC-MS data could assign fluxes in primary mouse hepatocytes, an experiment that is impractical by 2H or 13C NMR.
Collapse
Affiliation(s)
- Stanislaw Deja
- Center for Human Nutrition, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Xiaorong Fu
- Center for Human Nutrition, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Justin A Fletcher
- Center for Human Nutrition, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Blanka Kucejova
- Center for Human Nutrition, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Jeffrey D Browning
- Department of Clinical Nutrition, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Jamey D Young
- Department of Chemical and Biomolecular Engineering, Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37235, USA.
| | - Shawn C Burgess
- Center for Human Nutrition, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
10
|
Parkhitko AA, Jouandin P, Mohr SE, Perrimon N. Methionine metabolism and methyltransferases in the regulation of aging and lifespan extension across species. Aging Cell 2019; 18:e13034. [PMID: 31460700 PMCID: PMC6826121 DOI: 10.1111/acel.13034] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 07/11/2019] [Accepted: 08/08/2019] [Indexed: 12/20/2022] Open
Abstract
Methionine restriction (MetR) extends lifespan across different species and exerts beneficial effects on metabolic health and inflammatory responses. In contrast, certain cancer cells exhibit methionine auxotrophy that can be exploited for therapeutic treatment, as decreasing dietary methionine selectively suppresses tumor growth. Thus, MetR represents an intervention that can extend lifespan with a complementary effect of delaying tumor growth. Beyond its function in protein synthesis, methionine feeds into complex metabolic pathways including the methionine cycle, the transsulfuration pathway, and polyamine biosynthesis. Manipulation of each of these branches extends lifespan; however, the interplay between MetR and these branches during regulation of lifespan is not well understood. In addition, a potential mechanism linking the activity of methionine metabolism and lifespan is regulation of production of the methyl donor S-adenosylmethionine, which, after transferring its methyl group, is converted to S-adenosylhomocysteine. Methylation regulates a wide range of processes, including those thought to be responsible for lifespan extension by MetR. Although the exact mechanisms of lifespan extension by MetR or methionine metabolism reprogramming are unknown, it may act via reducing the rate of translation, modifying gene expression, inducing a hormetic response, modulating autophagy, or inducing mitochondrial function, antioxidant defense, or other metabolic processes. Here, we review the mechanisms of lifespan extension by MetR and different branches of methionine metabolism in different species and the potential for exploiting the regulation of methyltransferases to delay aging.
Collapse
Affiliation(s)
- Andrey A. Parkhitko
- Department of GeneticsBlavatnik InstituteHarvard Medical SchoolBostonMassachusetts
| | - Patrick Jouandin
- Department of GeneticsBlavatnik InstituteHarvard Medical SchoolBostonMassachusetts
| | - Stephanie E. Mohr
- Department of GeneticsBlavatnik InstituteHarvard Medical SchoolBostonMassachusetts
| | - Norbert Perrimon
- Department of GeneticsBlavatnik InstituteHarvard Medical SchoolBostonMassachusetts
- Howard Hughes Medical InstituteBostonMassachusetts
| |
Collapse
|
11
|
Real-time hyperpolarized 13C magnetic resonance detects increased pyruvate oxidation in pyruvate dehydrogenase kinase 2/4-double knockout mouse livers. Sci Rep 2019; 9:16480. [PMID: 31712597 PMCID: PMC6848094 DOI: 10.1038/s41598-019-52952-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 10/22/2019] [Indexed: 01/05/2023] Open
Abstract
The pyruvate dehydrogenase complex (PDH) critically regulates carbohydrate metabolism. Phosphorylation of PDH by one of the pyruvate dehydrogenase kinases 1-4 (PDK1-4) decreases the flux of carbohydrates into the TCA cycle. Inhibition of PDKs increases oxidative metabolism of carbohydrates, so targeting PDKs has emerged as an important therapeutic approach to manage various metabolic diseases. Therefore, it is highly desirable to begin to establish imaging tools for noninvasive measurements of PDH flux in rodent models. In this study, we used hyperpolarized (HP) 13C-magnetic resonance spectroscopy to study the impact of a PDK2/PDK4 double knockout (DKO) on pyruvate metabolism in perfused livers from lean and diet-induced obese (DIO) mice and validated the HP observations with high-resolution 13C-nuclear magnetic resonance (NMR) spectroscopy of tissue extracts and steady-state isotopomer analyses. We observed that PDK-deficient livers produce more HP-bicarbonate from HP-[1-13C]pyruvate than age-matched control livers. A steady-state 13C-NMR isotopomer analysis of tissue extracts confirmed that flux rates through PDH, as well as pyruvate carboxylase and pyruvate cycling activities, are significantly higher in PDK-deficient livers. Immunoblotting experiments confirmed that HP-bicarbonate production from HP-[1-13C]pyruvate parallels decreased phosphorylation of the PDH E1α subunit (pE1α) in liver tissue. Our findings indicate that combining real-time hyperpolarized 13C NMR spectroscopy and 13C isotopomer analysis provides quantitative insights into intermediary metabolism in PDK-knockout mice. We propose that this method will be useful in assessing metabolic disease states and developing therapies to improve PDH flux.
Collapse
|
12
|
Cappel DA, Deja S, Duarte JAG, Kucejova B, Iñigo M, Fletcher JA, Fu X, Berglund ED, Liu T, Elmquist JK, Hammer S, Mishra P, Browning JD, Burgess SC. Pyruvate-Carboxylase-Mediated Anaplerosis Promotes Antioxidant Capacity by Sustaining TCA Cycle and Redox Metabolism in Liver. Cell Metab 2019; 29:1291-1305.e8. [PMID: 31006591 PMCID: PMC6585968 DOI: 10.1016/j.cmet.2019.03.014] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 01/12/2019] [Accepted: 03/26/2019] [Indexed: 01/09/2023]
Abstract
The hepatic TCA cycle supports oxidative and biosynthetic metabolism. This dual responsibility requires anaplerotic pathways, such as pyruvate carboxylase (PC), to generate TCA cycle intermediates necessary for biosynthesis without disrupting oxidative metabolism. Liver-specific PC knockout (LPCKO) mice were created to test the role of anaplerotic flux in liver metabolism. LPCKO mice have impaired hepatic anaplerosis, diminution of TCA cycle intermediates, suppressed gluconeogenesis, reduced TCA cycle flux, and a compensatory increase in ketogenesis and renal gluconeogenesis. Loss of PC depleted aspartate and compromised urea cycle function, causing elevated urea cycle intermediates and hyperammonemia. Loss of PC prevented diet-induced hyperglycemia and insulin resistance but depleted NADPH and glutathione, which exacerbated oxidative stress and correlated with elevated liver inflammation. Thus, despite catalyzing the synthesis of intermediates also produced by other anaplerotic pathways, PC is specifically necessary for maintaining oxidation, biosynthesis, and pathways distal to the TCA cycle, such as antioxidant defenses.
Collapse
Affiliation(s)
- David A Cappel
- Center for Human Nutrition, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Stanisław Deja
- Center for Human Nutrition, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - João A G Duarte
- Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Blanka Kucejova
- Center for Human Nutrition, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Melissa Iñigo
- Center for Human Nutrition, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Justin A Fletcher
- Center for Human Nutrition, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xiaorong Fu
- Center for Human Nutrition, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Eric D Berglund
- Center for Hypothalamic Research, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Tiemin Liu
- Sate Key Laboratory of Genetic Engineering, School of Life Sciences, Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, Shanghai 200438, China
| | - Joel K Elmquist
- Center for Hypothalamic Research, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Suntrea Hammer
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Prashant Mishra
- Children's Medical Center Research Institute, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jeffrey D Browning
- Department of Clinical Nutrition, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shawn C Burgess
- Center for Human Nutrition, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Pharmacology, The University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
13
|
Carreau AM, Jin ES, Garcia-Reyes Y, Rahat H, Nadeau KJ, Malloy CR, Cree-Green M. A simple method to monitor hepatic gluconeogenesis and triglyceride synthesis following oral sugar tolerance test in obese adolescents. Am J Physiol Regul Integr Comp Physiol 2019; 317:R134-R142. [PMID: 31042400 DOI: 10.1152/ajpregu.00047.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Hepatic energy metabolism is a key element in many metabolic diseases. Hepatic anaplerosis provides carbons for gluconeogenesis (GNG) and triglyceride (TG) synthesis. We aimed to optimize a protocol that measures hepatic anaplerotic contribution for GNG, TG synthesis, and hepatic pentose phosphate pathway (PPP) activity using a single dose of oral [U-13C3]glycerol paired with an oral sugar tolerance test (OSTT) in a population with significant insulin resistance. The OSTT (75 g glucose + 25 g fructose) was administered to eight obese adolescents with polycystic ovarian syndrome (PCOS) followed by ingestion of [U-13C3]glycerol at t = 180 or t = 210 min. 13C-labeling patterns of serum glucose and TG-glycerol were determined by nuclear magnetic resonance. 13C enrichment in plasma TG-glycerol was detectable and stable from 240 to 390 min with the [U-13C3]glycerol drink at t = 180 min(3.65 ± 2.3 to 4.47 ± 1.4%; P > 0.4), but the enrichment was undetectable at 240 min with the glycerol drink at t = 210 min. The relative contribution from anaplerosis was determined at the end of the OSTT [18.5 ±3.4% (t = 180 min) vs. 16.0 ± 3.5% (t = 210 min); P = 0.27]. [U-13C3]glycerol was incorporated into GNG 390 min after the OSTT with an enrichment of 7.5-12.5%. Glucose derived from TCA cycle activity was 0.3-1%, and the PPP activity was 2.8-4.7%. In conclusion, it is possible to obtain relative measurements of hepatic anaplerotic contribution to both GNG and TG esterification following an OSTT in a highly insulin-resistant population using a minimally invasive technique. Tracer administration should be timed to allow enough de novo TG esterification and endogenous glucose release after the sugar drink.
Collapse
Affiliation(s)
- Anne-Marie Carreau
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Colorado Anschutz Medical Campus , Aurora, Colorado
| | - Eunsook S Jin
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center , Dallas, Texas
| | - Yesenia Garcia-Reyes
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Colorado Anschutz Medical Campus , Aurora, Colorado
| | - Haseeb Rahat
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Colorado Anschutz Medical Campus , Aurora, Colorado
| | - Kristen J Nadeau
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Colorado Anschutz Medical Campus , Aurora, Colorado.,Center for Women's Health Research , Aurora, Colorado
| | - Craig R Malloy
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center , Dallas, Texas
| | - Melanie Cree-Green
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Colorado Anschutz Medical Campus , Aurora, Colorado.,Center for Women's Health Research , Aurora, Colorado
| |
Collapse
|
14
|
Bian C, Bai B, Gao Q, Li S, Zhao Y. 17β-Estradiol Regulates Glucose Metabolism and Insulin Secretion in Rat Islet β Cells Through GPER and Akt/mTOR/GLUT2 Pathway. Front Endocrinol (Lausanne) 2019; 10:531. [PMID: 31447779 PMCID: PMC6691154 DOI: 10.3389/fendo.2019.00531] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 07/17/2019] [Indexed: 12/14/2022] Open
Abstract
Aims: To explore the molecular mechanism by which 17β-estradiol (estrogen 2, E2) regulates glucose transporter 2 (GLUT2) and insulin secretion in islet β cells through G protein-coupled estrogen receptor (GPER) via Akt/mTOR pathway. Methods: SPF-grade SD male rats were used to establish an in vivo type 2 diabetes model treated with E2. Rat insulinoma cells (INS-1) were cultured in normal or high glucose media with or without E2. Immunofluorescence double staining was used to detect GPER, GLUT2, insulin, and glucagon immunolocalization in rat islet tissues. Western blot was used to detect GPER, Akt, mTOR, and GLUT2 protein immunocontent. Real-time PCR detected Slc2a2 and glucose kinase (GK) content, and ELISA was used to detect insulin levels. Glucose uptake, GK activity and pyruvate dehydrogenase (PDH) activity were analyzed with glucose detection, GK activity and PDH activity assay kit. Results: Immunofluorescence double staining confocal indicated that E2 treatment up-regulated expression levels of GPER, GLUT2, and insulin, while down-regulated glucagon. Western blot results revealed E2 increased GPER, Akt/mTOR pathway, and GLUT2 protein immunocontent. Real-time PCR showed E2 elevated Slc2a2, GK content. Moreover, E2 improved insulin secretion, glucose uptake, GK activity, and PDH activity. Conclusion: Our findings indicated that exogenous E2 up-regulated GPER via the Akt/mTOR pathway to increase GLUT2 protein content and insulin secretion in islet β cells.
