1
|
Shichkova P, Coggan JS, Markram H, Keller D. Brain Metabolism in Health and Neurodegeneration: The Interplay Among Neurons and Astrocytes. Cells 2024; 13:1714. [PMID: 39451233 PMCID: PMC11506225 DOI: 10.3390/cells13201714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 08/31/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024] Open
Abstract
The regulation of energy in the brain has garnered substantial attention in recent years due to its significant implications in various disorders and aging. The brain's energy metabolism is a dynamic and tightly regulated network that balances energy demand and supply by engaging complementary molecular pathways. The crosstalk among these pathways enables the system to switch its preferred fuel source based on substrate availability, activity levels, and cell state-related factors such as redox balance. Brain energy production relies on multi-cellular cooperation and is continuously supplied by fuel from the blood due to limited internal energy stores. Astrocytes, which interface with neurons and blood vessels, play a crucial role in coordinating the brain's metabolic activity, and their dysfunction can have detrimental effects on brain health. This review characterizes the major energy substrates (glucose, lactate, glycogen, ketones and lipids) in astrocyte metabolism and their role in brain health, focusing on recent developments in the field.
Collapse
Affiliation(s)
- Polina Shichkova
- Blue Brain Project, École Polytechnique Fédérale de Lausanne, 1202 Geneva, Switzerland
| | - Jay S. Coggan
- Blue Brain Project, École Polytechnique Fédérale de Lausanne, 1202 Geneva, Switzerland
| | - Henry Markram
- Blue Brain Project, École Polytechnique Fédérale de Lausanne, 1202 Geneva, Switzerland
- Laboratory of Neural Microcircuitry, Brain Mind Institute, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Daniel Keller
- Blue Brain Project, École Polytechnique Fédérale de Lausanne, 1202 Geneva, Switzerland
| |
Collapse
|
2
|
Mitchell JM, Chi Y, Zheng S, Thapa M, Wang E, Li S. Annotation of Metabolites in Stable Isotope Tracing Untargeted Metabolomics via Khipu-web. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2024. [PMID: 39348378 DOI: 10.1021/jasms.4c00175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/02/2024]
Abstract
Stable isotope tracing is a crucial technique for understanding the metabolic wiring of biological systems, determining metabolic flux through pathways of interest, and detecting novel metabolites and pathways. Despite the potential insights provided by this technique, its application remains limited to a small number of targeted molecules and pathways. Because previous software tools usually require chemical formulas to find relevant features, and the data are highly complex, especially in untargeted metabolomics and when the downstream reactions and metabolites are poorly characterized. We report here Khipu version 2 and its new user-friendly web application. New functions are added to enhance analyzing stable isotope tracing data including metrics that evaluate peak enrichment in labeled samples, scoring methods to facilitate robust detection of intensity patterns and integrated natural abundance correction. We demonstrate that this approach can be applied to untargeted metabolomics to systematically extract isotope-labeled compounds and annotate the unidentified metabolites.
Collapse
Affiliation(s)
- Joshua M Mitchell
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, Connecticut 06032, United States
| | - Yuanye Chi
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, Connecticut 06032, United States
| | - Shujian Zheng
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, Connecticut 06032, United States
| | - Maheshwor Thapa
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, Connecticut 06032, United States
| | - Eric Wang
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, Connecticut 06032, United States
| | - Shuzhao Li
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, Connecticut 06032, United States
- University of Connecticut School of Medicine, Farmington, Connecticut 06032, United States
| |
Collapse
|
3
|
Menegollo M, Bentham RB, Henriques T, Ng SQ, Ren Z, Esculier C, Agarwal S, Tong ETY, Lo C, Ilangovan S, Szabadkai Z, Suman M, Patani N, Ghanate A, Bryson K, Stein RC, Yuneva M, Szabadkai G. Multistate Gene Cluster Switches Determine the Adaptive Mitochondrial and Metabolic Landscape of Breast Cancer. Cancer Res 2024; 84:2911-2925. [PMID: 38924467 PMCID: PMC11372374 DOI: 10.1158/0008-5472.can-23-3172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 04/17/2024] [Accepted: 06/20/2024] [Indexed: 06/28/2024]
Abstract
Adaptive metabolic switches are proposed to underlie conversions between cellular states during normal development as well as in cancer evolution. Metabolic adaptations represent important therapeutic targets in tumors, highlighting the need to characterize the full spectrum, characteristics, and regulation of the metabolic switches. To investigate the hypothesis that metabolic switches associated with specific metabolic states can be recognized by locating large alternating gene expression patterns, we developed a method to identify interspersed gene sets by massive correlated biclustering and to predict their metabolic wiring. Testing the method on breast cancer transcriptome datasets revealed a series of gene sets with switch-like behavior that could be used to predict mitochondrial content, metabolic activity, and central carbon flux in tumors. The predictions were experimentally validated by bioenergetic profiling and metabolic flux analysis of 13C-labeled substrates. The metabolic switch positions also distinguished between cellular states, correlating with tumor pathology, prognosis, and chemosensitivity. The method is applicable to any large and heterogeneous transcriptome dataset to discover metabolic and associated pathophysiological states. Significance: A method for identifying the transcriptomic signatures of metabolic switches underlying divergent routes of cellular transformation stratifies breast cancer into metabolic subtypes, predicting their biology, architecture, and clinical outcome.
Collapse
Affiliation(s)
- Michela Menegollo
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Robert B Bentham
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, United Kingdom
| | - Tiago Henriques
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Seow Q Ng
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, United Kingdom
| | - Ziyu Ren
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, United Kingdom
| | - Clarinde Esculier
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, United Kingdom
| | - Sia Agarwal
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, United Kingdom
| | - Emily T Y Tong
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, United Kingdom
| | - Clement Lo
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, United Kingdom
| | - Sanjana Ilangovan
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, United Kingdom
| | - Zorka Szabadkai
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, United Kingdom
| | - Matteo Suman
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Neill Patani
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, United Kingdom
- The Francis Crick Institute, London, United Kingdom
| | | | - Kevin Bryson
- Department of Computer Sciences, University College London, London, United Kingdom
| | - Robert C Stein
- Department of Oncology, University College London Hospitals, London, United Kingdom
- UCL Cancer Institute, University College London, London, United Kingdom
| | | | - Gyorgy Szabadkai
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, United Kingdom
- The Francis Crick Institute, London, United Kingdom
| |
Collapse
|
4
|
Malik DM, Rhoades SD, Zhang SL, Sengupta A, Barber A, Haynes P, Arnadottir ES, Pack A, Kibbey RG, Kain P, Sehgal A, Weljie AM. Glucose Challenge Uncovers Temporal Fungibility of Metabolic Homeostasis over a day:night cycle. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.30.564837. [PMID: 37961230 PMCID: PMC10634956 DOI: 10.1101/2023.10.30.564837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Rhythmicity is a cornerstone of behavioral and biological processes, especially metabolism, yet the mechanisms behind metabolite cycling remain elusive. This study uncovers a robust oscillation in key metabolite pathways downstream of glucose in humans. A purpose-built 13C6-glucose isotope tracing platform was used to sample Drosophila every 4h and probe these pathways, revealing a striking peak in biosynthesis shortly after lights-on in wild-type flies. A hyperactive mutant (fumin) demonstrates increased Krebs cycle labelling and dawn-specific glycolysis labelling. Surprisingly, neither underlying feeding rhythms nor the presence of food availability explain the rhythmicity of glucose processing across genotypes, suggesting a robust internal mechanism for metabolic control of glucose processing. These results align with clinical data highlighting detrimental effects of mistimed energy intake. Our approach offers a unique insight into the dynamic range of daily metabolic processing and provides a mechanistic foundation for exploring circadian metabolic homeostasis in disease contexts.
