9
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Impaired skeletal muscle mitochondrial pyruvate uptake rewires glucose metabolism to drive whole-body leanness

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Metabolic cycles are a fundamental element of cellular and organismal function. Among the most critical in higher organisms is the Cori Cycle, the systemic cycling between lactate and glucose. Here, skeletal muscle-specific Mitochondrial Pyruvate Carrier (MPC) deletion in mice diverted pyruvate into circulating lactate. This switch disinhibited muscle fatty acid oxidation and drove Cori Cycling that contributed to increased energy expenditure. Loss of muscle MPC activity led to strikingly decreased adiposity with complete muscle mass and strength retention. Notably, despite decreasing muscle glucose oxidation, muscle MPC disruption increased muscle glucose uptake and whole-body insulin sensitivity. Furthermore, chronic and acute muscle MPC deletion accelerated fat mass loss on a normal diet after high fat diet-induced obesity. Our results illuminate the role of the skeletal muscle MPC as a whole-body carbon flux control point. They highlight the potential utility of modulating muscle pyruvate utilization to ameliorate obesity and type 2 diabetes.

          Related collections

          Most cited references38

          • Record: found
          • Abstract: found
          • Article: not found

          A mitochondrial pyruvate carrier required for pyruvate uptake in yeast, Drosophila, and humans.

          Pyruvate constitutes a critical branch point in cellular carbon metabolism. We have identified two proteins, Mpc1 and Mpc2, as essential for mitochondrial pyruvate transport in yeast, Drosophila, and humans. Mpc1 and Mpc2 associate to form an ~150-kilodalton complex in the inner mitochondrial membrane. Yeast and Drosophila mutants lacking MPC1 display impaired pyruvate metabolism, with an accumulation of upstream metabolites and a depletion of tricarboxylic acid cycle intermediates. Loss of yeast Mpc1 results in defective mitochondrial pyruvate uptake, and silencing of MPC1 or MPC2 in mammalian cells impairs pyruvate oxidation. A point mutation in MPC1 provides resistance to a known inhibitor of the mitochondrial pyruvate carrier. Human genetic studies of three families with children suffering from lactic acidosis and hyperpyruvatemia revealed a causal locus that mapped to MPC1, changing single amino acids that are conserved throughout eukaryotes. These data demonstrate that Mpc1 and Mpc2 form an essential part of the mitochondrial pyruvate carrier.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Glutamine oxidation maintains the TCA cycle and cell survival during impaired mitochondrial pyruvate transport.

            Alternative modes of metabolism enable cells to resist metabolic stress. Inhibiting these compensatory pathways may produce synthetic lethality. We previously demonstrated that glucose deprivation stimulated a pathway in which acetyl-CoA was formed from glutamine downstream of glutamate dehydrogenase (GDH). Here we show that import of pyruvate into the mitochondria suppresses GDH and glutamine-dependent acetyl-CoA formation. Inhibiting the mitochondrial pyruvate carrier (MPC) activates GDH and reroutes glutamine metabolism to generate both oxaloacetate and acetyl-CoA, enabling persistent tricarboxylic acid (TCA) cycle function. Pharmacological blockade of GDH elicited largely cytostatic effects in culture, but these effects became cytotoxic when combined with MPC inhibition. Concomitant administration of MPC and GDH inhibitors significantly impaired tumor growth compared to either inhibitor used as a single agent. Together, the data define a mechanism to induce glutaminolysis and uncover a survival pathway engaged during compromised supply of pyruvate to the mitochondria. Copyright © 2014 Elsevier Inc. All rights reserved.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              Age-Associated Loss of OPA1 in Muscle Impacts Muscle Mass, Metabolic Homeostasis, Systemic Inflammation, and Epithelial Senescence

              Summary Mitochondrial dysfunction occurs during aging, but its impact on tissue senescence is unknown. Here, we find that sedentary but not active humans display an age-related decline in the mitochondrial protein, optic atrophy 1 (OPA1), that is associated with muscle loss. In adult mice, acute, muscle-specific deletion of Opa1 induces a precocious senescence phenotype and premature death. Conditional and inducible Opa1 deletion alters mitochondrial morphology and function but not DNA content. Mechanistically, the ablation of Opa1 leads to ER stress, which signals via the unfolded protein response (UPR) and FoxOs, inducing a catabolic program of muscle loss and systemic aging. Pharmacological inhibition of ER stress or muscle-specific deletion of FGF21 compensates for the loss of Opa1, restoring a normal metabolic state and preventing muscle atrophy and premature death. Thus, mitochondrial dysfunction in the muscle can trigger a cascade of signaling initiated at the ER that systemically affects general metabolism and aging.
                Bookmark