Collapse
Affiliation(s)
- Che Bian
- Department of Endocrinology, The First Affiliated Hospital of China Medical University, Shenyang, China
- Department of Endocrinology and Metabolism, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Bowen Bai
- Department of Endocrinology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Qian Gao
- Department of Endocrinology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Siyi Li
- Department of Endocrinology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yuyan Zhao
- Department of Endocrinology, The First Affiliated Hospital of China Medical University, Shenyang, China
- *Correspondence: Yuyan Zhao
| |
Collapse
|
15
|
An adapted isotope dilution 1H- 13C heteronuclear single-quantum correlation (ID-HSQC) for rapid and accurate quantification of endogenous and exogenous plasma glucose. Anal Bioanal Chem 2018; 410:6705-6711. [PMID: 30054692 DOI: 10.1007/s00216-018-1276-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 07/07/2018] [Accepted: 07/17/2018] [Indexed: 02/04/2023]
Abstract
A wide variety of methods, such as enzymatic methods, LC-MS, and LC-MS/MS, are currently available for the concentration determination of plasma glucose in studies of diabetes, obesity, exercise, etc. However, these methods rarely discriminate endogenous and exogenous glucose in plasma. A novel NMR strategy for discriminative quantification of the endogenous and exogenous glucose in plasma has been developed using an adapted isotope dilution 1H-13C heteronuclear single-quantum correlation (ID-HSQC) with uniformly 13C-labeled glucose as a tracer of exogenous glucose. This method takes advantage of the distinct 1H-13C chemical shifts of the hemiacetal group of the α-D-glucopyranose and makes use of the 13C-13C one-bond J-coupling (1JCC) in uniformly 13C-labeled glucose to differentiate the 1H-13C HSQC signal of labeled glucose from that of its natural counterpart when data are acquired in high-resolution mode. The molar ratio between the endogenous and exogenous plasma glucose can then be calculated from the peak intensities of the natural and labeled glucose. The accuracy and precision of the method were evaluated using a series of standard mixtures of natural and uniformly 13C-labeled glucose with varied but known concentrations. Application of this method is demonstrated for the quantification of endogenous and exogenous glucose in plasma derived from healthy and diabetic cynomolgus monkeys. The results nicely agree with our previous LC-MS/MS results. Considering the natural abundance of 13C isotope at the level of 1.1% in endogenous glucose, comparable peak intensities of quantitatively measurable signals derived from natural and labeled glucose imply that the ID-HSQC can tolerate a significantly high ratio of isotope dilution, with labeled/natural glucose at ~ 1%. We expect that the ID-HSQC method can serve as an alternative approach to the biomedical or clinical studies of glucose metabolism.
Collapse
|
16
|
Jin ES, Browning JD, Murphy RE, Malloy CR. Fatty liver disrupts glycerol metabolism in gluconeogenic and lipogenic pathways in humans. J Lipid Res 2018; 59:1685-1694. [PMID: 30054343 PMCID: PMC6121920 DOI: 10.1194/jlr.m086405] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 07/27/2018] [Indexed: 12/29/2022] Open
Abstract
It is a challenge to assess metabolic dysregulation in fatty liver of human patients prior to clinical manifestations. Here, we recruited obese, but otherwise healthy, subjects to examine biochemical processes in the liver with simple triglyceride accumulation using stable isotopes and NMR analysis of metabolic products in blood. Intrahepatic triglycerides were measured using 1H magnetic resonance spectroscopy, and volunteers received 2H2O and [U-13C3]glycerol orally, followed by a series of blood draws. NMR analysis of plasma triglycerides and glucose provided detailed information about metabolic pathways in patients with simple hepatic steatosis. Compared with subjects with low hepatic fat, patients with hepatic steatosis were characterized by the following: lower 13C enrichments in the glycerol backbones of triglycerides (i.e., TG-[13C]glycerol), higher [U-13C3]glycerol metabolism through the tricarboxylic acid (TCA) cycle, delayed gluconeogenesis from [U-13C3]glycerol, and less flexibility in adjusting supporting fluxes of glucose production upon an oral load of glycerol. In summary, simple hepatic steatosis was associated with enhanced [U-13C3]glycerol metabolism through pathways that intersect the TCA cycle and delayed gluconeogenesis from glycerol.
Collapse
Affiliation(s)
- Eunsook S Jin
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390.
| | - Jeffrey D Browning
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390; Department of Clinical Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Rebecca E Murphy
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Craig R Malloy
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390; Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390; Veterans Affairs North Texas Health Care System, Dallas, TX 75216
| |
Collapse
|
17
|
d'Avignon DA, Puchalska P, Ercal B, Chang Y, Martin SE, Graham MJ, Patti GJ, Han X, Crawford PA. Hepatic ketogenic insufficiency reprograms hepatic glycogen metabolism and the lipidome. JCI Insight 2018; 3:99762. [PMID: 29925686 DOI: 10.1172/jci.insight.99762] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 05/17/2018] [Indexed: 02/06/2023] Open
Abstract
While several molecular targets are under consideration, mechanistic underpinnings of the transition from uncomplicated nonalcoholic fatty liver disease (NAFLD) to nonalcoholic steatohepatitis (NASH) remain unresolved. Here we apply multiscale chemical profiling technologies to mouse models of deranged hepatic ketogenesis to uncover potential NAFLD driver signatures. Use of stable-isotope tracers, quantitatively tracked by nuclear magnetic resonance (NMR) spectroscopy, supported previous observations that livers of wild-type mice maintained long term on a high-fat diet (HFD) exhibit a marked increase in hepatic energy charge. Fed-state ketogenesis rates increased nearly 3-fold in livers of HFD-fed mice, a greater proportionate increase than that observed for tricarboxylic acid (TCA) cycle flux, but both of these contributors to overall hepatic energy homeostasis fueled markedly increased hepatic glucose production (HGP). Thus, to selectively determine the role of the ketogenic conduit on HGP and oxidative hepatic fluxes, we studied a ketogenesis-insufficient mouse model generated by knockdown of the mitochondrial isoform of 3-hydroxymethylglutaryl-CoA synthase (HMGCS2). In response to ketogenic insufficiency, TCA cycle flux in the fed state doubled and HGP increased more than 60%, sourced by a 3-fold increase in glycogenolysis. Finally, high-resolution untargeted metabolomics and shotgun lipidomics performed using ketogenesis-insufficient livers in the fed state revealed accumulation of bis(monoacylglycero)phosphates, which also accumulated in livers of other models commonly used to study NAFLD. In summary, natural and interventional variations in ketogenesis in the fed state strongly influence hepatic energy homeostasis, glucose metabolism, and the lipidome. Importantly, HGP remains tightly linked to overall hepatic energy charge, which includes both terminal fat oxidation through the TCA cycle and partial oxidation via ketogenesis.
Collapse
Affiliation(s)
- D André d'Avignon
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA.,Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA
| | - Patrycja Puchalska
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA.,Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA
| | - Baris Ercal
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA.,Department of Psychiatry, Washington University, St. Louis, Missouri, USA
| | - YingJu Chang
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA.,Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA
| | - Shannon E Martin
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA.,Pathobiology Graduate Program, Brown University, Providence, Rhode Island, USA
| | | | - Gary J Patti
- Department of Chemistry, Washington University, St. Louis, Missouri, USA
| | - Xianlin Han
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA.,Barshop Institute for Longevity and Aging Studies, Department of Medicine, Division of Diabetes, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Peter A Crawford
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA.,Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA.,Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
18
|
Atzrodt J, Derdau V, Kerr WJ, Reid M. Deuterium- und tritiummarkierte Verbindungen: Anwendungen in den modernen Biowissenschaften. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201704146] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Jens Atzrodt
- Isotope Chemistry and Metabolite Synthesis, Integrated Drug Discovery, Medicinal Chemistry; Industriepark Höchst, G876 65926 Frankfurt Deutschland
| | - Volker Derdau
- Isotope Chemistry and Metabolite Synthesis, Integrated Drug Discovery, Medicinal Chemistry; Industriepark Höchst, G876 65926 Frankfurt Deutschland
| | - William J. Kerr
- Department of Pure and Applied Chemistry, WestCHEM; University of Strathclyde; 295 Cathedral Street Glasgow Scotland G1 1XL Großbritannien
| | - Marc Reid
- Department of Pure and Applied Chemistry, WestCHEM; University of Strathclyde; 295 Cathedral Street Glasgow Scotland G1 1XL Großbritannien
| |
Collapse
|
19
|
Atzrodt J, Derdau V, Kerr WJ, Reid M. Deuterium- and Tritium-Labelled Compounds: Applications in the Life Sciences. Angew Chem Int Ed Engl 2018; 57:1758-1784. [PMID: 28815899 DOI: 10.1002/anie.201704146] [Citation(s) in RCA: 421] [Impact Index Per Article: 70.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 07/27/2017] [Indexed: 12/19/2022]
Abstract
Hydrogen isotopes are unique tools for identifying and understanding biological and chemical processes. Hydrogen isotope labelling allows for the traceless and direct incorporation of an additional mass or radioactive tag into an organic molecule with almost no changes in its chemical structure, physical properties, or biological activity. Using deuterium-labelled isotopologues to study the unique mass-spectrometric patterns generated from mixtures of biologically relevant molecules drastically simplifies analysis. Such methods are now providing unprecedented levels of insight in a wide and continuously growing range of applications in the life sciences and beyond. Tritium (3 H), in particular, has seen an increase in utilization, especially in pharmaceutical drug discovery. The efforts and costs associated with the synthesis of labelled compounds are more than compensated for by the enhanced molecular sensitivity during analysis and the high reliability of the data obtained. In this Review, advances in the application of hydrogen isotopes in the life sciences are described.