Collapse
Affiliation(s)
- Dania M. Malik
- Pharmacology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Department of Systems Pharmacology and Translational Therapeutics
- Institute for Translational Medicine and Therapeutics
- These authors contributed equally
| | - Seth D. Rhoades
- Pharmacology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Department of Systems Pharmacology and Translational Therapeutics
- Institute for Translational Medicine and Therapeutics
- Fulgens Consulting, LLC, Cambridge, Massachusetts 02142, USA
- These authors contributed equally
| | - Shirley L. Zhang
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Howard Hughes Medical Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Arjun Sengupta
- Department of Systems Pharmacology and Translational Therapeutics
- Institute for Translational Medicine and Therapeutics
| | - Annika Barber
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Howard Hughes Medical Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers, The State University of New Jersey, New Brunswick, New Jersey 08854, USA
| | - Paula Haynes
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Howard Hughes Medical Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Erna Sif Arnadottir
- Division of Sleep Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Center for Sleep and Circadian Neurobiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Allan Pack
- Division of Sleep Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Center for Sleep and Circadian Neurobiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Richard G. Kibbey
- Department of Internal Medicine, Department of Cellular & Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | - Pinky Kain
- Department of Systems Pharmacology and Translational Therapeutics
- Institute for Translational Medicine and Therapeutics
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Amita Sehgal
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Howard Hughes Medical Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Aalim M. Weljie
- Department of Systems Pharmacology and Translational Therapeutics
- Institute for Translational Medicine and Therapeutics
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
5
|
Racine L, Parmentier R, Niphadkar S, Chhun J, Martignoles JA, Delhommeau F, Laxman S, Paldi A. Metabolic adaptation pilots the differentiation of human hematopoietic cells. Life Sci Alliance 2024; 7:e202402747. [PMID: 38802246 PMCID: PMC11130395 DOI: 10.26508/lsa.202402747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/12/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024] Open
Abstract
A continuous supply of energy is an essential prerequisite for survival and represents the highest priority for the cell. We hypothesize that cell differentiation is a process of optimization of energy flow in a changing environment through phenotypic adaptation. The mechanistic basis of this hypothesis is provided by the established link between core energy metabolism and epigenetic covalent modifications of chromatin. This theory predicts that early metabolic perturbations impact subsequent differentiation. To test this, we induced transient metabolic perturbations in undifferentiated human hematopoietic cells using pharmacological inhibitors targeting key metabolic reactions. We recorded changes in chromatin structure and gene expression, as well as phenotypic alterations by single-cell ATAC and RNA sequencing, time-lapse microscopy, and flow cytometry. Our observations suggest that these metabolic perturbations are shortly followed by alterations in chromatin structure, leading to changes in gene expression. We also show that these transient fluctuations alter the differentiation potential of the cells.
Collapse
Affiliation(s)
- Laëtitia Racine
- https://ror.org/02en5vm52 Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, Paris, France
- https://ror.org/046b3cj80 Ecole Pratique des Hautes Etudes, PSL Research University, Paris, France
- AP-HP, SIRIC CURAMUS, Hôpital Saint-Antoine, Service d'Hématologie Biologique, Paris, France
- OPALE Carnot Institute, Paris, France
| | - Romuald Parmentier
- https://ror.org/02en5vm52 Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, Paris, France
- https://ror.org/046b3cj80 Ecole Pratique des Hautes Etudes, PSL Research University, Paris, France
- AP-HP, SIRIC CURAMUS, Hôpital Saint-Antoine, Service d'Hématologie Biologique, Paris, France
- OPALE Carnot Institute, Paris, France
| | - Shreyas Niphadkar
- Institute for Stem Cell Science and Regenerative Medicine (DBT-inStem), Bangalore, India
| | - Julie Chhun
- https://ror.org/02en5vm52 Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, Paris, France
- https://ror.org/046b3cj80 Ecole Pratique des Hautes Etudes, PSL Research University, Paris, France
- AP-HP, SIRIC CURAMUS, Hôpital Saint-Antoine, Service d'Hématologie Biologique, Paris, France
- OPALE Carnot Institute, Paris, France
| | - Jean-Alain Martignoles
- https://ror.org/02en5vm52 Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, Paris, France
- AP-HP, SIRIC CURAMUS, Hôpital Saint-Antoine, Service d'Hématologie Biologique, Paris, France
- OPALE Carnot Institute, Paris, France
| | - François Delhommeau
- https://ror.org/02en5vm52 Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, Paris, France
- AP-HP, SIRIC CURAMUS, Hôpital Saint-Antoine, Service d'Hématologie Biologique, Paris, France
- OPALE Carnot Institute, Paris, France
| | - Sunil Laxman
- Institute for Stem Cell Science and Regenerative Medicine (DBT-inStem), Bangalore, India
| | - Andras Paldi
- https://ror.org/02en5vm52 Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, Paris, France
- https://ror.org/046b3cj80 Ecole Pratique des Hautes Etudes, PSL Research University, Paris, France
- AP-HP, SIRIC CURAMUS, Hôpital Saint-Antoine, Service d'Hématologie Biologique, Paris, France
- OPALE Carnot Institute, Paris, France
| |
Collapse
|
6
|
Mann CG, MacArthur MR, Zhang J, Gong S, AbuSalim JE, Hunter CJ, Lu W, Agius T, Longchamp A, Allagnat F, Rabinowitz J, Mitchell JR, De Bock K, Mitchell SJ. Sulfur Amino Acid Restriction Enhances Exercise Capacity in Mice by Boosting Fat Oxidation in Muscle. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.27.601041. [PMID: 39005372 PMCID: PMC11244859 DOI: 10.1101/2024.06.27.601041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Dietary restriction of the sulfur-containing amino acids methionine and cysteine (SAAR) improves body composition, enhances insulin sensitivity, and extends lifespan; benefits seen also with endurance exercise. Yet, the impact of SAAR on skeletal muscle remains largely unexplored. Here we demonstrate that one week of SAAR in sedentary, young, male mice increases endurance exercise capacity. Indirect calorimetry showed that SAAR increased lipid oxidation at rest and delayed the onset of carbohydrate utilization during exercise. Transcriptomic analysis revealed increased expression of genes involved in fatty acid catabolism especially in glycolytic muscle following SAAR. These findings were functionally supported by increased fatty acid circulatory turnover flux and muscle β-oxidation. Reducing lipid uptake from circulation through endothelial cell (EC)-specific CD36 deletion attenuated the running phenotype. Mechanistically, VEGF-signaling inhibition prevented exercise increases following SAAR, without affecting angiogenesis, implicating noncanonical VEGF signaling and EC CD36-dependent fatty acid transport in regulating exercise capacity by influencing muscle substrate availability.
Collapse
Affiliation(s)
- Charlotte G Mann
- Department of Health Sciences and Technology, ETH Zurich, Zurich 8092, Switzerland
| | - Michael R MacArthur
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
- Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
- Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ 08544, USA
| | - Jing Zhang
- Department of Health Sciences and Technology, ETH Zurich, Zurich 8092, Switzerland
| | - Songlin Gong
- Department of Health Sciences and Technology, ETH Zurich, Zurich 8092, Switzerland
| | - Jenna E AbuSalim
- Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
- Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ 08544, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Craig J. Hunter
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
- Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
- Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ 08544, USA
| | - Wenyun Lu
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
- Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Thomas Agius
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), Lausanne 1005, Switzerland
| | - Alban Longchamp
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), Lausanne 1005, Switzerland
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Florent Allagnat
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), Lausanne 1005, Switzerland
| | - Joshua Rabinowitz
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
- Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
- Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ 08544, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - James R Mitchell
- Department of Health Sciences and Technology, ETH Zurich, Zurich 8092, Switzerland
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Katrien De Bock
- Department of Health Sciences and Technology, ETH Zurich, Zurich 8092, Switzerland
| | - Sarah J Mitchell
- Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|
7
|
Bae H, Kim BR, Jung S, Le J, van der Heide D, Yu W, Park SH, Hilkin BM, Gansemer ND, Powers LS, Kang T, Meyerholz DK, Schuster VL, Jang C, Welsh MJ. Arteriovenous metabolomics in pigs reveals CFTR regulation of metabolism in multiple organs. J Clin Invest 2024; 134:e174500. [PMID: 38743489 PMCID: PMC11213515 DOI: 10.1172/jci174500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 05/07/2024] [Indexed: 05/16/2024] Open
Abstract
Mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene cause cystic fibrosis (CF), a multiorgan disease that is characterized by diverse metabolic defects. However, other than specific CFTR mutations, the factors that influence disease progression and severity remain poorly understood. Aberrant metabolite levels have been reported, but whether CFTR loss itself or secondary abnormalities (infection, inflammation, malnutrition, and various treatments) drive metabolic defects is uncertain. Here, we implemented comprehensive arteriovenous metabolomics in newborn CF pigs, and the results revealed CFTR as a bona fide regulator of metabolism. CFTR loss impaired metabolite exchange across organs, including disruption of lung uptake of fatty acids, yet enhancement of uptake of arachidonic acid, a precursor of proinflammatory cytokines. CFTR loss also impaired kidney reabsorption of amino acids and lactate and abolished renal glucose homeostasis. These and additional unexpected metabolic defects prior to disease manifestations reveal a fundamental role for CFTR in controlling multiorgan metabolism. Such discovery informs a basic understanding of CF, provides a foundation for future investigation, and has implications for developing therapies targeting only a single tissue.