Collapse
Affiliation(s)
- Jens Atzrodt
- Isotope Chemistry and Metabolite Synthesis, Integrated Drug Discovery, Medicinal Chemistry, Industriepark Höchst, G876, 65926, Frankfurt, Germany
| | - Volker Derdau
- Isotope Chemistry and Metabolite Synthesis, Integrated Drug Discovery, Medicinal Chemistry, Industriepark Höchst, G876, 65926, Frankfurt, Germany
| | - William J Kerr
- Department of Pure and Applied Chemistry, WestCHEM, University of Strathclyde, 295 Cathedral Street, Glasgow, Scotland, G1 1XL, UK
| | - Marc Reid
- Department of Pure and Applied Chemistry, WestCHEM, University of Strathclyde, 295 Cathedral Street, Glasgow, Scotland, G1 1XL, UK
| |
Collapse
|
20
|
Carvalho RA, Jarak I. Mitochondrial Bioenergetics by 13C-NMR Isotopomer Analysis. Methods Mol Biol 2018; 1782:229-247. [PMID: 29851003 DOI: 10.1007/978-1-4939-7831-1_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Metabolic reprogramming has been associated to a plethora of diseases, and there has been increased demand for methodologies able to determine the metabolic alterations that characterize the pathological states and help developing metabolically centered therapies. In this chapter, methodologies for monitoring TCA cycle turnover and its interaction with pyruvate cycling and anaplerotic reactions will be presented. These methodologies are based in the application of stable 13C isotope "tracers"/substrates and 13C-NMR isotopomer analysis of metabolic intermediates. These methodologies can be applied at several organizational levels, ranging from isolated organelles and organs to whole organisms/humans. For the sake of simplicity, only very simple and well-defined models will be presented, including isolated heart mitochondria and isolated perfused hearts and livers.
Collapse
Affiliation(s)
- Rui A Carvalho
- Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal.
- Centre for Functional Ecology, University of Coimbra, Coimbra, Portugal.
| | - Ivana Jarak
- Centre for Functional Ecology, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
21
|
Pachanski MJ, Kirkland ME, Kosinski DT, Mane J, Cheewatrakoolpong B, Xue J, Szeto D, Forrest G, Miller C, Bunzel M, Plummer CW, Chobanian HR, Miller MW, Souza S, Thomas-Fowlkes BS, Ogawa AM, Weinglass AB, Di Salvo J, Li X, Feng Y, Tatosian DA, Howard AD, Colletti SL, Trujillo ME. GPR40 partial agonists and AgoPAMs: Differentiating effects on glucose and hormonal secretions in the rodent. PLoS One 2017; 12:e0186033. [PMID: 29053717 PMCID: PMC5650142 DOI: 10.1371/journal.pone.0186033] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 09/23/2017] [Indexed: 01/14/2023] Open
Abstract
GPR40 agonists are effective antidiabetic agents believed to lower glucose through direct effects on the beta cell to increase glucose stimulated insulin secretion. However, not all GPR40 agonists are the same. Partial agonists lower glucose through direct effects on the pancreas, whereas GPR40 AgoPAMs may incorporate additional therapeutic effects through increases in insulinotrophic incretins secreted by the gut. Here we describe how GPR40 AgoPAMs stimulate both insulin and incretin secretion in vivo over time in diabetic GK rats. We also describe effects of AgoPAMs in vivo to lower glucose and body weight beyond what is seen with partial GPR40 agonists in both the acute and chronic setting. Further comparisons of the glucose lowering profile of AgoPAMs suggest these compounds may possess greater glucose control even in the presence of elevated glucagon secretion, an unexpected feature observed with both acute and chronic treatment with AgoPAMs. Together these studies highlight the complexity of GPR40 pharmacology and the potential additional benefits AgoPAMs may possess above partial agonists for the diabetic patient.
Collapse
Affiliation(s)
- Michele J. Pachanski
- In Vivo Pharmacology, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Melissa E. Kirkland
- In Vivo Pharmacology, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Daniel T. Kosinski
- In Vivo Pharmacology, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Joel Mane
- In Vivo Pharmacology, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | | | - Jiyan Xue
- In Vivo Pharmacology, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Daphne Szeto
- In Vivo Pharmacology, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Gail Forrest
- In Vivo Pharmacology, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Corin Miller
- Translational Imaging Biomarkers, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Michelle Bunzel
- Translational Imaging Biomarkers, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Christopher W. Plummer
- Department of Medicinal Chemistry, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Harry R. Chobanian
- Department of Medicinal Chemistry, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Michael W. Miller
- Department of Medicinal Chemistry, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Sarah Souza
- In Vivo Pharmacology, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | | | - Aimie M. Ogawa
- In Vivo Pharmacology, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Adam B. Weinglass
- In Vivo Pharmacology, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Jerry Di Salvo
- In Vivo Pharmacology, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Xiaoyan Li
- Department of Cardio Metabolic Diseases, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Yue Feng
- Department of Cardio Metabolic Diseases, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Daniel A. Tatosian
- Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Andrew D. Howard
- Department of Cardio Metabolic Diseases, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Steven L. Colletti
- Department of Medicinal Chemistry, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Maria E. Trujillo
- In Vivo Pharmacology, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
- * E-mail:
| |
Collapse
|
22
|
Non-invasive assessment of hepatic mitochondrial metabolism by positional isotopomer NMR tracer analysis (PINTA). Nat Commun 2017; 8:798. [PMID: 28986525 PMCID: PMC5630596 DOI: 10.1038/s41467-017-01143-w] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 08/22/2017] [Indexed: 01/03/2023] Open
Abstract
Hepatic mitochondria play a central role in the regulation of intermediary metabolism and maintenance of normoglycemia, and there is great interest in assessing rates of hepatic mitochondrial citrate synthase flux (VCS) and pyruvate carboxylase flux (VPC) in vivo. Here, we show that a positional isotopomer NMR tracer analysis (PINTA) method can be used to non-invasively assess rates of VCS and VPC fluxes using a combined NMR/gas chromatography-mass spectrometry analysis of plasma following infusion of [3-13C]lactate and glucose tracer. PINTA measures VCS and VPC fluxes over a wide range of physiological conditions with minimal pyruvate cycling and detects increased hepatic VCS following treatment with a liver-targeted mitochondrial uncoupler. Finally, validation studies in humans demonstrate that the VPC/VCS ratio measured by PINTA is similar to that determined by in vivo NMR spectroscopy. This method will provide investigators with a relatively simple tool to non-invasively examine the role of altered hepatic mitochondrial metabolism. Liver mitochondrial metabolism plays an important role for glucose and lipid homeostasis and its alterations contribute to metabolic disorders, including fatty liver and diabetes. Here Perry et al. develop a method for the measurement of hepatic fluxes by using lactate and glucose tracers in combination with NMR spectroscopy.
Collapse
|
23
|
Miller C, Pachanski MJ, Kirkland ME, Kosinski DT, Mane J, Bunzel M, Cao J, Souza S, Thomas-Fowlkes B, Di Salvo J, Weinglass AB, Li X, Myers RW, Knagge K, Carrington PE, Hagmann WK, Trujillo ME. GPR40 partial agonist MK-2305 lower fasting glucose in the Goto Kakizaki rat via suppression of endogenous glucose production. PLoS One 2017; 12:e0176182. [PMID: 28542610 PMCID: PMC5441580 DOI: 10.1371/journal.pone.0176182] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 04/06/2017] [Indexed: 11/19/2022] Open
Abstract
GPR40 (FFA1) is a fatty acid receptor whose activation results in potent glucose lowering and insulinotropic effects in vivo. Several reports illustrate that GPR40 agonists exert glucose lowering in diabetic humans. To assess the mechanisms by which GPR40 partial agonists improve glucose homeostasis, we evaluated the effects of MK-2305, a potent and selective partial GPR40 agonist, in diabetic Goto Kakizaki rats. MK-2305 decreased fasting glucose after acute and chronic treatment. MK-2305-mediated changes in glucose were coupled with increases in plasma insulin during hyperglycemia and glucose challenges but not during fasting, when glucose was normalized. To determine the mechanism(s) mediating these changes in glucose metabolism, we measured the absolute contribution of precursors to glucose production in the presence or absence of MK-2305. MK-2305 treatment resulted in decreased endogenous glucose production (EGP) driven primarily through changes in gluconeogenesis from substrates entering at the TCA cycle. The decrease in EGP was not likely due to a direct effect on the liver, as isolated perfused liver studies showed no effect of MK-2305 ex vivo and GPR40 is not expressed in the liver. Taken together, our results suggest MK-2305 treatment increases glucose stimulated insulin secretion (GSIS), resulting in changes to hepatic substrate handling that improve glucose homeostasis in the diabetic state. Importantly, these data extend our understanding of the underlying mechanisms by which GPR40 partial agonists reduce hyperglycemia.
Collapse
Affiliation(s)
- Corin Miller
- Departments of Translational Imaging Biomarkers, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Michele J. Pachanski
- In Vivo Pharmacology, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Melissa E. Kirkland
- In Vivo Pharmacology, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Daniel T. Kosinski
- In Vivo Pharmacology, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Joel Mane
- In Vivo Pharmacology, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Michelle Bunzel
- Departments of Translational Imaging Biomarkers, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Jin Cao
- Departments of Translational Imaging Biomarkers, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Sarah Souza
- In Vitro Pharmacology, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Brande Thomas-Fowlkes
- In Vitro Pharmacology, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Jerry Di Salvo
- In Vitro Pharmacology, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Adam B. Weinglass
- In Vitro Pharmacology, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Xiaoyan Li
- Cardio-Metabolic Diseases, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Robert W. Myers
- In Vitro Pharmacology, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Kevin Knagge
- David H Murdock Research Institute, Kannapolis, North Carolina, United States of America
| | - Paul E. Carrington
- Cardio-Metabolic Diseases, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - William K. Hagmann
- Chemistry, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Maria E. Trujillo
- In Vivo Pharmacology, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
- * E-mail:
| |
Collapse
|
24
|
Neeland IJ, Hughes C, Ayers CR, Malloy CR, Jin ES. Effects of visceral adiposity on glycerol pathways in gluconeogenesis. Metabolism 2017; 67:80-89. [PMID: 28081781 PMCID: PMC5244471 DOI: 10.1016/j.metabol.2016.11.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 10/27/2016] [Accepted: 11/22/2016] [Indexed: 01/23/2023]
Abstract
OBJECTIVE To determine the feasibility of using oral 13C labeled glycerol to assess effects of visceral adiposity on gluconeogenic pathways in obese humans. RESEARCH DESIGN AND METHODS Obese (BMI ≥30kg/m2) participants without type 2 diabetes underwent visceral adipose tissue (VAT) assessment and stratification by median VAT into high VAT-fasting (n=3), low VAT-fasting (n=4), and high VAT-refed (n=2) groups. Participants ingested [U-13C3] glycerol and blood samples were subsequently analyzed at multiple time points over 3h by NMR spectroscopy. The fractions of plasma glucose (enrichment) derived from [U-13C3] glycerol via hepatic gluconeogenesis, pentose phosphate pathway (PPP), and tricarboxylic acid (TCA) cycle were assessed using 13C NMR analysis of glucose. Mixed linear models were used to compare 13C enrichment in glucose between groups. RESULTS Mean age, BMI, and baseline glucose were 49years, 40.1kg/m2, and 98mg/dl, respectively. Up to 20% of glycerol was metabolized in the TCA cycle prior to gluconeogenesis and PPP activity was minor (<1% of total glucose) in all participants. There was a 21% decrease in 13C enrichment in plasma glucose in the high VAT-fasting compared with low VAT-fasting group (p=0.03), suggesting dilution by endogenous glycerol. High VAT-refed participants had 37% less 13C enrichment in glucose compared with high VAT-fasting (p=0.02). There was a trend toward lower [1,2-13C2] (via PPP) and [5,6-13C2]/[4,5,6-13C3] (via TCA cycle) glucose in high VAT versus low VAT groups. CONCLUSIONS We applied a simple method to detect gluconeogenesis from glycerol in obese humans. Our findings provide preliminary evidence that excess visceral fat disrupts multiple pathways in hepatic gluconeogenesis from glycerol.