Collapse
Affiliation(s)
- Hosung Bae
- Department of Biological Chemistry, University of California – Irvine, Irvine, California, USA
| | - Bo Ram Kim
- Department of Internal Medicine, Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
- Howard Hughes Medical Institute, University of Iowa, Iowa City, Iowa, USA
| | - Sunhee Jung
- Department of Biological Chemistry, University of California – Irvine, Irvine, California, USA
| | - Johnny Le
- Department of Biological Chemistry, University of California – Irvine, Irvine, California, USA
| | | | - Wenjie Yu
- Department of Internal Medicine, Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
- Howard Hughes Medical Institute, University of Iowa, Iowa City, Iowa, USA
| | - Sang Hee Park
- Department of Biological Chemistry, University of California – Irvine, Irvine, California, USA
| | - Brieanna M. Hilkin
- Department of Internal Medicine, Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Nicholas D. Gansemer
- Department of Internal Medicine, Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Linda S. Powers
- Department of Internal Medicine, Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Taekyung Kang
- Department of Biological Chemistry, University of California – Irvine, Irvine, California, USA
| | - David K. Meyerholz
- Department of Pathology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Victor L. Schuster
- Department of Internal Medicine, Albert Einstein College of Medicine, Bronx, New York, New York, USA
| | - Cholsoon Jang
- Department of Biological Chemistry, University of California – Irvine, Irvine, California, USA
- Center for Complex Biological Systems and
- Center for Epigenetics and Metabolism, University of California – Irvine, Irvine, California, USA
| | - Michael J. Welsh
- Department of Internal Medicine, Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
- Howard Hughes Medical Institute, University of Iowa, Iowa City, Iowa, USA
- Department of Molecular Physiology and Biophysics, Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
8
|
Verkerke ARP, Wang D, Yoshida N, Taxin ZH, Shi X, Zheng S, Li Y, Auger C, Oikawa S, Yook JS, Granath-Panelo M, He W, Zhang GF, Matsushita M, Saito M, Gerszten RE, Mills EL, Banks AS, Ishihama Y, White PJ, McGarrah RW, Yoneshiro T, Kajimura S. BCAA-nitrogen flux in brown fat controls metabolic health independent of thermogenesis. Cell 2024; 187:2359-2374.e18. [PMID: 38653240 PMCID: PMC11145561 DOI: 10.1016/j.cell.2024.03.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 01/07/2024] [Accepted: 03/21/2024] [Indexed: 04/25/2024]
Abstract
Brown adipose tissue (BAT) is best known for thermogenesis. Rodent studies demonstrated that enhanced BAT thermogenesis is tightly associated with increased energy expenditure, reduced body weight, and improved glucose homeostasis. However, human BAT is protective against type 2 diabetes, independent of body weight. The mechanism underlying this dissociation remains unclear. Here, we report that impaired mitochondrial catabolism of branched-chain amino acids (BCAAs) in BAT, by deleting mitochondrial BCAA carriers (MBCs), caused systemic insulin resistance without affecting energy expenditure and body weight. Brown adipocytes catabolized BCAA in the mitochondria as nitrogen donors for the biosynthesis of non-essential amino acids and glutathione. Impaired mitochondrial BCAA-nitrogen flux in BAT resulted in increased oxidative stress, decreased hepatic insulin signaling, and decreased circulating BCAA-derived metabolites. A high-fat diet attenuated BCAA-nitrogen flux and metabolite synthesis in BAT, whereas cold-activated BAT enhanced the synthesis. This work uncovers a metabolite-mediated pathway through which BAT controls metabolic health beyond thermogenesis.
Collapse
Affiliation(s)
- Anthony R P Verkerke
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, and Howard Hughes Medical Institute, Boston, MA, USA
| | - Dandan Wang
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, and Howard Hughes Medical Institute, Boston, MA, USA
| | - Naofumi Yoshida
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, and Howard Hughes Medical Institute, Boston, MA, USA
| | - Zachary H Taxin
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, and Howard Hughes Medical Institute, Boston, MA, USA
| | - Xu Shi
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Shuning Zheng
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Yuka Li
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Christopher Auger
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, and Howard Hughes Medical Institute, Boston, MA, USA
| | - Satoshi Oikawa
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, and Howard Hughes Medical Institute, Boston, MA, USA
| | - Jin-Seon Yook
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, and Howard Hughes Medical Institute, Boston, MA, USA
| | - Melia Granath-Panelo
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, and Howard Hughes Medical Institute, Boston, MA, USA
| | - Wentao He
- Duke Molecular Physiology Institute, Duke School of Medicine, Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, Duke University, Durham, NC, USA
| | - Guo-Fang Zhang
- Duke Molecular Physiology Institute, Duke School of Medicine, Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, Duke University, Durham, NC, USA
| | - Mami Matsushita
- Department of Nutrition, School of Nursing and Nutrition, Tenshi College, Sapporo, Japan
| | - Masayuki Saito
- Laboratory of Biochemistry, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Robert E Gerszten
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Evanna L Mills
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute and Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Alexander S Banks
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Yasushi Ishihama
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Phillip J White
- Duke Molecular Physiology Institute, Duke School of Medicine, Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Robert W McGarrah
- Duke Molecular Physiology Institute, Duke School of Medicine, Sarah W. Stedman Nutrition and Metabolism Center, Department of Medicine, Division of Cardiology, Duke University, Durham, NC, USA
| | - Takeshi Yoneshiro
- Division of Metabolic Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan; Division of Molecular Physiology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shingo Kajimura
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, and Howard Hughes Medical Institute, Boston, MA, USA.
| |
Collapse
|
9
|
Bowman CE, Neinast MD, Jang C, Patel J, Blair MC, Mirek ET, Jonsson WO, Chu Q, Merlo L, Mandik-Nayak L, Anthony TG, Rabinowitz JD, Arany Z. Off-target depletion of plasma tryptophan by allosteric inhibitors of BCKDK. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.05.582974. [PMID: 38496495 PMCID: PMC10942310 DOI: 10.1101/2024.03.05.582974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
The activation of branched chain amino acid (BCAA) catabolism has garnered interest as a potential therapeutic approach to improve insulin sensitivity, enhance recovery from heart failure, and blunt tumor growth. Evidence for this interest relies in part on BT2, a small molecule that promotes BCAA oxidation and is protective in mouse models of these pathologies. BT2 and other analogs allosterically inhibit branched chain ketoacid dehydrogenase kinase (BCKDK) to promote BCAA oxidation, which is presumed to underlie the salutary effects of BT2. Potential "off-target" effects of BT2 have not been considered, however. We therefore tested for metabolic off-target effects of BT2 in Bckdk-/- animals. As expected, BT2 failed to activate BCAA oxidation in these animals. Surprisingly, however, BT2 strongly reduced plasma tryptophan levels and promoted catabolism of tryptophan to kynurenine in both control and Bckdk-/- mice. Mechanistic studies revealed that none of the principal tryptophan catabolic or kynurenine-producing/consuming enzymes (TDO, IDO1, IDO2, or KATs) were required for BT2-mediated lowering of plasma tryptophan. Instead, using equilibrium dialysis assays and mice lacking albumin, we show that BT2 avidly binds plasma albumin and displaces tryptophan, releasing it for catabolism. These data confirm that BT2 activates BCAA oxidation via inhibition of BCKDK but also reveal a robust off-target effect on tryptophan metabolism via displacement from serum albumin. The data highlight a potential confounding effect for pharmaceutical compounds that compete for binding with albumin-bound tryptophan.
Collapse
Affiliation(s)
- Caitlyn E. Bowman
- Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
- Present address: Biology Department, Williams College, Williamstown, MA, USA
| | - Michael D. Neinast
- Department of Chemistry and Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Cholsoon Jang
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Jiten Patel
- Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Megan C. Blair
- Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Emily T. Mirek
- Department of Nutritional Sciences, Rutgers School of Environmental and Biological Sciences, New Brunswick, NJ, USA
| | - William O. Jonsson
- Department of Nutritional Sciences, Rutgers School of Environmental and Biological Sciences, New Brunswick, NJ, USA
| | - Qingwei Chu
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lauren Merlo
- Lankenau Institute for Medical Research, Wynnewood, PA, USA
| | | | - Tracy G. Anthony
- Department of Nutritional Sciences, Rutgers School of Environmental and Biological Sciences, New Brunswick, NJ, USA
| | - Joshua D. Rabinowitz
- Department of Chemistry and Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Zolt Arany
- Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
10
|
Membrez M, Migliavacca E, Christen S, Yaku K, Trieu J, Lee AK, Morandini F, Giner MP, Stiner J, Makarov MV, Garratt ES, Vasiloglou MF, Chanvillard L, Dalbram E, Ehrlich AM, Sanchez-Garcia JL, Canto C, Karagounis LG, Treebak JT, Migaud ME, Heshmat R, Razi F, Karnani N, Ostovar A, Farzadfar F, Tay SKH, Sanders MJ, Lillycrop KA, Godfrey KM, Nakagawa T, Moco S, Koopman R, Lynch GS, Sorrentino V, Feige JN. Trigonelline is an NAD + precursor that improves muscle function during ageing and is reduced in human sarcopenia. Nat Metab 2024; 6:433-447. [PMID: 38504132 DOI: 10.1038/s42255-024-00997-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 01/26/2024] [Indexed: 03/21/2024]
Abstract
Mitochondrial dysfunction and low nicotinamide adenine dinucleotide (NAD+) levels are hallmarks of skeletal muscle ageing and sarcopenia1-3, but it is unclear whether these defects result from local changes or can be mediated by systemic or dietary cues. Here we report a functional link between circulating levels of the natural alkaloid trigonelline, which is structurally related to nicotinic acid4, NAD+ levels and muscle health in multiple species. In humans, serum trigonelline levels are reduced with sarcopenia and correlate positively with muscle strength and mitochondrial oxidative phosphorylation in skeletal muscle. Using naturally occurring and isotopically labelled trigonelline, we demonstrate that trigonelline incorporates into the NAD+ pool and increases NAD+ levels in Caenorhabditis elegans, mice and primary myotubes from healthy individuals and individuals with sarcopenia. Mechanistically, trigonelline does not activate GPR109A but is metabolized via the nicotinate phosphoribosyltransferase/Preiss-Handler pathway5,6 across models. In C. elegans, trigonelline improves mitochondrial respiration and biogenesis, reduces age-related muscle wasting and increases lifespan and mobility through an NAD+-dependent mechanism requiring sirtuin. Dietary trigonelline supplementation in male mice enhances muscle strength and prevents fatigue during ageing. Collectively, we identify nutritional supplementation of trigonelline as an NAD+-boosting strategy with therapeutic potential for age-associated muscle decline.