Collapse
Affiliation(s)
- Ian J Neeland
- Department of Internal Medicine, Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Connor Hughes
- Department of Internal Medicine, Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Colby R Ayers
- Department of Clinical Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Craig R Malloy
- Advanced Imaging Research Center and Departments of Internal Medicine and Radiology, University of Texas Southwestern Medical Center and VA North Texas Healthcare System, Dallas, TX, USA
| | - Eunsook S Jin
- Advanced Imaging Research Center and Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
25
|
Jin ES, Sherry AD, Malloy CR. An Oral Load of [13C3]Glycerol and Blood NMR Analysis Detect Fatty Acid Esterification, Pentose Phosphate Pathway, and Glycerol Metabolism through the Tricarboxylic Acid Cycle in Human Liver. J Biol Chem 2016; 291:19031-41. [PMID: 27432878 DOI: 10.1074/jbc.m116.742262] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Indexed: 11/06/2022] Open
Abstract
Drugs and other interventions for high impact hepatic diseases often target biochemical pathways such as gluconeogenesis, lipogenesis, or the metabolic response to oxidative stress. However, traditional liver function tests do not provide quantitative data about these pathways. In this study, we developed a simple method to evaluate these processes by NMR analysis of plasma metabolites. Healthy subjects ingested [U-(13)C3]glycerol, and blood was drawn at multiple times. Each subject completed three visits under differing nutritional states. High resolution (13)C NMR spectra of plasma triacylglycerols and glucose provided new insights into a number of hepatic processes including fatty acid esterification, the pentose phosphate pathway, and gluconeogenesis through the tricarboxylic acid cycle. Fasting stimulated pentose phosphate pathway activity and metabolism of [U-(13)C3]glycerol in the tricarboxylic acid cycle prior to gluconeogenesis or glyceroneogenesis. Fatty acid esterification was transient in the fasted state but continuous under fed conditions. We conclude that a simple NMR analysis of blood metabolites provides an important biomarker of pentose phosphate pathway activity, triacylglycerol synthesis, and flux through anaplerotic pathways in mitochondria of human liver.
Collapse
Affiliation(s)
- Eunsook S Jin
- From the Advanced Imaging Research Center and the Departments of Internal Medicine and
| | - A Dean Sherry
- From the Advanced Imaging Research Center and Radiology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390, the Department of Chemistry, University of Texas at Dallas, Richardson, Texas 75080, and
| | - Craig R Malloy
- From the Advanced Imaging Research Center and the Departments of Internal Medicine and Radiology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390, the VA North Texas Health Care System, Dallas, Texas 75216
| |
Collapse
|
26
|
Assessment of Hepatic Mitochondrial Oxidation and Pyruvate Cycling in NAFLD by (13)C Magnetic Resonance Spectroscopy. Cell Metab 2016; 24:167-71. [PMID: 27411016 PMCID: PMC4946568 DOI: 10.1016/j.cmet.2016.06.005] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Revised: 05/07/2016] [Accepted: 06/08/2016] [Indexed: 01/02/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease, and there is great interest in understanding the potential role of alterations in mitochondrial metabolism in its pathogenesis. To address this question, we assessed rates of hepatic mitochondrial oxidation in subjects with and without NAFLD by monitoring the rate of (13)C labeling in hepatic [5-(13)C]glutamate and [1-(13)C]glutamate by (13)C MRS during an infusion of [1-(13)C]acetate. We found that rates of hepatic mitochondrial oxidation were similar between NAFLD and control subjects. We also assessed rates of hepatic pyruvate cycling during an infusion of [3-(13)C]lactate by monitoring the (13)C label in hepatic [2-(13)C]alanine and [2-(13)C]glutamate and found that this flux was also similar between groups and more than 10-fold lower than previously reported. Contrary to previous studies, we show that hepatic mitochondrial oxidation and pyruvate cycling are not altered in NAFLD and do not account for the hepatic fat accumulation.
Collapse
|
27
|
Shestov AA, Lee SC, Nath K, Guo L, Nelson DS, Roman JC, Leeper DB, Wasik MA, Blair IA, Glickson JD. (13)C MRS and LC-MS Flux Analysis of Tumor Intermediary Metabolism. Front Oncol 2016; 6:135. [PMID: 27379200 PMCID: PMC4908130 DOI: 10.3389/fonc.2016.00135] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 05/23/2016] [Indexed: 01/09/2023] Open
Abstract
We present the first validated metabolic network model for analysis of flux through key pathways of tumor intermediary metabolism, including glycolysis, the oxidative and non-oxidative arms of the pentose pyrophosphate shunt, the TCA cycle as well as its anaplerotic pathways, pyruvate-malate shuttling, glutaminolysis, and fatty acid biosynthesis and oxidation. The model that is called Bonded Cumomer Analysis for application to (13)C magnetic resonance spectroscopy ((13)C MRS) data and Fragmented Cumomer Analysis for mass spectrometric data is a refined and efficient form of isotopomer analysis that can readily be expanded to incorporate glycogen, phospholipid, and other pathways thereby encompassing all the key pathways of tumor intermediary metabolism. Validation was achieved by demonstrating agreement of experimental measurements of the metabolic rates of oxygen consumption, glucose consumption, lactate production, and glutamate pool size with independent measurements of these parameters in cultured human DB-1 melanoma cells. These cumomer models have been applied to studies of DB-1 melanoma and DLCL2 human diffuse large B-cell lymphoma cells in culture and as xenografts in nude mice at 9.4 T. The latter studies demonstrate the potential translation of these methods to in situ studies of human tumor metabolism by MRS with stable (13)C isotopically labeled substrates on instruments operating at high magnetic fields (≥7 T). The melanoma studies indicate that this tumor line obtains 51% of its ATP by mitochondrial metabolism and 49% by glycolytic metabolism under both euglycemic (5 mM glucose) and hyperglycemic conditions (26 mM glucose). While a high level of glutamine uptake is detected corresponding to ~50% of TCA cycle flux under hyperglycemic conditions, and ~100% of TCA cycle flux under euglycemic conditions, glutaminolysis flux and its contributions to ATP synthesis were very small. Studies of human lymphoma cells demonstrated that inhibition of mammalian target of rapamycin (mTOR) signaling produced changes in flux through the glycolytic, pentose shunt, and TCA cycle pathways that were evident within 8 h of treatment and increased at 24 and 48 h. Lactate was demonstrated to be a suitable biomarker of mTOR inhibition that could readily be monitored by (1)H MRS and perhaps also by FDG-PET and hyperpolarized (13)C MRS methods.
Collapse
Affiliation(s)
- Alexander A Shestov
- Laboratory of Molecular Imaging, Department of Radiology, Perelman School of Medicine, University of Pennsylvania , Philadelphia, PA , USA
| | - Seung-Cheol Lee
- Laboratory of Molecular Imaging, Department of Radiology, Perelman School of Medicine, University of Pennsylvania , Philadelphia, PA , USA
| | - Kavindra Nath
- Laboratory of Molecular Imaging, Department of Radiology, Perelman School of Medicine, University of Pennsylvania , Philadelphia, PA , USA
| | - Lili Guo
- Department of Systems Pharmacology and Translational Therapeutics, Center for Cancer Pharmacology, Perelman School of Medicine, University of Pennsylvania , Philadelphia, PA , USA
| | - David S Nelson
- Laboratory of Molecular Imaging, Department of Radiology, Perelman School of Medicine, University of Pennsylvania , Philadelphia, PA , USA
| | - Jeffrey C Roman
- Laboratory of Molecular Imaging, Department of Radiology, Perelman School of Medicine, University of Pennsylvania , Philadelphia, PA , USA
| | - Dennis B Leeper
- Department of Radiation Oncology, Thomas Jefferson University , Philadelphia, PA , USA
| | - Mariusz A Wasik
- Laboratory Medicine, Department of Pathology, Perelman School of Medicine, University of Pennsylvania , Philadelphia, PA , USA
| | - Ian A Blair
- Department of Systems Pharmacology and Translational Therapeutics, Center for Cancer Pharmacology, Perelman School of Medicine, University of Pennsylvania , Philadelphia, PA , USA
| | - Jerry D Glickson
- Laboratory of Molecular Imaging, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
28
|
Patterson RE, Kalavalapalli S, Williams CM, Nautiyal M, Mathew JT, Martinez J, Reinhard MK, McDougall DJ, Rocca JR, Yost RA, Cusi K, Garrett TJ, Sunny NE. Lipotoxicity in steatohepatitis occurs despite an increase in tricarboxylic acid cycle activity. Am J Physiol Endocrinol Metab 2016; 310:E484-94. [PMID: 26814015 PMCID: PMC4824140 DOI: 10.1152/ajpendo.00492.2015] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 01/25/2016] [Indexed: 02/07/2023]
Abstract
The hepatic tricarboxylic acid (TCA) cycle is central to integrating macronutrient metabolism and is closely coupled to cellular respiration, free radical generation, and inflammation. Oxidative flux through the TCA cycle is induced during hepatic insulin resistance, in mice and humans with simple steatosis, reflecting early compensatory remodeling of mitochondrial energetics. We hypothesized that progressive severity of hepatic insulin resistance and the onset of nonalcoholic steatohepatitis (NASH) would impair oxidative flux through the hepatic TCA cycle. Mice (C57/BL6) were fed a high-trans-fat high-fructose diet (TFD) for 8 wk to induce simple steatosis and NASH by 24 wk. In vivo fasting hepatic mitochondrial fluxes were determined by(13)C-nuclear magnetic resonance (NMR)-based isotopomer analysis. Hepatic metabolic intermediates were quantified using mass spectrometry-based targeted metabolomics. Hepatic triglyceride accumulation and insulin resistance preceded alterations in mitochondrial metabolism, since TCA cycle fluxes remained normal during simple steatosis. However, mice with NASH had a twofold induction (P< 0.05) of mitochondrial fluxes (μmol/min) through the TCA cycle (2.6 ± 0.5 vs. 5.4 ± 0.6), anaplerosis (9.1 ± 1.2 vs. 16.9 ± 2.2), and pyruvate cycling (4.9 ± 1.0 vs. 11.1 ± 1.9) compared with their age-matched controls. Induction of the TCA cycle activity during NASH was concurrent with blunted ketogenesis and accumulation of hepatic diacylglycerols (DAGs), ceramides (Cer), and long-chain acylcarnitines, suggesting inefficient oxidation and disposal of excess free fatty acids (FFA). Sustained induction of mitochondrial TCA cycle failed to prevent accretion of "lipotoxic" metabolites in the liver and could hasten inflammation and the metabolic transition to NASH.
Collapse
Affiliation(s)
| | - Srilaxmi Kalavalapalli
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Florida, Gainesville, Florida
| | - Caroline M Williams
- Department of Integrative Biology, University of California, Berkeley, California
| | - Manisha Nautiyal
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Florida, Gainesville, Florida
| | - Justin T Mathew
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Florida, Gainesville, Florida
| | - Janie Martinez
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Florida, Gainesville, Florida
| | - Mary K Reinhard
- Animal Care Services, University of Florida, Gainesville, Florida
| | | | - James R Rocca
- Advanced Magnetic Resonance Imaging and Spectroscopy Facility, McKnight Brain Institute, University of Florida, Gainesville, Florida
| | - Richard A Yost
- Department of Chemistry, University of Florida, Gainesville, Florida; Department of Pathology, University of Florida, Gainesville, Florida
| | - Kenneth Cusi
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Florida, Gainesville, Florida; Division of Endocrinology, Diabetes, and Metabolism, Malcom Randall Veterans Administration Medical Center, Gainesville, Florida; Division of Diabetes, the University of Texas Health Science Center at San Antonio, San Antonio, Texas; Division of Diabetes, Audie L. Murphy Veterans Administration Medical Center, San Antonio, Texas; and
| | - Timothy J Garrett
- Department of Pathology, University of Florida, Gainesville, Florida
| | - Nishanth E Sunny
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Florida, Gainesville, Florida;
| |
Collapse
|
29
|
Jin ES, Moreno KX, Wang JX, Fidelino L, Merritt ME, Sherry AD, Malloy CR. Metabolism of hyperpolarized [1-(13)C]pyruvate through alternate pathways in rat liver. NMR IN BIOMEDICINE 2016; 29:466-74. [PMID: 26836042 PMCID: PMC4805436 DOI: 10.1002/nbm.3479] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 11/18/2015] [Accepted: 12/04/2015] [Indexed: 05/07/2023]
Abstract
The source of hyperpolarized (HP) [(13)C]bicarbonate in the liver during metabolism of HP [1-(13)C]pyruvate is uncertain and likely changes with physiology. Multiple processes including decarboxylation through pyruvate dehydrogenase or pyruvate carboxylase followed by subsequent decarboxylation via phosphoenolpyruvate carboxykinase (gluconeogenesis) could play a role. Here we tested which metabolic fate of pyruvate contributed to the appearance of HP [(13)C]bicarbonate during metabolism of HP [1-(13)C]pyruvate by the liver in rats after 21 h of fasting compared to rats with free access to food. The (13)C NMR of HP [(13)C]bicarbonate was observed in the liver of fed rats, but not in fasted rats where pyruvate carboxylation and gluconeogenesis was active. To further explore the relative fluxes through pyruvate carboxylase versus pyruvate dehydrogenase in the liver under typical conditions of hyperpolarization studies, separate parallel experiments were performed with rats given non-hyperpolarized [2,3-(13)C]pyruvate. (13)C NMR analysis of glutamate isolated from the liver of rats revealed that flux from injected pyruvate through pyruvate dehydrogenase was dominant under fed conditions whereas flux through pyruvate carboxylase dominated under fasted conditions. The NMR signal of HP [(13)C]bicarbonate does not parallel pyruvate carboxylase activity followed by subsequent decarboxylation reaction leading to glucose production. In the liver of healthy well-fed rats, the appearance of HP [(13)C]bicarbonate exclusively reflects decarboxylation of HP [1-(13)C]pyruvate via pyruvate dehydrogenase.
Collapse
Affiliation(s)
- Eunsook S Jin
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Karlos X Moreno
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Jian-Xiong Wang
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Radiology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Leila Fidelino
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Matthew E Merritt
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Radiology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - A Dean Sherry
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Radiology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Chemistry, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Craig R Malloy
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Radiology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- VA North Texas Health Care System, Dallas, TX, 75216, USA
| |
Collapse
|
30
|
Perry RJ, Borders CB, Cline GW, Zhang XM, Alves TC, Petersen KF, Rothman DL, Kibbey RG, Shulman GI. Propionate Increases Hepatic Pyruvate Cycling and Anaplerosis and Alters Mitochondrial Metabolism. J Biol Chem 2016; 291:12161-70. [PMID: 27002151 PMCID: PMC4933266 DOI: 10.1074/jbc.m116.720631] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Indexed: 02/03/2023] Open
Abstract
In mammals, pyruvate kinase (PK) plays a key role in regulating the balance between glycolysis and gluconeogenesis; however, in vivo regulation of PK flux by gluconeogenic hormones and substrates is poorly understood. To this end, we developed a novel NMR-liquid chromatography/tandem-mass spectrometry (LC-MS/MS) method to directly assess pyruvate cycling relative to mitochondrial pyruvate metabolism (VPyr-Cyc/VMito) in vivo using [3-(13)C]lactate as a tracer. Using this approach, VPyr-Cyc/VMito was only 6% in overnight fasted rats. In contrast, when propionate was infused simultaneously at doses previously used as a tracer, it increased VPyr-Cyc/VMito by 20-30-fold, increased hepatic TCA metabolite concentrations 2-3-fold, and increased endogenous glucose production rates by 20-100%. The physiologic stimuli, glucagon and epinephrine, both increased hepatic glucose production, but only glucagon suppressed VPyr-Cyc/VMito These data show that under fasting conditions, when hepatic gluconeogenesis is stimulated, pyruvate recycling is relatively low in liver compared with VMito flux and that liver metabolism, in particular pyruvate cycling, is sensitive to propionate making it an unsuitable tracer to assess hepatic glycolytic, gluconeogenic, and mitochondrial metabolism in vivo.
Collapse
Affiliation(s)
| | | | | | | | | | - Kitt Falk Petersen
- From the Departments of Internal Medicine, The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen DK 1017, Denmark
| | - Douglas L Rothman
- Radiology and Biomedical Imaging, and the Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06519, and
| | - Richard G Kibbey
- From the Departments of Internal Medicine, Cellular and Molecular Physiology, and
| | - Gerald I Shulman
- From the Departments of Internal Medicine, The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen DK 1017, Denmark Cellular and Molecular Physiology, and the Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut 06519,
| |
Collapse
|
31
|
Fan TWM, Lane AN. Applications of NMR spectroscopy to systems biochemistry. PROGRESS IN NUCLEAR MAGNETIC RESONANCE SPECTROSCOPY 2016; 92-93:18-53. [PMID: 26952191 PMCID: PMC4850081 DOI: 10.1016/j.pnmrs.2016.01.005] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 01/26/2016] [Accepted: 01/28/2016] [Indexed: 05/05/2023]
Abstract
The past decades of advancements in NMR have made it a very powerful tool for metabolic research. Despite its limitations in sensitivity relative to mass spectrometric techniques, NMR has a number of unparalleled advantages for metabolic studies, most notably the rigor and versatility in structure elucidation, isotope-filtered selection of molecules, and analysis of positional isotopomer distributions in complex mixtures afforded by multinuclear and multidimensional experiments. In addition, NMR has the capacity for spatially selective in vivo imaging and dynamical analysis of metabolism in tissues of living organisms. In conjunction with the use of stable isotope tracers, NMR is a method of choice for exploring the dynamics and compartmentation of metabolic pathways and networks, for which our current understanding is grossly insufficient. In this review, we describe how various direct and isotope-edited 1D and 2D NMR methods can be employed to profile metabolites and their isotopomer distributions by stable isotope-resolved metabolomic (SIRM) analysis. We also highlight the importance of sample preparation methods including rapid cryoquenching, efficient extraction, and chemoselective derivatization to facilitate robust and reproducible NMR-based metabolomic analysis. We further illustrate how NMR has been applied in vitro, ex vivo, or in vivo in various stable isotope tracer-based metabolic studies, to gain systematic and novel metabolic insights in different biological systems, including human subjects. The pathway and network knowledge generated from NMR- and MS-based tracing of isotopically enriched substrates will be invaluable for directing functional analysis of other 'omics data to achieve understanding of regulation of biochemical systems, as demonstrated in a case study. Future developments in NMR technologies and reagents to enhance both detection sensitivity and resolution should further empower NMR in systems biochemical research.
Collapse
Affiliation(s)
- Teresa W-M Fan
- Department of Toxicology and Cancer Biology, University of Kentucky, 789 S. Limestone St., Lexington, KY 40536, United States.
| | - Andrew N Lane
- Department of Toxicology and Cancer Biology, University of Kentucky, 789 S. Limestone St., Lexington, KY 40536, United States.
| |
Collapse
|
32
|
Abstract
Gluconeogenesis is a complex metabolic process that involves multiple enzymatic steps regulated by myriad factors, including substrate concentrations, the redox state, activation and inhibition of specific enzyme steps, and hormonal modulation. At present, the most widely accepted technique to determine gluconeogenesis is by measuring the incorporation of deuterium from the body water pool into newly formed glucose. However, several techniques using radioactive and stable-labeled isotopes have been used to quantitate the contribution and regulation of gluconeogenesis in humans. Each method has its advantages, methodological assumptions, and set of propagated errors. In this review, we examine the strengths and weaknesses of the most commonly used stable isotopes methods to measure gluconeogenesis in vivo. We discuss the advantages and limitations of each method and summarize the applicability of these measurements in understanding normal and pathophysiological conditions.
Collapse
Affiliation(s)
- Stephanie T Chung
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - Shaji K Chacko
- U.S. Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX
| | - Agneta L Sunehag
- U.S. Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX
| | - Morey W Haymond
- U.S. Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX
| |
Collapse
|
33
|
Satapati S, Kucejova B, Duarte JAG, Fletcher JA, Reynolds L, Sunny NE, He T, Nair LA, Livingston KA, Fu X, Merritt ME, Sherry AD, Malloy CR, Shelton JM, Lambert J, Parks EJ, Corbin I, Magnuson MA, Browning JD, Burgess SC. Mitochondrial metabolism mediates oxidative stress and inflammation in fatty liver. J Clin Invest 2015; 125:4447-62. [PMID: 26571396 DOI: 10.1172/jci82204] [Citation(s) in RCA: 283] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 10/08/2015] [Indexed: 02/06/2023] Open
Abstract
Mitochondria are critical for respiration in all tissues; however, in liver, these organelles also accommodate high-capacity anaplerotic/cataplerotic pathways that are essential to gluconeogenesis and other biosynthetic activities. During nonalcoholic fatty liver disease (NAFLD), mitochondria also produce ROS that damage hepatocytes, trigger inflammation, and contribute to insulin resistance. Here, we provide several lines of evidence indicating that induction of biosynthesis through hepatic anaplerotic/cataplerotic pathways is energetically backed by elevated oxidative metabolism and hence contributes to oxidative stress and inflammation during NAFLD. First, in murine livers, elevation of fatty acid delivery not only induced oxidative metabolism, but also amplified anaplerosis/cataplerosis and caused a proportional rise in oxidative stress and inflammation. Second, loss of anaplerosis/cataplerosis via genetic knockdown of phosphoenolpyruvate carboxykinase 1 (Pck1) prevented fatty acid-induced rise in oxidative flux, oxidative stress, and inflammation. Flux appeared to be regulated by redox state, energy charge, and metabolite concentration, which may also amplify antioxidant pathways. Third, preventing elevated oxidative metabolism with metformin also normalized hepatic anaplerosis/cataplerosis and reduced markers of inflammation. Finally, independent histological grades in human NAFLD biopsies were proportional to oxidative flux. Thus, hepatic oxidative stress and inflammation are associated with elevated oxidative metabolism during an obesogenic diet, and this link may be provoked by increased work through anabolic pathways.
Collapse
|
34
|
Jin ES, Sherry AD, Malloy CR. Lactate Contributes to Glyceroneogenesis and Glyconeogenesis in Skeletal Muscle by Reversal of Pyruvate Kinase. J Biol Chem 2015; 290:30486-97. [PMID: 26491014 DOI: 10.1074/jbc.m115.689174] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Indexed: 11/06/2022] Open
Abstract
Phosphoenolpyruvate (PEP) generated from pyruvate is required for de novo synthesis of glycerol and glycogen in skeletal muscle. One possible pathway involves synthesis of PEP from the citric acid cycle intermediates via PEP carboxykinase, whereas another could involve reversal of pyruvate kinase (PK). Earlier studies have reported that reverse flux through PK can contribute carbon precursors for glycogen synthesis in muscle, but the physiological importance of this pathway remains uncertain especially in the setting of high plasma glucose. In addition, although PEP is a common intermediate for both glyconeogenesis and glyceroneogenesis, the importance of reverse PK in de novo glycerol synthesis has not been examined. Here we studied the contribution of reverse PK to synthesis of glycogen and the glycerol moiety of acylglycerols in skeletal muscle of animals with high plasma glucose. Rats received a single intraperitoneal bolus of glucose, glycerol, and lactate under a fed or fasted state. Only one of the three substrates was (13)C-labeled in each experiment. After 3 h of normal awake activity, the animals were sacrificed, and the contribution from each substrate to glycogen and the glycerol moiety of acylglycerols was evaluated. The fraction of (13)C labeling in glycogen and the glycerol moiety exceeded the possible contribution from either plasma glucose or muscle oxaloacetate. The reverse PK served as a common route for both glyconeogenesis and glyceroneogenesis in the skeletal muscle of rats with high plasma glucose. The activity of pyruvate carboxylase was low in muscle, and no PEP carboxykinase activity was detected.