Collapse
Affiliation(s)
- Mathieu Membrez
- Nestlé Institute of Health Sciences, Nestlé Research, Lausanne, Switzerland
| | | | - Stefan Christen
- Nestlé Institute of Food Safety and Analytical Sciences, Nestlé Research, Lausanne, Switzerland
| | - Keisuke Yaku
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Jennifer Trieu
- Centre for Muscle Research, Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Alaina K Lee
- Centre for Muscle Research, Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Francesco Morandini
- Nestlé Institute of Health Sciences, Nestlé Research, Lausanne, Switzerland
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Maria Pilar Giner
- Nestlé Institute of Food Safety and Analytical Sciences, Nestlé Research, Lausanne, Switzerland
| | - Jade Stiner
- Nestlé Institute of Health Sciences, Nestlé Research, Lausanne, Switzerland
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Mikhail V Makarov
- Mitchell Cancer Institute, Department of Pharmacology, F. P. Whiddon College of Medicine, University of South Alabama, Mobile, AL, USA
| | - Emma S Garratt
- Institute of Developmental Sciences, Human Developmental and Health, Faculty of Medicine, University of Southampton, Southampton, UK
- National Institute for Health and Care Research, Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Maria F Vasiloglou
- Nestlé Institute of Health Sciences, Nestlé Research, Lausanne, Switzerland
| | - Lucie Chanvillard
- Nestlé Institute of Health Sciences, Nestlé Research, Lausanne, Switzerland
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Emilie Dalbram
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Amy M Ehrlich
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Carles Canto
- Nestlé Institute of Health Sciences, Nestlé Research, Lausanne, Switzerland
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Leonidas G Karagounis
- Nestlé Health Science, Translation Research, Lausanne, Switzerland
- Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Victoria, Australia
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | - Jonas T Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Marie E Migaud
- Mitchell Cancer Institute, Department of Pharmacology, F. P. Whiddon College of Medicine, University of South Alabama, Mobile, AL, USA
| | - Ramin Heshmat
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Farideh Razi
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Science Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Neerja Karnani
- Singapore Institute for Clinical Sciences (A*STAR), Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Afshin Ostovar
- Osteoporosis Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Farshad Farzadfar
- Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Stacey K H Tay
- KTP-National University Children's Medical Institute, National University Hospital, Singapore, Singapore
| | - Matthew J Sanders
- Nestlé Institute of Health Sciences, Nestlé Research, Lausanne, Switzerland
| | - Karen A Lillycrop
- Institute of Developmental Sciences, Human Developmental and Health, Faculty of Medicine, University of Southampton, Southampton, UK
- National Institute for Health and Care Research, Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
| | - Keith M Godfrey
- Institute of Developmental Sciences, Human Developmental and Health, Faculty of Medicine, University of Southampton, Southampton, UK
- National Institute for Health and Care Research, Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
- Medical Research Council Lifecourse Epidemiology Centre, University of Southampton, Southampton, UK
| | - Takashi Nakagawa
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Sofia Moco
- Nestlé Institute of Food Safety and Analytical Sciences, Nestlé Research, Lausanne, Switzerland
- Division of Molecular and Computational Toxicology, Department of Chemistry and Pharmaceutical Sciences, Amsterdam Institute for Molecular and Life Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - René Koopman
- Centre for Muscle Research, Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Gordon S Lynch
- Centre for Muscle Research, Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Vincenzo Sorrentino
- Nestlé Institute of Health Sciences, Nestlé Research, Lausanne, Switzerland.
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Jerome N Feige
- Nestlé Institute of Health Sciences, Nestlé Research, Lausanne, Switzerland.
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.
| |
Collapse
|
11
|
McBride MJ, Hunter CJ, Zhang Z, TeSlaa T, Xu X, Ducker GS, Rabinowitz JD. Glycine homeostasis requires reverse SHMT flux. Cell Metab 2024; 36:103-115.e4. [PMID: 38171330 DOI: 10.1016/j.cmet.2023.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 10/09/2023] [Accepted: 12/04/2023] [Indexed: 01/05/2024]
Abstract
The folate-dependent enzyme serine hydroxymethyltransferase (SHMT) reversibly converts serine into glycine and a tetrahydrofolate-bound one-carbon unit. Such one-carbon unit production plays a critical role in development, the immune system, and cancer. Using rodent models, here we show that the whole-body SHMT flux acts to net consume rather than produce glycine. Pharmacological inhibition of whole-body SHMT1/2 and genetic knockout of liver SHMT2 elevated circulating glycine levels up to eight-fold. Stable-isotope tracing revealed that the liver converts glycine to serine, which is then converted by serine dehydratase into pyruvate and burned in the tricarboxylic acid cycle. In response to diets deficient in serine and glycine, de novo biosynthetic flux was unaltered, but SHMT2- and serine-dehydratase-mediated catabolic flux was lower. Thus, glucose-derived serine synthesis is largely insensitive to systemic demand. Instead, circulating serine and glycine homeostasis is maintained through variable consumption, with liver SHMT2 a major glycine-consuming enzyme.
Collapse
Affiliation(s)
- Matthew J McBride
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Craig J Hunter
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Zhaoyue Zhang
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Tara TeSlaa
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Xincheng Xu
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Gregory S Ducker
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Joshua D Rabinowitz
- Department of Chemistry, Princeton University, Princeton, NJ, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
12
|
Ritterhoff J, Tian R. Metabolic mechanisms in physiological and pathological cardiac hypertrophy: new paradigms and challenges. Nat Rev Cardiol 2023; 20:812-829. [PMID: 37237146 DOI: 10.1038/s41569-023-00887-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/02/2023] [Indexed: 05/28/2023]
Abstract
Cardiac metabolism is vital for heart function. Given that cardiac contraction requires a continuous supply of ATP in large quantities, the role of fuel metabolism in the heart has been mostly considered from the perspective of energy production. However, the consequence of metabolic remodelling in the failing heart is not limited to a compromised energy supply. The rewired metabolic network generates metabolites that can directly regulate signalling cascades, protein function, gene transcription and epigenetic modifications, thereby affecting the overall stress response of the heart. In addition, metabolic changes in both cardiomyocytes and non-cardiomyocytes contribute to the development of cardiac pathologies. In this Review, we first summarize how energy metabolism is altered in cardiac hypertrophy and heart failure of different aetiologies, followed by a discussion of emerging concepts in cardiac metabolic remodelling, that is, the non-energy-generating function of metabolism. We highlight challenges and open questions in these areas and finish with a brief perspective on how mechanistic research can be translated into therapies for heart failure.
Collapse
Affiliation(s)
- Julia Ritterhoff
- Molecular and Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg, Germany.
- Mitochondria and Metabolism Center, Department of Anaesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA.
| | - Rong Tian
- Mitochondria and Metabolism Center, Department of Anaesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
13
|
Li C, Cheng K, Zhao Q, Jin L, Wang X, Liufu T, Zhao X, Li X, Wang X, Lyu J, Huang D, Li P, Chen XW, Wang Z, Hu X, Quan L, Chen Z. Diazo-carboxyl Click Derivatization Enables Sensitive Analysis of Carboxylic Acid Metabolites in Biosamples. Anal Chem 2023; 95:16976-16986. [PMID: 37943785 DOI: 10.1021/acs.analchem.3c03277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2023]
Abstract
Carboxylic acids are central metabolites in bioenergetics, signal transduction, and post-translation protein regulation. However, the quantitative analysis of carboxylic acids as an indispensable part of metabolomics is prohibitively challenging, particularly in trace amounts of biosamples. Here we report a diazo-carboxyl/hydroxylamine-ketone double click derivatization method for the sensitive analysis of hydrophilic, low-molecular-weight carboxylic acids. In general, our method renders a 5- to 2000-fold higher response in mass spectrometry along with improved chromatographic separation. With this method, we presented the near-single-cell analysis of carboxylic acid metabolites in 10 mouse egg cells before and after fertilization. Malate, fumarate, and β-hydroxybutyrate were found to decrease after fertilization. We also monitored the isotope labeling kinetics of carboxylic acids inside adherent cells cultured in 96-well plates during drug treatment. Finally, we applied this method to plasma or serum samples (5 μL) collected from mice and humans under pathological and physiological conditions. The double click derivatization method paves a way toward single-cell metabolomics and bedside diagnostics.
Collapse
Affiliation(s)
- Cong Li
- College of Future Technology, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
- Peking-Tsinghua Center for Life Science, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Kunlun Cheng
- College of Future Technology, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Qijin Zhao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Li Jin
- College of Future Technology, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Xuelian Wang
- College of Future Technology, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Tongling Liufu
- Department of Neurology, Peking University First Hospital, Beijing 100034, China
| | - Xutong Zhao
- Department of Neurology, Peking University First Hospital, Beijing 100034, China
| | - Xiaochuan Li
- College of Future Technology, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Xiao Wang
- College of Future Technology, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Jia Lyu
- College of Future Technology, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Dong Huang
- College of Future Technology, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Pingping Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xiao-Wei Chen
- College of Future Technology, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
- Peking-Tsinghua Center for Life Science, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Zhaoxia Wang
- Department of Neurology, Peking University First Hospital, Beijing 100034, China
- Beijing Key Laboratory of Neurovascular Disease Discovery, Beijing 100034, China
| | - Xinli Hu
- College of Future Technology, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Li Quan
- College of Future Technology, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Zhixing Chen
- College of Future Technology, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
- Peking-Tsinghua Center for Life Science, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
- National Biomedical Imaging Center, Beijing 100871, China
| |
Collapse
|
14
|
Saini P, Anugula S, Fong YW. The Role of ATP-Binding Cassette Proteins in Stem Cell Pluripotency. Biomedicines 2023; 11:1868. [PMID: 37509507 PMCID: PMC10377311 DOI: 10.3390/biomedicines11071868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/20/2023] [Accepted: 06/22/2023] [Indexed: 07/30/2023] Open
Abstract
Pluripotent stem cells (PSCs) are highly proliferative cells that can self-renew indefinitely in vitro. Upon receiving appropriate signals, PSCs undergo differentiation and can generate every cell type in the body. These unique properties of PSCs require specific gene expression patterns that define stem cell identity and dynamic regulation of intracellular metabolism to support cell growth and cell fate transitions. PSCs are prone to DNA damage due to elevated replicative and transcriptional stress. Therefore, mechanisms to prevent deleterious mutations in PSCs that compromise stem cell function or increase the risk of tumor formation from becoming amplified and propagated to progenitor cells are essential for embryonic development and for using PSCs including induced PSCs (iPSCs) as a cell source for regenerative medicine. In this review, we discuss the role of the ATP-binding cassette (ABC) superfamily in maintaining PSC homeostasis, and propose how their activities can influence cellular signaling and stem cell fate decisions. Finally, we highlight recent discoveries that not all ABC family members perform only canonical metabolite and peptide transport functions in PSCs; rather, they can participate in diverse cellular processes from genome surveillance to gene transcription and mRNA translation, which are likely to maintain the pristine state of PSCs.
Collapse
Affiliation(s)
- Prince Saini
- Brigham Regenerative Medicine Center, Brigham and Women’s Hospital, Boston, MA 02115, USA; (P.S.); (S.A.)
- Department of Medicine, Cardiovascular Medicine Division, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Sharath Anugula
- Brigham Regenerative Medicine Center, Brigham and Women’s Hospital, Boston, MA 02115, USA; (P.S.); (S.A.)
- Department of Medicine, Cardiovascular Medicine Division, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Yick W. Fong
- Brigham Regenerative Medicine Center, Brigham and Women’s Hospital, Boston, MA 02115, USA; (P.S.); (S.A.)
- Department of Medicine, Cardiovascular Medicine Division, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| |
Collapse
|
15
|
Anisimova AS, Kolyupanova NM, Makarova NE, Egorov AA, Kulakovskiy IV, Dmitriev SE. Human Tissues Exhibit Diverse Composition of Translation Machinery. Int J Mol Sci 2023; 24:8361. [PMID: 37176068 PMCID: PMC10179197 DOI: 10.3390/ijms24098361] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/26/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
While protein synthesis is vital for the majority of cell types of the human body, diversely differentiated cells require specific translation regulation. This suggests the specialization of translation machinery across tissues and organs. Using transcriptomic data from GTEx, FANTOM, and Gene Atlas, we systematically explored the abundance of transcripts encoding translation factors and aminoacyl-tRNA synthetases (ARSases) in human tissues. We revised a few known and identified several novel translation-related genes exhibiting strict tissue-specific expression. The proteins they encode include eEF1A1, eEF1A2, PABPC1L, PABPC3, eIF1B, eIF4E1B, eIF4ENIF1, and eIF5AL1. Furthermore, our analysis revealed a pervasive tissue-specific relative abundance of translation machinery components (e.g., PABP and eRF3 paralogs, eIF2B and eIF3 subunits, eIF5MPs, and some ARSases), suggesting presumptive variance in the composition of translation initiation, elongation, and termination complexes. These conclusions were largely confirmed by the analysis of proteomic data. Finally, we paid attention to sexual dimorphism in the repertoire of translation factors encoded in sex chromosomes (eIF1A, eIF2γ, and DDX3), and identified the testis and brain as organs with the most diverged expression of translation-associated genes.
Collapse
Affiliation(s)
- Aleksandra S. Anisimova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Natalia M. Kolyupanova
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Nadezhda E. Makarova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Artyom A. Egorov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Ivan V. Kulakovskiy
- Vavilov Institute of General Genetics, Russian Academy of Sciences, 117971 Moscow, Russia;
- Institute of Protein Research, Russian Academy of Sciences, 142290 Pushchino, Russia
- Laboratory of Regulatory Genomics, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Sergey E. Dmitriev
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia
| |
Collapse
|
16
|
Towanou R, Konmy B, Yovo M, Dansou CC, Dougnon V, Loko FS, Akpovi CD, Baba-Moussa L. Phytochemical Screening, Antioxidant Activity, and Acute Toxicity Evaluation of Senna italica Extract Used in Traditional Medicine. J Toxicol 2023; 2023:6405415. [PMID: 36968147 PMCID: PMC10038741 DOI: 10.1155/2023/6405415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/24/2022] [Accepted: 03/03/2023] [Indexed: 03/19/2023] Open
Abstract
Medicinal plants such as Senna italica are increasingly used for their purgative virtues to treat stomach aches, fever, and jaundice. This study aims to screen the phytochemical compounds and to assess the antioxidant activity in vitro and the acute oral toxicity in vivo of Senna italica leaves. The plant was harvested, dried, pulverized, and preserved. Phytochemical screening was performed using different laboratory protocols. Ethanolic and aqueous extracts were, respectively, obtained by maceration and decoction technics. The assay for free radical scavenging was used to examine the antioxidant activity using DPPH. Acute oral toxicity was performed with aqueous and ethanolic extracts at 5000 mg/kg of body weight on female albinos Wistar rats, weighing 152.44 ± 3.68 g. Subjects were checked for any signs of mortality and macroscopy toxicity during the 14 days of the study. Biochemical and hematological parameters were measured to assess liver and kidney functions, and histological analysis of these organs was conducted. Phytochemical analysis highlighted the presence of total phenols, flavones, tannins, alkaloids, and quinone derivatives. Semiethanolic (78 μg/mL), ethanolic (9.7 μg/mL), and aqueous extract (9.2 μg/mL) showed an interesting antioxidant activity. Biochemical and hematological parameters were normal and not significantly different (p > 0.05). The plant extracts did not produce any toxic effect or mortality at the provided dose. Senna italica extracts induced an increase in the volume of liver and kidney tissues but no necrosis. Thus, lethal dose 50 of Senna italica leaf extract is probably higher than 5000 mg/kg.