Collapse
Affiliation(s)
- Eunsook S Jin
- From the Advanced Imaging Research Center, Department of Internal Medicine, and
| | - A Dean Sherry
- From the Advanced Imaging Research Center, Radiology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, Department of Chemistry, University of Texas at Dallas, Richardson, Texas 75080, and
| | - Craig R Malloy
- From the Advanced Imaging Research Center, Department of Internal Medicine, and Radiology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, Veterans Affairs North Texas Health Care System, Dallas, Texas 75216
| |
Collapse
|
35
|
Previs SF, Kelley DE. Tracer-based assessments of hepatic anaplerotic and TCA cycle flux: practicality, stoichiometry, and hidden assumptions. Am J Physiol Endocrinol Metab 2015; 309:E727-35. [PMID: 26330343 DOI: 10.1152/ajpendo.00216.2015] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 08/24/2015] [Indexed: 11/22/2022]
Abstract
Two groups recently used different tracer methods to quantify liver-specific flux rates. The studies had a similar goal, i.e., to characterize mitochondrial oxidative function. These efforts could have a direct impact on our ability to understand metabolic abnormalities that affect the pathophysiology of fatty liver and allow us to examine mechanisms surrounding potential therapeutic interventions. Briefly, one method couples the continuous infusion of [(13)C]acetate with direct real-time measurements of [(13)C]glutamate labeling in liver; the other method administers [(13)C]propionate, in combination with other tracers, and subsequently measures the (13)C labeling of plasma glucose and/or acetaminophen-glucuronide. It appears that a controversy has arisen, since the respective methods yielded different estimates of the anaplerotic/TCA flux ratio (VANA:VTCA) in "control" subjects, i.e., the [(13)C]acetate- and [(13)C]propionate-derived VANA:VTCA flux ratios appear to be ∼1.4 and ∼5, respectively. While the deep expertise in the respective groups makes it somewhat trivial for each to perform the tracer studies, the data interpretation is inherently difficult. The current perspective was undertaken to examine potential factors that could account for or contribute to the apparent differences. Attention was directed toward 1) matters of practicality, 2) issues surrounding stoichiometry, and 3) hidden assumptions. We believe that the [(13)C]acetate method has certain weaknesses that limit its utility; in contrast, the [(13)C]propionate method likely yields a more correct answer. We hope our discussion will help clarify the differences in the recent reports. Presumably this will be of interest to investigators who are considering tracer-based studies of liver metabolism.
Collapse
|
36
|
Previs SF, Herath K, Castro-Perez J, Mahsut A, Zhou H, McLaren DG, Shah V, Rohm RJ, Stout SJ, Zhong W, Wang SP, Johns DG, Hubbard BK, Cleary MA, Roddy TP. Effect of Error Propagation in Stable Isotope Tracer Studies: An Approach for Estimating Impact on Apparent Biochemical Flux. Methods Enzymol 2015; 561:331-58. [PMID: 26358910 DOI: 10.1016/bs.mie.2015.06.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Stable isotope tracers are widely used to quantify metabolic rates, and yet a limited number of studies have considered the impact of analytical error on estimates of flux. For example, when estimating the contribution of de novo lipogenesis, one typically measures a minimum of four isotope ratios, i.e., the precursor and product labeling pre- and posttracer administration. This seemingly simple problem has 1 correct solution and 80 erroneous outcomes. In this report, we outline a methodology for evaluating the effect of error propagation on apparent physiological endpoints. We demonstrate examples of how to evaluate the influence of analytical error in case studies concerning lipid and protein synthesis; we have focused on (2)H2O as a tracer and contrast different mass spectrometry platforms including GC-quadrupole-MS, GC-pyrolysis-IRMS, LC-quadrupole-MS, and high-resolution FT-ICR-MS. The method outlined herein can be used to determine how to minimize variations in the apparent biology by altering the dose and/or the type of tracer. Likewise, one can facilitate biological studies by estimating the reduction in the noise of an outcome that is expected for a given increase in the number of replicate injections.
Collapse
Affiliation(s)
| | | | | | - Ablatt Mahsut
- Merck Research Laboratories, Kenilworth, New Jersey, USA
| | - Haihong Zhou
- Merck Research Laboratories, Kenilworth, New Jersey, USA
| | | | - Vinit Shah
- Merck Research Laboratories, Kenilworth, New Jersey, USA
| | - Rory J Rohm
- Merck Research Laboratories, Kenilworth, New Jersey, USA
| | - Steven J Stout
- Merck Research Laboratories, Kenilworth, New Jersey, USA
| | - Wendy Zhong
- Merck Research Laboratories, Kenilworth, New Jersey, USA
| | | | | | | | | | - Thomas P Roddy
- Merck Research Laboratories, Kenilworth, New Jersey, USA
| |
Collapse
|
37
|
Sunny NE, Kalavalapalli S, Bril F, Garrett TJ, Nautiyal M, Mathew JT, Williams CM, Cusi K. Cross-talk between branched-chain amino acids and hepatic mitochondria is compromised in nonalcoholic fatty liver disease. Am J Physiol Endocrinol Metab 2015; 309:E311-9. [PMID: 26058864 PMCID: PMC4537921 DOI: 10.1152/ajpendo.00161.2015] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 06/04/2015] [Indexed: 12/20/2022]
Abstract
Elevated plasma branched-chain amino acids (BCAA) in the setting of insulin resistance have been relevant in predicting type 2 diabetes mellitus (T2DM) onset, but their role in the etiology of hepatic insulin resistance remains uncertain. We determined the link between BCAA and dysfunctional hepatic tricarboxylic acid (TCA) cycle, which is a central feature of hepatic insulin resistance and nonalcoholic fatty liver disease (NAFLD). Plasma metabolites under basal fasting and euglycemic hyperinsulinemic clamps (insulin stimulation) were measured in 94 human subjects with varying degrees of insulin sensitivity to identify their relationships with insulin resistance. Furthermore, the impact of elevated BCAA on hepatic TCA cycle was determined in a diet-induced mouse model of NAFLD, utilizing targeted metabolomics and nuclear magnetic resonance (NMR)-based metabolic flux analysis. Insulin stimulation revealed robust relationships between human plasma BCAA and indices of insulin resistance, indicating chronic metabolic overload from BCAA. Human plasma BCAA and long-chain acylcarnitines also showed a positive correlation, suggesting modulation of mitochondrial metabolism by BCAA. Concurrently, mice with NAFLD failed to optimally induce hepatic mTORC1, plasma ketones, and hepatic long-chain acylcarnitines, following acute elevation of plasma BCAA. Furthermore, elevated BCAA failed to induce multiple fluxes through hepatic TCA cycle in mice with NAFLD. Our data suggest that BCAA are essential to mediate efficient channeling of carbon substrates for oxidation through mitochondrial TCA cycle. Impairment of BCAA-mediated upregulation of the TCA cycle could be a significant contributor to mitochondrial dysfunction in NAFLD.
Collapse
Affiliation(s)
- Nishanth E Sunny
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, and
| | | | - Fernando Bril
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, and
| | - Timothy J Garrett
- Department of Pathology, University of Florida, Gainesville, Florida
| | - Manisha Nautiyal
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, and
| | - Justin T Mathew
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, and
| | - Caroline M Williams
- Department of Integrative Biology, University of California, Berkeley, California
| | - Kenneth Cusi
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, and Division of Endocrinology, Diabetes, and Metabolism, Malcom Randall Veterans Administration Medical Center (VAMC), Gainesville, Florida; Division of Diabetes, University of Texas Health Science Center at San Antonio, and Division of Diabetes, Audie L. Murphy VAMC, San Antonio, Texas; and
| |
Collapse
|
38
|
Efficient Modeling of MS/MS Data for Metabolic Flux Analysis. PLoS One 2015; 10:e0130213. [PMID: 26230524 PMCID: PMC4521746 DOI: 10.1371/journal.pone.0130213] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 05/16/2015] [Indexed: 01/25/2023] Open
Abstract
Metabolic flux analysis (MFA) is a widely used method for quantifying intracellular metabolic fluxes. It works by feeding cells with isotopic labeled nutrients, measuring metabolite isotopic labeling, and computationally interpreting the measured labeling data to estimate flux. Tandem mass-spectrometry (MS/MS) has been shown to be useful for MFA, providing positional isotopic labeling data. Specifically, MS/MS enables the measurement of a metabolite tandem mass-isotopomer distribution, representing the abundance in which certain parent and product fragments of a metabolite have different number of labeled atoms. However, a major limitation in using MFA with MS/MS data is the lack of a computationally efficient method for simulating such isotopic labeling data. Here, we describe the tandemer approach for efficiently computing metabolite tandem mass-isotopomer distributions in a metabolic network, given an estimation of metabolic fluxes. This approach can be used by MFA to find optimal metabolic fluxes, whose induced metabolite labeling patterns match tandem mass-isotopomer distributions measured by MS/MS. The tandemer approach is applied to simulate MS/MS data in a small-scale metabolic network model of mammalian methionine metabolism and in a large-scale metabolic network model of E. coli. It is shown to significantly improve the running time by between two to three orders of magnitude compared to the state-of-the-art, cumomers approach. We expect the tandemer approach to promote broader usage of MS/MS technology in metabolic flux analysis. Implementation is freely available at www.cs.technion.ac.il/~tomersh/methods.html
Collapse
|
39
|
Hasenour CM, Wall ML, Ridley DE, Hughey CC, James FD, Wasserman DH, Young JD. Mass spectrometry-based microassay of (2)H and (13)C plasma glucose labeling to quantify liver metabolic fluxes in vivo. Am J Physiol Endocrinol Metab 2015; 309:E191-203. [PMID: 25991647 PMCID: PMC4504936 DOI: 10.1152/ajpendo.00003.2015] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 05/14/2015] [Indexed: 01/06/2023]
Abstract
Mouse models designed to examine hepatic metabolism are critical to diabetes and obesity research. Thus, a microscale method to quantitatively assess hepatic glucose and intermediary metabolism in conscious, unrestrained mice was developed. [(13)C3]propionate, [(2)H2]water, and [6,6-(2)H2]glucose isotopes were delivered intravenously in short- (9 h) and long-term-fasted (19 h) C57BL/6J mice. GC-MS and mass isotopomer distribution (MID) analysis were performed on three 40-μl arterial plasma glucose samples obtained during the euglycemic isotopic steady state. Model-based regression of hepatic glucose and citric acid cycle (CAC)-related fluxes was performed using a comprehensive isotopomer model to track carbon and hydrogen atom transitions through the network and thereby simulate the MIDs of measured fragment ions. Glucose-6-phosphate production from glycogen diminished, and endogenous glucose production was exclusively gluconeogenic with prolonged fasting. Gluconeogenic flux from phosphoenolpyruvate (PEP) remained stable, whereas that from glycerol modestly increased from short- to long-term fasting. CAC flux [i.e., citrate synthase (VCS)] was reduced with long-term fasting. Interestingly, anaplerosis and cataplerosis increased with fast duration; accordingly, pyruvate carboxylation and the conversion of oxaloacetate to PEP were severalfold higher than VCS in long-term fasted mice. This method utilizes state-of-the-art in vivo methodology and comprehensive isotopomer modeling to quantify hepatic glucose and intermediary fluxes during physiological stress in mice. The small plasma requirements permit serial sampling without stress and the affirmation of steady-state glucose kinetics. Furthermore, the approach can accommodate a broad range of modeling assumptions, isotope tracers, and measurement inputs without the need to introduce ad hoc mathematical approximations.