Collapse
Affiliation(s)
- Rodrigue Towanou
- Non-Communicable Diseases and Cancer Research Unit, Laboratory of Applied Biology Research, Polytechnic School of Abomey-Calavi, Abomey-Calavi, Benin
- Research Unit in Applied Microbiology and Pharmacology of Natural Substances, Research Laboratory in Applied Biology, Polytechnic School of Abomey-Calavi, University of Abomey-Calavi, Godomey, Benin
- Laboratory of Biology and Molecular Typing in Microbiology, Faculty of Science and Technology, University of Abomey-Calavi, Cotonou, Benin
| | - Basile Konmy
- Laboratory of Biology and Molecular Typing in Microbiology, Faculty of Science and Technology, University of Abomey-Calavi, Cotonou, Benin
- Zootechnical Research and Livestock System Unit, Laboratory of Animal and Fisheries Science (LaSAH), National University of Agriculture, Ketou, Benin
| | - Mahudro Yovo
- Research and Training Laboratory in Applied Chemistry (LERCA), Polytechnic School of Abomey-Calavi, University of Abomey-Calavi, Cotonou, Benin
| | - Christian C. Dansou
- Zootechnical Research and Livestock System Unit, Laboratory of Animal and Fisheries Science (LaSAH), National University of Agriculture, Ketou, Benin
| | - Victorien Dougnon
- Research Unit in Applied Microbiology and Pharmacology of Natural Substances, Research Laboratory in Applied Biology, Polytechnic School of Abomey-Calavi, University of Abomey-Calavi, Godomey, Benin
| | - Frédéric S. Loko
- Research and Training Laboratory in Applied Chemistry (LERCA), Polytechnic School of Abomey-Calavi, University of Abomey-Calavi, Cotonou, Benin
| | - Casimir D. Akpovi
- Non-Communicable Diseases and Cancer Research Unit, Laboratory of Applied Biology Research, Polytechnic School of Abomey-Calavi, Abomey-Calavi, Benin
| | - Lamine Baba-Moussa
- Laboratory of Biology and Molecular Typing in Microbiology, Faculty of Science and Technology, University of Abomey-Calavi, Cotonou, Benin
| |
Collapse
|
17
|
Comparison of fecal and blood metabolome reveals inconsistent associations of the gut microbiota with cardiometabolic diseases. Nat Commun 2023; 14:571. [PMID: 36732517 PMCID: PMC9894915 DOI: 10.1038/s41467-023-36256-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 01/20/2023] [Indexed: 02/04/2023] Open
Abstract
Blood metabolome is commonly used in human studies to explore the associations of gut microbiota-derived metabolites with cardiometabolic diseases. Here, in a cohort of 1007 middle-aged and elderly adults with matched fecal metagenomic (149 species and 214 pathways) and paired fecal and blood targeted metabolomics data (132 metabolites), we find disparate associations with taxonomic composition and microbial pathways when using fecal or blood metabolites. For example, we observe that fecal, but not blood butyric acid significantly associates with both gut microbiota and prevalent type 2 diabetes. These findings are replicated in an independent validation cohort involving 103 adults. Our results suggest that caution should be taken when inferring microbiome-cardiometabolic disease associations from either blood or fecal metabolome data.
Collapse
|
18
|
McBride MJ, Hunter CJ, Rabinowitz JD. Glycine homeostasis requires reverse SHMT flux. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.11.523668. [PMID: 36711816 PMCID: PMC9882094 DOI: 10.1101/2023.01.11.523668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The folate-dependent enzyme serine hydroxymethyltransferase (SHMT) reversibly converts serine into glycine and a tetrahydrofolate-bound one-carbon unit. Such one-carbon unit production plays a critical role in development, the immune system, and cancer. Here we show that the whole-body SHMT flux acts to net consume rather than produce glycine. Pharmacological inhibition of whole-body SHMT1/2 and genetic knockout of liver SHMT2 elevated circulating glycine levels up to eight-fold. Stable isotope tracing revealed that the liver converts glycine to serine, which is then converted by serine dehydratase into pyruvate and burned in the tricarboxylic acid cycle. In response to diets deficient in serine and glycine, de novo biosynthetic flux was unaltered but SHMT2- and serine dehydratase-mediated catabolic flux was lower. Thus, glucose-derived serine synthesis does not respond to systemic demand. Instead, circulating serine and glycine homeostasis is maintained through variable consumption, with liver SHMT2 as a major glycine-consuming enzyme.
Collapse
|
19
|
Tong W, Hannou SA, Wang Y, Astapova I, Sargsyan A, Monn R, Thiriveedi V, Li D, McCann JR, Rawls JF, Roper J, Zhang GF, Herman MA. The intestine is a major contributor to circulating succinate in mice. FASEB J 2022; 36:e22546. [PMID: 36106538 PMCID: PMC9523828 DOI: 10.1096/fj.202200135rr] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 08/24/2022] [Accepted: 08/29/2022] [Indexed: 10/03/2023]
Abstract
The tricarboxylic acid (TCA) cycle is the epicenter of cellular aerobic metabolism. TCA cycle intermediates facilitate energy production and provide anabolic precursors, but also function as intra- and extracellular metabolic signals regulating pleiotropic biological processes. Despite the importance of circulating TCA cycle metabolites as signaling molecules, the source of circulating TCA cycle intermediates remains uncertain. We observe that in mice, the concentration of TCA cycle intermediates in the portal blood exceeds that in tail blood indicating that the gut is a major contributor to circulating TCA cycle metabolites. With a focus on succinate as a representative of a TCA cycle intermediate with signaling activities and using a combination of gut microbiota depletion mouse models and isotopomer tracing, we demonstrate that intestinal microbiota is not a major contributor to circulating succinate. Moreover, we demonstrate that endogenous succinate production is markedly higher than intestinal succinate absorption in normal physiological conditions. Altogether, these results indicate that endogenous succinate production within the intestinal tissue is a major physiological source of circulating succinate. These results provide a foundation for an investigation into the role of the intestine in regulating circulating TCA cycle metabolites and their potential signaling effects on health and disease.
Collapse
Affiliation(s)
- Wenxin Tong
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina, USA
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, USA
| | - Sarah A. Hannou
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, USA
| | - You Wang
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, USA
| | - Inna Astapova
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, USA
- Division of Endocrinology, Metabolism, and Nutrition, Duke University, Durham, North Carolina, USA
| | - Ashot Sargsyan
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, USA
| | - Ruby Monn
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, USA
| | | | - Diana Li
- Division of Gastroenterology, Duke University, Durham, North Carolina, USA
| | - Jessica R. McCann
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University, Durham, NC, USA
| | - John F. Rawls
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University, Durham, NC, USA
| | - Jatin Roper
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina, USA
- Division of Gastroenterology, Duke University, Durham, North Carolina, USA
| | - Guo-fang Zhang
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, USA
| | - Mark A. Herman
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, USA
- Division of Endocrinology, Metabolism, and Nutrition, Duke University, Durham, North Carolina, USA
| |
Collapse
|
20
|
Kaymak I, Luda KM, Duimstra LR, Ma EH, Longo J, Dahabieh MS, Faubert B, Oswald BM, Watson MJ, Kitchen-Goosen SM, DeCamp LM, Compton SE, Fu Z, DeBerardinis RJ, Williams KS, Sheldon RD, Jones RG. Carbon source availability drives nutrient utilization in CD8 + T cells. Cell Metab 2022; 34:1298-1311.e6. [PMID: 35981545 PMCID: PMC10068808 DOI: 10.1016/j.cmet.2022.07.012] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 05/04/2022] [Accepted: 07/23/2022] [Indexed: 12/23/2022]
Abstract
How environmental nutrient availability impacts T cell metabolism and function remains poorly understood. Here, we report that the presence of physiologic carbon sources (PCSs) in cell culture medium broadly impacts glucose utilization by CD8+ T cells, independent of transcriptional changes in metabolic reprogramming. The presence of PCSs reduced glucose contribution to the TCA cycle and increased effector function of CD8+ T cells, with lactate directly fueling the TCA cycle. In fact, CD8+ T cells responding to Listeria infection preferentially consumed lactate over glucose as a TCA cycle substrate in vitro, with lactate enhancing T cell bioenergetic and biosynthetic capacity. Inhibiting lactate-dependent metabolism in CD8+ T cells by silencing lactate dehydrogenase A (Ldha) impaired both T cell metabolic homeostasis and proliferative expansion in vivo. Together, our data indicate that carbon source availability shapes T cell glucose metabolism and identifies lactate as a bioenergetic and biosynthetic fuel for CD8+ effector T cells.
Collapse
Affiliation(s)
- Irem Kaymak
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA
| | - Katarzyna M Luda
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA; University of Copenhagen, Novo Nordisk Foundation Center for Basic Metabolic Research, Blegdamsvej 3B, 2200 København, Denmark
| | - Lauren R Duimstra
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA
| | - Eric H Ma
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA
| | - Joseph Longo
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA
| | - Michael S Dahabieh
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA
| | - Brandon Faubert
- Department of Medicine-Hematology and Oncology, University of Chicago, Chicago, IL, USA
| | - Brandon M Oswald
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA
| | - McLane J Watson
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA
| | - Susan M Kitchen-Goosen
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA
| | - Lisa M DeCamp
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA
| | - Shelby E Compton
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA
| | - Zhen Fu
- Bioinformatics and Biostatistics Core Facility, Van Andel Institute, Grand Rapids, MI, USA
| | - Ralph J DeBerardinis
- Children's Medical Center Research Institute, University of Texas (UT) Southwestern Medical Center, Dallas, TX, USA; Howard Hughes Medical Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Kelsey S Williams
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA
| | - Ryan D Sheldon
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA; Metabolomics and Bioenergetics Core Facility, Van Andel Institute, Grand Rapids, MI, USA
| | - Russell G Jones
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA.
| |
Collapse
|
21
|
Leandro J, Khamrui S, Suebsuwong C, Chen PJ, Secor C, Dodatko T, Yu C, Sanchez R, DeVita RJ, Houten SM, Lazarus MB. Characterization and structure of the human lysine-2-oxoglutarate reductase domain, a novel therapeutic target for treatment of glutaric aciduria type 1. Open Biol 2022; 12:220179. [PMID: 36128717 PMCID: PMC9490328 DOI: 10.1098/rsob.220179] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 08/30/2022] [Indexed: 12/26/2022] Open
Abstract
In humans, a single enzyme 2-aminoadipic semialdehyde synthase (AASS) catalyses the initial two critical reactions in the lysine degradation pathway. This enzyme evolved to be a bifunctional enzyme with both lysine-2-oxoglutarate reductase (LOR) and saccharopine dehydrogenase domains (SDH). Moreover, AASS is a unique drug target for inborn errors of metabolism such as glutaric aciduria type 1 that arise from deficiencies downstream in the lysine degradation pathway. While work has been done to elucidate the SDH domain structurally and to develop inhibitors, neither has been done for the LOR domain. Here, we purify and characterize LOR and show that it is activated by alkylation of cysteine 414 by N-ethylmaleimide. We also provide evidence that AASS is rate-limiting upon high lysine exposure of mice. Finally, we present the crystal structure of the human LOR domain. Our combined work should enable future efforts to identify inhibitors of this novel drug target.
Collapse
Affiliation(s)
- João Leandro
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Susmita Khamrui
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Drug Discovery Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Chalada Suebsuwong
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Drug Discovery Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Peng-Jen Chen
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Drug Discovery Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Cody Secor
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Drug Discovery Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Tetyana Dodatko
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Chunli Yu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Mount Sinai Genomics, Inc, Stamford, CT 06902, USA
| | - Roberto Sanchez
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Drug Discovery Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Robert J. DeVita
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Drug Discovery Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sander M. Houten
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Michael B. Lazarus
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Drug Discovery Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
22
|
Bae H, Lam K, Jang C. Metabolic flux between organs measured by arteriovenous metabolite gradients. Exp Mol Med 2022; 54:1354-1366. [PMID: 36075951 PMCID: PMC9534916 DOI: 10.1038/s12276-022-00803-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/15/2022] [Accepted: 03/04/2022] [Indexed: 12/15/2022] Open
Abstract
Mammalian organs convert dietary nutrients into circulating metabolites and share them to maintain whole-body metabolic homeostasis. While the concentrations of circulating metabolites have been frequently measured in a variety of pathophysiological conditions, the exchange flux of circulating metabolites between organs is not easily measurable due to technical difficulties. Isotope tracing is useful for measuring such fluxes for a metabolite of interest, but the shuffling of isotopic atoms between metabolites requires mathematical modeling. Arteriovenous metabolite gradient measurements can complement isotope tracing to infer organ-specific net fluxes of many metabolites simultaneously. Here, we review the historical development of arteriovenous measurements and discuss their advantages and limitations with key example studies that have revealed metabolite exchange flux between organs in diverse pathophysiological contexts.
Collapse
Affiliation(s)
- Hosung Bae
- Department of Biological Chemistry, Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
| | - Katie Lam
- Department of Biological Chemistry, Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
| | - Cholsoon Jang
- Department of Biological Chemistry, Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
23
|
Flam E, Jang C, Murashige D, Yang Y, Morley MP, Jung S, Kantner DS, Pepper H, Bedi KC, Brandimarto J, Prosser BL, Cappola T, Snyder NW, Rabinowitz JD, Margulies KB, Arany Z. Integrated landscape of cardiac metabolism in end-stage human nonischemic dilated cardiomyopathy. NATURE CARDIOVASCULAR RESEARCH 2022; 1:817-829. [PMID: 36776621 PMCID: PMC9910091 DOI: 10.1038/s44161-022-00117-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 07/08/2022] [Indexed: 01/03/2023]
Abstract
Heart failure (HF) is a leading cause of mortality. Failing hearts undergo profound metabolic changes, but a comprehensive evaluation in humans is lacking. We integrate plasma and cardiac tissue metabolomics of 678 metabolites, genome-wide RNA-sequencing, and proteomic studies to examine metabolic status in 87 explanted human hearts from 39 patients with end-stage HF compared with 48 nonfailing donors. We confirm bioenergetic defects in human HF and reveal selective depletion of adenylate purines required for maintaining ATP levels. We observe substantial reductions in fatty acids and acylcarnitines in failing tissue, despite plasma elevations, suggesting defective import of fatty acids into cardiomyocytes. Glucose levels, in contrast, are elevated. Pyruvate dehydrogenase, which gates carbohydrate oxidation, is de-repressed, allowing increased lactate and pyruvate burning. Tricarboxylic acid cycle intermediates are significantly reduced. Finally, bioactive lipids are profoundly reprogrammed, with marked reductions in ceramides and elevations in lysoglycerophospholipids. These data unveil profound metabolic abnormalities in human failing hearts.
Collapse
Affiliation(s)
- Emily Flam
- Perelman School of Medicine, Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Cholsoon Jang
- Department of Chemistry and Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Danielle Murashige
- Perelman School of Medicine, Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Yifan Yang
- Perelman School of Medicine, Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael P. Morley
- Perelman School of Medicine, Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Sunhee Jung
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Daniel S. Kantner
- Center for Metabolic Disease Research, Department of Cardiovascular Science, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Hannah Pepper
- Center for Metabolic Disease Research, Department of Cardiovascular Science, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Kenneth C. Bedi
- Perelman School of Medicine, Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Jeff Brandimarto
- Perelman School of Medicine, Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Benjamin L. Prosser
- Perelman School of Medicine, Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
- Department of Physiology, Pennsylvania Muscle Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Thomas Cappola
- Perelman School of Medicine, Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Nathaniel W. Snyder
- Center for Metabolic Disease Research, Department of Cardiovascular Science, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Joshua D. Rabinowitz
- Department of Chemistry and Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Kenneth B. Margulies
- Perelman School of Medicine, Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Zolt Arany
- Perelman School of Medicine, Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
24
|
Quek LE, van Geldermalsen M, Guan YF, Wahi K, Mayoh C, Balaban S, Pang A, Wang Q, Cowley MJ, Brown KK, Turner N, Hoy AJ, Holst J. Glutamine addiction promotes glucose oxidation in triple-negative breast cancer. Oncogene 2022; 41:4066-4078. [PMID: 35851845 PMCID: PMC9391225 DOI: 10.1038/s41388-022-02408-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 06/30/2022] [Accepted: 07/01/2022] [Indexed: 11/30/2022]
Abstract
Glutamine is a conditionally essential nutrient for many cancer cells, but it remains unclear how consuming glutamine in excess of growth requirements confers greater fitness to glutamine-addicted cancers. By contrasting two breast cancer subtypes with distinct glutamine dependencies, we show that glutamine-indispensable triple-negative breast cancer (TNBC) cells rely on a non-canonical glutamine-to-glutamate overflow, with glutamine carbon routed once through the TCA cycle. Importantly, this single-pass glutaminolysis increases TCA cycle fluxes and replenishes TCA cycle intermediates in TNBC cells, a process that achieves net oxidation of glucose but not glutamine. The coupling of glucose and glutamine catabolism appears hard-wired via a distinct TNBC gene expression profile biased to strip and then sequester glutamine nitrogen, but hampers the ability of TNBC cells to oxidise glucose when glutamine is limiting. Our results provide a new understanding of how metabolically rigid TNBC cells are sensitive to glutamine deprivation and a way to select vulnerable TNBC subtypes that may be responsive to metabolic-targeted therapies.