Collapse
Affiliation(s)
| | - Martha L Wall
- Department of Chemical and Biomolecular Engineering, and
| | | | | | - Freyja D James
- Mouse Metabolic Phenotyping Center, Vanderbilt University, Nashville, Tennessee
| | - David H Wasserman
- Department of Molecular Physiology and Biophysics, Mouse Metabolic Phenotyping Center, Vanderbilt University, Nashville, Tennessee
| | - Jamey D Young
- Department of Molecular Physiology and Biophysics, Department of Chemical and Biomolecular Engineering, and
| |
Collapse
|
40
|
Wang LF, Yang GQ, Yang S, Yang GY, Li M, Zhu HS, Wang YY, Han LQ, Liu RY, Jia SD, Song F. Alteration of factors associated with hepatic gluconeogenesis in response to acute lipopolysaccharide in dairy goat1. J Anim Sci 2015; 93:2767-77. [DOI: 10.2527/jas.2014-8718] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
41
|
Coelho M, Valente-Silva P, Tylki-Szymanska A, Henriques T, Barosa C, Carvalho F, Jones JG. Demonstration of glucose-6-phosphate hydrogen 5 enrichment from deuterated water by transaldolase-mediated exchange alone. Magn Reson Med 2015; 75:1781-6. [PMID: 25995077 DOI: 10.1002/mrm.25749] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Revised: 04/02/2015] [Accepted: 04/02/2015] [Indexed: 11/11/2022]
Abstract
PURPOSE Enrichment of glucose position 5 (H5) from deuterated water ((2)H2O) is widely used for quantifying gluconeogenesis. Exchanges of hexose and triose phosphates mediated by transaldolase have been postulated to enrich H5 independently of gluconeogenesis, but to date this mechanism has not been proven. We determined the enrichment of glucose-6-phosphate (G6P), the immediate precursor of endogenously produced glucose, from (2)H2O in erythrocyte hemolysate preparations. Here, transaldolase exchange is active but gluconeogenesis is absent. METHODS Hemolysates were prepared from human erythrocytes and incubated with a buffer containing 5% [U-(13)C]G6P, unlabeled fructose 1,6-bisphosphate, and 10% (2)H2O. G6P (2)H-enrichment and (13)C-isotopomer distributions were analyzed by (2)H and (13)C NMR following derivatization to monoacetone glucose. RESULTS (2)H NMR analysis revealed high (2)H-enrichment of G6P hydrogens 2, 4, and 5; low enrichment of hydrogen 3, and residual enrichments of hydrogens 1, 6R, and 6S. (13)C NMR isotopomer analysis revealed that [U-(13)C]G6P was converted to [1,2,3-(13)C3]G6P, a predicted product of transaldolase-mediated exchange, as well as [1,2-(13)C2]G6P and [3-(13)C]G6P, predicted products of combined transaldolase and transketolase exchanges. CONCLUSION Hydrogen 5 of G6P was enriched from (2)H2O through exchanges mediated by transaldolase. These studies prove that G6P can be enriched in hydrogen 5 by (2)H2O independently of gluconeogenesis.
Collapse
Affiliation(s)
- Margarida Coelho
- Center for Neurosciences and Cell Biology, University of Coimbra, Portugal
| | | | - Anna Tylki-Szymanska
- Department of Pediatrics, Nutrition and Metabolic Disease, The Children's Memorial Health Institute, Warsaw, Poland
| | - Tiago Henriques
- Center for Neurosciences and Cell Biology, University of Coimbra, Portugal
| | - Cristina Barosa
- Center for Neurosciences and Cell Biology, University of Coimbra, Portugal
| | - Filipa Carvalho
- Center for Neurosciences and Cell Biology, University of Coimbra, Portugal
| | - John G Jones
- Center for Neurosciences and Cell Biology, University of Coimbra, Portugal.,APDP-Portuguese Diabetes Association, Lisbon, Portugal
| |
Collapse
|
42
|
Allen DK, Bates PD, Tjellström H. Tracking the metabolic pulse of plant lipid production with isotopic labeling and flux analyses: Past, present and future. Prog Lipid Res 2015; 58:97-120. [PMID: 25773881 DOI: 10.1016/j.plipres.2015.02.002] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 01/30/2015] [Accepted: 02/11/2015] [Indexed: 11/25/2022]
Abstract
Metabolism is comprised of networks of chemical transformations, organized into integrated biochemical pathways that are the basis of cellular operation, and function to sustain life. Metabolism, and thus life, is not static. The rate of metabolites transitioning through biochemical pathways (i.e., flux) determines cellular phenotypes, and is constantly changing in response to genetic or environmental perturbations. Each change evokes a response in metabolic pathway flow, and the quantification of fluxes under varied conditions helps to elucidate major and minor routes, and regulatory aspects of metabolism. To measure fluxes requires experimental methods that assess the movements and transformations of metabolites without creating artifacts. Isotopic labeling fills this role and is a long-standing experimental approach to identify pathways and quantify their metabolic relevance in different tissues or under different conditions. The application of labeling techniques to plant science is however far from reaching it potential. In light of advances in genetics and molecular biology that provide a means to alter metabolism, and given recent improvements in instrumentation, computational tools and available isotopes, the use of isotopic labeling to probe metabolism is becoming more and more powerful. We review the principal analytical methods for isotopic labeling with a focus on seminal studies of pathways and fluxes in lipid metabolism and carbon partitioning through central metabolism. Central carbon metabolic steps are directly linked to lipid production by serving to generate the precursors for fatty acid biosynthesis and lipid assembly. Additionally some of the ideas for labeling techniques that may be most applicable for lipid metabolism in the future were originally developed to investigate other aspects of central metabolism. We conclude by describing recent advances that will play an important future role in quantifying flux and metabolic operation in plant tissues.
Collapse
Affiliation(s)
- Doug K Allen
- United States Department of Agriculture, Agricultural Research Service, 975 North Warson Road, St. Louis, MO 63132, United States; Donald Danforth Plant Science Center, 975 North Warson Road, St. Louis, MO 63132, United States.
| | - Philip D Bates
- Department of Chemistry and Biochemistry, University of Southern Mississippi, Hattiesburg, MS 39406, United States
| | - Henrik Tjellström
- Department of Plant Biology, Michigan State University, East Lansing, MI 48824, United States; Great Lakes Bioenergy Research Center, Michigan State University, East Lansing, MI 48824, United States
| |
Collapse
|
43
|
Coelho M, Nunes P, Mendes VM, Manadas B, Heerschap A, Jones JG. Effect of Global ATGL Knockout on Murine Fasting Glucose Kinetics. J Diabetes Res 2015; 2015:542029. [PMID: 26236747 PMCID: PMC4506825 DOI: 10.1155/2015/542029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 06/08/2015] [Indexed: 12/25/2022] Open
Abstract
Mice deficient in adipose triglyceride lipase (ATGL(-/-)) present elevated ectopic lipid levels but are paradoxically glucose-tolerant. Measurement of endogenous glucose production (EGP) and Cori cycle activity provide insights into the maintenance of glycemic control in these animals. These parameters were determined in 7 wild-type (ATGL(+/-)) and 6 ATGL(-/-) mice by a primed-infusion of [U-(13)C6]glucose followed by LC-MS/MS targeted mass-isotopomer analysis of blood glucose. EGP was quantified by isotope dilution of [U-(13)C6]glucose while Cori cycling was estimated by analysis of glucose triose (13)C-isotopomers. Fasting plasma free fatty-acids were significantly lower in ATGL(-/-) versus control mice (0.43 ± 0.05 mM versus 0.73 ± 0.11 mM, P < 0.05). Six-hour fasting EGP rates were identical for both ATGL(-/-) and control mice (79 ± 11 versus 71 ± 7 μmol/kg/min, resp.). Peripheral glucose metabolism was dominated by Cori cycling (80 ± 2% and 82 ± 7% of glucose disposal for ATGL(-/-) and control mice, resp.) indicating that peripheral glucose oxidation was not significantly upregulated in ATGL(-/-) mice under these conditions. The glucose (13)C-isotopomer distributions in both ATGL(-/-) and control mice were consistent with extensive hepatic pyruvate recycling. This suggests that gluconeogenic outflow from the Krebs cycle was also well compensated in ATGL(-/-) mice.
Collapse
Affiliation(s)
- Margarida Coelho
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | | | - Vera M. Mendes
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Bruno Manadas
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Arend Heerschap
- Department of Radiology, Radboud University Nijmegen Medical Centre, Nijmegen, Netherlands
| | - John G. Jones
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Portuguese Diabetes Association (APDP), Lisbon, Portugal
- *John G. Jones:
| |
Collapse
|
44
|
Jin ES, Szuszkiewicz-Garcia M, Browning JD, Baxter JD, Abate N, Malloy CR. Influence of liver triglycerides on suppression of glucose production by insulin in men. J Clin Endocrinol Metab 2015; 100:235-43. [PMID: 25250633 PMCID: PMC4283006 DOI: 10.1210/jc.2014-2404] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
CONTEXT The ability of insulin to suppress hepatic glucose production is impaired among subjects with increased intrahepatic triglycerides (IHTG). However, little is known about the roles of insulin on the supporting fluxes of glucose production among patients with fatty liver. OBJECTIVE To evaluate the effects of insulin on fluxes through the three potential sources of plasma glucose (glycerol, the citric acid cycle, and glycogen) among patients with fatty liver. Design, Settings, Participants, and Intervention: Nineteen men with a range of IHTG (∼0.5% to 23%) were studied after an overnight fast and during hyperinsulinemia using magnetic resonance spectroscopy and stable isotope tracers. MAIN OUTCOME MEASURES IHTG, gluconeogenesis from glycerol, gluconeogenesis from the citric acid cycle, glycogenolysis, and (13)C-labeled glucose produced from the citric acid cycle during hyperinsulinemia were measured. RESULTS Men with high IHTG had higher fluxes through all pathways contributing to glucose production during hyperinsulinemia, compared to men with low IHTG, but they had similar fluxes after the fast. Consequently, men with fatty liver had impaired insulin efficiency in suppressing total glucose production as well as fluxes through all three biochemical pathways contributing to glucose. The detection of glucose isotopomers with (13)C arising from [U-(13)C3]propionate ingested during hyperinsulinemia demonstrated continuous gluconeogenesis from the citric acid cycle in all subjects. CONCLUSIONS These findings challenge the concept that individual glucose production pathways are selectively dysregulated during hepatic insulin resistance. Overproduction of glucose during hyperinsulinemia in men with fatty liver results from inadequate suppression of all the supporting fluxes of glucose production in response to insulin.