Collapse
Affiliation(s)
- Lake-Ee Quek
- School of Mathematics and Statistics, The University of Sydney, Camperdown, NSW, Australia.
| | - Michelle van Geldermalsen
- Origins of Cancer Program, Centenary Institute, The University of Sydney, Camperdown, NSW, Australia
- Sydney Medical School, The University of Sydney, Camperdown, NSW, Australia
| | - Yi Fang Guan
- School of Medical Sciences and School of Clinical Medicine, UNSW Sydney, Kensington, NSW, Australia
| | - Kanu Wahi
- School of Medical Sciences and School of Clinical Medicine, UNSW Sydney, Kensington, NSW, Australia
| | - Chelsea Mayoh
- Children's Cancer Institute, Lowy Cancer Centre, UNSW Sydney, Kensington, NSW, Australia
- School of Women's and Children's Health, UNSW Sydney, Kensington, NSW, Australia
| | - Seher Balaban
- School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia
| | - Angel Pang
- School of Medical Sciences and School of Clinical Medicine, UNSW Sydney, Kensington, NSW, Australia
| | - Qian Wang
- School of Medical Sciences and School of Clinical Medicine, UNSW Sydney, Kensington, NSW, Australia
| | - Mark J Cowley
- Children's Cancer Institute, Lowy Cancer Centre, UNSW Sydney, Kensington, NSW, Australia
- School of Women's and Children's Health, UNSW Sydney, Kensington, NSW, Australia
| | - Kristin K Brown
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
- Department of Biochemistry and Pharmacology, The University of Melbourne, Melbourne, VIC, Australia
| | - Nigel Turner
- Department of Pharmacology, School of Medical Sciences, UNSW Sydney, Kensington, NSW, Australia
| | - Andrew J Hoy
- School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia
| | - Jeff Holst
- School of Medical Sciences and School of Clinical Medicine, UNSW Sydney, Kensington, NSW, Australia.
| |
Collapse
|
25
|
Nemkov T, Skinner S, Diaw M, Diop S, Samb A, Connes P, D’Alessandro A. Plasma Levels of Acyl-Carnitines and Carboxylic Acids Correlate With Cardiovascular and Kidney Function in Subjects With Sickle Cell Trait. Front Physiol 2022; 13:916197. [PMID: 35910560 PMCID: PMC9326174 DOI: 10.3389/fphys.2022.916197] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 06/22/2022] [Indexed: 11/13/2022] Open
Abstract
Subjects with sickle cell trait (SCT) carry one copy of mutated β-globin gene at position E6V at the origin of the production of sickle hemoglobin (HbS). Indeed, individuals with SCT have both normal hemoglobin and HbS, in contrast to patients with sickle cell disease who inherited of two copies of the mutated gene. Although SCT is generally benign/asymptomatic, carriers may develop certain adverse outcomes such as renal complications, venous thromboembolism, exercise-induced rhabdomyolysis … However, little is known about whether similar metabolic pathways are affected in individuals with SCT and whether these metabolic derangements, if present, correlate to clinically relevant parameters. In this study, we performed metabolomics analysis of plasma from individuals with sickle cell trait (n = 34) compared to healthy controls (n = 30). Results indicated a significant increase in basal circulating levels of hemolysis markers, mono- (pyruvate, lactate), di- and tri-carboxylates (including all Krebs cycle intermediates), suggestive of systems-wide mitochondrial dysfunction in individuals with SCT. Elevated levels of kynurenines and indoles were observed in SCT samples, along with increases in the levels of oxidative stress markers (advanced glycation and protein-oxidation end-products, malondialdehyde, oxylipins, eicosanoids). Increases in circulating levels of acyl-carnitines and fatty acids were observed, consistent with increased membrane lipid damage in individuals with sickle cell trait. Finally, correlation analyses to clinical co-variates showed that alterations in the aforementioned pathways strongly correlated with clinical measurements of blood viscosity, renal (glomerular filtration rate, microalbuminuria, uremia) and cardiovascular function (carotid-femoral pulse wave velocity, blood pressure).
Collapse
Affiliation(s)
- Travis Nemkov
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO, United States
| | - Sarah Skinner
- Inter-university Laboratory of Biology of Motor Function EA7424, Vascular Biology and the Red Blood Cell Team, Claude Bernard University Lyon 1, Lyon, France
| | - Mor Diaw
- Laboratory of Physiology and Functional Exploration, FMPO, UCAD, Dakar, Senegal
- IRL3189 Environnement, Santé, Sociétés CNRS/UCAD Dakar/ UGB Saint-Louis/ USTT Bamako/ CNRST Ouagadougou, Dakar, Senegal
| | - Saliou Diop
- Laboratory of Hemato-immunology, FMPO, UCAD, Dakar, Senegal
| | - Abdoulaye Samb
- Laboratory of Physiology and Functional Exploration, FMPO, UCAD, Dakar, Senegal
- IRL3189 Environnement, Santé, Sociétés CNRS/UCAD Dakar/ UGB Saint-Louis/ USTT Bamako/ CNRST Ouagadougou, Dakar, Senegal
| | - Philippe Connes
- Inter-university Laboratory of Biology of Motor Function EA7424, Vascular Biology and the Red Blood Cell Team, Claude Bernard University Lyon 1, Lyon, France
| | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO, United States
| |
Collapse
|
26
|
Fulghum K, Collins HE, Jones SP, Hill BG. Influence of biological sex and exercise on murine cardiac metabolism. JOURNAL OF SPORT AND HEALTH SCIENCE 2022; 11:479-494. [PMID: 35688382 PMCID: PMC9338340 DOI: 10.1016/j.jshs.2022.06.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 04/07/2022] [Accepted: 04/27/2022] [Indexed: 05/03/2023]
Abstract
Although the structural and functional effects of exercise on the heart are well established, the metabolic changes that occur in the heart during and after exercise remain unclear. In this study, we used metabolomics to assess time-dependent changes in the murine cardiac metabolome following 1 session of treadmill exercise. After the exercise bout, we also recorded blood lactate, glucose, and ketone body levels and measured cardiac mitochondrial respiration. In both male and female mice, moderate- and high-intensity exercise acutely increased blood lactate levels. In both sexes, low- and moderate-intensity exercise augmented circulating 3-hydroxybutryrate levels immediately after the exercise bout; however, only in female mice did high-intensity exercise increase 3-hydroxybutyrate levels, with significant increases occurring 1 h after the exercise session. Untargeted metabolomics analyses of sedentary female and male hearts suggest considerable sex-dependent differences in basal cardiac metabolite levels, with female hearts characterized by higher levels of pantothenate, pyridoxamine, homoarginine, tryptophan, and several glycerophospholipid and sphingomyelin species and lower levels of numerous metabolites, including acetyl coenzyme A, glucuronate, gulonate, hydroxyproline, prolyl-hydroxyproline, carnosine, anserine, and carnitinylated and glycinated species, as compared with male hearts. Immediately after a bout of treadmill exercise, both male and female hearts had higher levels of corticosterone; however, female mice showed more extensive exercise-induced changes in the cardiac metabolome, characterized by significant, time-dependent changes in amino acids (e.g., serine, alanine, tyrosine, tryptophan, branched-chain amino acids) and the ketone body 3-hydroxybutyrate. Results from experiments using isolated cardiac mitochondria suggest that high-intensity treadmill exercise does not acutely affect respiration or mitochondrial coupling; however, female cardiac mitochondria demonstrate generally higher adenosine diphosphate sensitivity compared with male cardiac mitochondria. Collectively, these findings in mice reveal key sex-dependent differences in cardiac metabolism and suggest that the metabolic network in the female heart is more responsive to physiological stress caused by exercise.
Collapse
Affiliation(s)
- Kyle Fulghum
- Diabetes and Obesity Center, Department of Medicine, Division of Environmental Medicine, Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY 40202, USA; Department of Physiology, University of Louisville, Louisville, KY 40202, USA
| | - Helen E Collins
- Diabetes and Obesity Center, Department of Medicine, Division of Environmental Medicine, Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY 40202, USA
| | - Steven P Jones
- Diabetes and Obesity Center, Department of Medicine, Division of Environmental Medicine, Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY 40202, USA
| | - Bradford G Hill
- Diabetes and Obesity Center, Department of Medicine, Division of Environmental Medicine, Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY 40202, USA.
| |
Collapse
|
27
|
Van Tine BA, Lyssiotis CA. In an era of ctDNA, is metabolomics the new kid on the block? Clin Cancer Res 2022; 28:1477-1478. [PMID: 35140120 DOI: 10.1158/1078-0432.ccr-21-4180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/01/2022] [Accepted: 02/02/2022] [Indexed: 11/16/2022]
Abstract
Mass action drives the serum homeostasis of metabolites. Recent developments in biofluid metabolomics suggest the potential to harness these changes using small volumes of blood to diagnosis, monitor, and risk stratify cancer patients. This current study may represent a complementary technology to circulating tumor DNA detection.
Collapse
Affiliation(s)
| | - Costas A Lyssiotis
- Department of Molecular & Integrative Physiology and Department of Internal Medicine, Division of Gastroenterology, University of Michigan–Ann Arbor
| |
Collapse
|
28
|
Wallace M. Metabolic balance-a masterclass in mass action. Nat Metab 2022; 4:17-18. [PMID: 35058632 DOI: 10.1038/s42255-021-00522-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Martina Wallace
- Institute of Food and Health, School of Agriculture and Food Science, University College Dublin, Dublin, Ireland.
| |
Collapse
|