Collapse
Affiliation(s)
- Eunsook S Jin
- Advanced Imaging Research Center (E.S.J., J.D.Br., J.D.Ba., C.R.M.), University of Texas Southwestern Medical Center, Dallas, Texas 75390-8568; Department of Internal Medicine (E.S.J., M.S.-G., J.D.B., C.R.M.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; Department of Medicine (N.A.), Division of Endocrinology, University of Texas Medical Branch at Galveston, Texas 77555; Department of Radiology (C.R.M.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; and VA North Texas Health Care System (C.R.M.), Dallas, Texas 75216
| | | | | | | | | | | |
Collapse
|
45
|
Moreno KX, Satapati S, DeBerardinis RJ, Burgess SC, Malloy CR, Merritt ME. Real-time detection of hepatic gluconeogenic and glycogenolytic states using hyperpolarized [2-13C]dihydroxyacetone. J Biol Chem 2014; 289:35859-67. [PMID: 25352600 DOI: 10.1074/jbc.m114.613265] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Glycogenolysis and gluconeogenesis are sensitive to nutritional state, and the net direction of flux is controlled by multiple enzymatic steps. This delicate balance in the liver is disrupted by a variety of pathological states including cancer and diabetes mellitus. Hyperpolarized carbon-13 magnetic resonance is a new metabolic imaging technique that can probe intermediary metabolism nondestructively. There are currently no methods to rapidly distinguish livers in a gluconeogenic from glycogenolytic state. Here we use the gluconeogenic precursor dihydroxyacetone (DHA) to deliver hyperpolarized carbon-13 to the perfused mouse liver. DHA enters gluconeogenesis at the level of the trioses. Perfusion conditions were designed to establish either a gluconeogenic or a glycogenolytic state. Unexpectedly, we found that [2-(13)C]DHA was metabolized within a few seconds to the common intermediates and end products of both glycolysis and gluconeogenesis under both conditions, including [2,5-(13)C]glucose, [2-(13)C]glycerol 3-phosphate, [2-(13)C]phosphoenolpyruvate (PEP), [2-(13)C]pyruvate, [2-(13)C]alanine, and [2-(13)C]lactate. [2-(13)C]Phosphoenolpyruvate, a key branch point in gluconeogenesis and glycolysis, was monitored in functioning tissue for the first time. Observation of [2-(13)C]PEP was not anticipated as the free energy difference between PEP and pyruvate is large. Pyruvate kinase is the only regulatory step of the common glycolytic-gluconeogenic pathway that appears to exert significant control over the kinetics of any metabolites of DHA. A ratio of glycolytic to gluconeogenic products distinguished the gluconeogenic from glycogenolytic state in these functioning livers.
Collapse
Affiliation(s)
| | | | | | - Shawn C Burgess
- From the Advanced Imaging Research Center, Departments of Pharmacology and
| | - Craig R Malloy
- From the Advanced Imaging Research Center, Radiology, University of Texas - Southwestern Medical Center, Dallas, Texas 75390 and the Veterans Affairs North Texas Healthcare System, Lancaster, Texas 75216
| | - Matthew E Merritt
- From the Advanced Imaging Research Center, Radiology, University of Texas - Southwestern Medical Center, Dallas, Texas 75390 and
| |
Collapse
|
46
|
Jin ES, Sherry AD, Malloy CR. Interaction between the pentose phosphate pathway and gluconeogenesis from glycerol in the liver. J Biol Chem 2014; 289:32593-603. [PMID: 25288790 DOI: 10.1074/jbc.m114.577692] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
After exposure to [U-(13)C3]glycerol, the liver produces primarily [1,2,3-(13)C3]- and [4,5,6-(13)C3]glucose in equal proportions through gluconeogenesis from the level of trioses. Other (13)C-labeling patterns occur as a consequence of alternative pathways for glucose production. The pentose phosphate pathway (PPP), metabolism in the citric acid cycle, incomplete equilibration by triose phosphate isomerase, or the transaldolase reaction all interact to produce complex (13)C-labeling patterns in exported glucose. Here, we investigated (13)C labeling in plasma glucose in rats given [U-(13)C3]glycerol under various nutritional conditions. Blood was drawn at multiple time points to extract glucose for NMR analysis. Because the transaldolase reaction and incomplete equilibrium by triose phosphate isomerase cannot break a (13)C-(13)C bond within the trioses contributing to glucose, the appearance of [1,2-(13)C2]-, [2,3-(13)C2]-, [5,6-(13)C2]-, and [4,5-(13)C2]glucose provides direct evidence for metabolism of glycerol in the citric acid cycle or the PPP but not an influence of either triose phosphate isomerase or the transaldolase reaction. In all animals, [1,2-(13)C2]glucose/[2,3-(13)C2]glucose was significantly greater than [5,6-(13)C2]glucose/[4,5-(13)C2]glucose, a relationship that can only arise from gluconeogenesis followed by passage of substrates through the PPP. In summary, the hepatic PPP in vivo can be detected by (13)C distribution in blood glucose after [U-(13)C3]glycerol administration.
Collapse
Affiliation(s)
- Eunsook S Jin
- From the Advanced Imaging Research Center and Departments of Internal Medicine and
| | - A Dean Sherry
- From the Advanced Imaging Research Center and Radiology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, the Department of Chemistry, University of Texas at Dallas, Richardson, Texas 75080, and
| | - Craig R Malloy
- From the Advanced Imaging Research Center and Departments of Internal Medicine and Radiology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, the Veterans Affairs North Texas Health Care System, Dallas, Texas 75216
| |
Collapse
|
47
|
Hughey CC, Wasserman DH, Lee-Young RS, Lantier L. Approach to assessing determinants of glucose homeostasis in the conscious mouse. Mamm Genome 2014; 25:522-38. [PMID: 25074441 DOI: 10.1007/s00335-014-9533-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 07/04/2014] [Indexed: 01/11/2023]
Abstract
Obesity and type 2 diabetes lessen the quality of life of those afflicted and place considerable burden on the healthcare system. Furthermore, the detrimental impact of these pathologies is expected to persist or even worsen. Diabetes is characterized by impaired insulin action and glucose homeostasis. This has led to a rapid increase in the number of mouse models of metabolic disease being used in the basic sciences to assist in facilitating a greater understanding of the metabolic dysregulation associated with obesity and diabetes, the identification of therapeutic targets, and the discovery of effective treatments. This review briefly describes the most frequently utilized models of metabolic disease. A presentation of standard methods and technologies on the horizon for assessing metabolic phenotypes in mice, with particular emphasis on glucose handling and energy balance, is provided. The article also addresses issues related to study design, selection and execution of metabolic tests of glucose metabolism, the presentation of data, and interpretation of results.
Collapse
Affiliation(s)
- Curtis C Hughey
- Department of Molecular Physiology and Biophysics, School of Medicine, Vanderbilt University, 823 Light Hall, 2215 Garland Ave, Nashville, TN, 37232, USA,
| | | | | | | |
Collapse
|
48
|
Herath KB, Zhong W, Yang J, Mahsut A, Rohm RJ, Shah V, Castro-Perez J, Zhou H, Attygalle AB, Kang L, Singh S, Johns DG, Cleary MA, Hubbard BK, Previs SF, Roddy TP. Determination of low levels of 2H-labeling using high-resolution mass spectrometry: application in studies of lipid flux and beyond. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2014; 28:239-244. [PMID: 24375874 DOI: 10.1002/rcm.6776] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Revised: 10/23/2013] [Accepted: 10/27/2013] [Indexed: 06/03/2023]
Abstract
RATIONALE The ability to measure low levels of (2)H-labeling is important in studies of metabolic flux, e.g. one can estimate lipid synthesis by administering (2)H2O and then measuring the incorporation of (2)H into fatty acids. Unfortunately, the analyses are complicated by the presence of more abundant naturally occurring stable isotopes, e.g. (13)C. Conventional approaches rely on coupling gas chromatographic separation of lipids with either quadrupole-mass spectrometry (q-MS) and/or pyrolysis-isotope ratio mass spectrometry (IRMS). The former is limited by high background labeling (primarily from (13)C) whereas the latter is not suitable for routine high-throughput analyses. METHODS We have contrasted the use of continuous flow-pyrolysis-IRMS against high-resolution mass spectrometry (i.e. Qq-FT-ICR MS) for measuring the (2)H-enrichment of fatty acids and peptides. RESULTS In contrast to IRMS, which requires ~30 min per analysis, it is possible to measure the (2)H-enrichment of palmitate via direct infusion high-resolution mass spectrometry (HRMS) in ~3 min per sample. In addition, Qq-FT-ICR MS enabled measurements of the (2)H-enrichment of peptides (which is not possible using IRMS). CONCLUSIONS High-resolution mass spectrometry can be used to measure low levels of (2)H-labeling so we expect that this approach will enhance studies of metabolic flux that rely on (2)H-labeled tracers, e.g. (2)H2O. However, since the high-resolution analyses require greater amounts of a given analyte one potential limitation centers on the overall sensitivity. Presumably, future advances can overcome this barrier.
Collapse
|
49
|
Shlomi T, Fan J, Tang B, Kruger WD, Rabinowitz JD. Quantitation of cellular metabolic fluxes of methionine. Anal Chem 2014; 86:1583-91. [PMID: 24397525 DOI: 10.1021/ac4032093] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
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.
Collapse
Affiliation(s)
- Tomer Shlomi
- Dept. of Computer Science, Technion - Israel Institute of Technology, Haifa 32000, Israel
| | | | | | | | | |
Collapse
|
50
|
Befroy DE, Perry RJ, Jain N, Dufour S, Cline GW, Trimmer JK, Brosnan J, Rothman DL, Petersen KF, Shulman GI. Direct assessment of hepatic mitochondrial oxidative and anaplerotic fluxes in humans using dynamic 13C magnetic resonance spectroscopy. Nat Med 2014; 20:98-102. [PMID: 24317120 PMCID: PMC3947269 DOI: 10.1038/nm.3415] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Accepted: 10/29/2013] [Indexed: 12/11/2022]
Abstract
Despite the central role of the liver in the regulation of glucose and lipid metabolism, there are currently no methods to directly assess hepatic oxidative metabolism in humans in vivo. By using a new (13)C-labeling strategy in combination with (13)C magnetic resonance spectroscopy, we show that rates of mitochondrial oxidation and anaplerosis in human liver can be directly determined noninvasively. Using this approach, we found the mean rates of hepatic tricarboxylic acid (TCA) cycle flux (VTCA) and anaplerotic flux (VANA) to be 0.43 ± 0.04 μmol g(-1) min(-1) and 0.60 ± 0.11 μmol g(-1) min(-1), respectively, in twelve healthy, lean individuals. We also found the VANA/VTCA ratio to be 1.39 ± 0.22, which is severalfold lower than recently published estimates using an indirect approach. This method will be useful for understanding the pathogenesis of nonalcoholic fatty liver disease and type 2 diabetes, as well as for assessing the effectiveness of new therapies targeting these pathways in humans.
Collapse
Affiliation(s)
- Douglas E Befroy
- 1] Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, Connecticut, USA. [2] Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA. [3]
| | - Rachel J Perry
- 1] Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA. [2] Department of Cellular & Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut, USA. [3]
| | - Nimit Jain
- Department of Biomedical Engineering, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Sylvie Dufour
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Gary W Cline
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | | | - Douglas L Rothman
- 1] Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, Connecticut, USA. [2] Department of Biomedical Engineering, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Kitt Falk Petersen
- 1] Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA. [2] Novo Nordisk Foundation Center for Basic Metabolic Research, Copenhagen, Denmark
| | - Gerald I Shulman
- 1] Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA. [2] Department of Cellular & Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut, USA. [3] Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA. [4] Novo Nordisk Foundation Center for Basic Metabolic Research, Copenhagen, Denmark
| |
Collapse
|