                Author and article information

                Contributors
                Role: Senior Editor
                Role: Reviewing Editor
                Journal
                eLife
                Elife
                eLife
                eLife
                eLife Sciences Publications, Ltd
                2050-084X
                18 July 2019
                2019
                : 8
                : e45873
                Affiliations
                [1 ]deptDepartment of Biochemistry, Carver College of Medicine University of Iowa Iowa CityUnited States
                [2 ]deptDepartment of Internal Medicine, Carver College of Medicine University of Iowa Iowa CityUnited States
                [3 ]deptDepartment of Chemistry, School of Medicine Washington University St. LouisUnited States
                [4 ]deptFraternal Order of the Eagles Diabetes Research Center (FOEDRC), Carver College of Medicine University of Iowa Iowa CityUnited States
                [5 ]deptFOEDRC Metabolic Phenotyping Core Facility, Carver College of Medicine University of Iowa Iowa CityUnited States
                [6 ]deptDepartment of Biochemistry, School of Medicine University of Utah Salt Lake CityUnited States
                [7 ]deptMetabolomics Core Research Facility, School of Medicine University of Utah Salt Lake CityUnited States
                [8 ]deptDepartment of Molecular Physiology and Biophysics, Carver College of Medicine University of Iowa Iowa CityUnited States
                [9 ]deptPappajohn Biomedical Institute, Carver College of Medicine University of Iowa Iowa CityUnited States
                [10 ]deptAbboud Cardiovascular Research Center, Carver College of Medicine University of Iowa Iowa CityUnited States
                [11 ]deptDepartment of Physical Therapy and Rehabilitation Science, Carver College of Medicine University of Iowa Iowa CityUnited States
                [12 ]deptDepartment of Pediatrics, Carver College of Medicine University of Iowa Iowa CityUnited States
                [13 ]deptDepartment of Veterans Affairs, Medical Center, Carver College of Medicine University of Iowa Iowa CityUnited States
                [14 ]deptFOEDRC Metabolomics Core Facility, Carver College of Medicine University of Iowa Iowa CityUnited States
                University of Oxford United Kingdom
                The University of Sydney Australia
                The University of Sydney Australia
                East Carolina University United States
                Yale University United States
                Duke University United States
                Author notes
                [†]

                These authors contributed equally to this work.

                Author information
                https://orcid.org/0000-0002-6132-1129
                https://orcid.org/0000-0001-9425-8448
                https://orcid.org/0000-0002-4101-0075
                http://orcid.org/0000-0002-9495-802X
                http://orcid.org/0000-0002-3748-6193
                https://orcid.org/0000-0003-4549-6567
                Article
                45873
                10.7554/eLife.45873
                6684275
                31305240
                b8a11a37-6c40-40a5-b23c-300b5c3870d7
                © 2019, Sharma et al

                This article is distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use and redistribution provided that the original author and source are credited.

                History
                : 07 February 2019
                : 15 July 2019
                Funding
                Funded by: FundRef http://dx.doi.org/10.13039/100000002, National Institutes of Health;
                Award ID: DK104998
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/100000002, National Institutes of Health;
                Award ID: AR059190
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/100000002, National Institutes of Health;
                Award ID: HD084645
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/100000002, National Institutes of Health;
                Award ID: HD082109
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/100000002, National Institutes of Health;
                Award ID: DK092412
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/100000002, National Institutes of Health;
                Award ID: ES028365
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/100000002, National Institutes of Health;
                Award ID: HL130146
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/100000002, National Institutes of Health;
                Award ID: HL007344
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/100000002, National Institutes of Health;
                Award ID: DK116522
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/100000002, National Institutes of Health;
                Award ID: GM007337
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/100000002, National Institutes of Health;
                Award ID: HL007638
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/100000002, National Institutes of Health;
                Award ID: DK101183
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/100000041, American Diabetes Association;
                Award ID: 1-18-PDF-060
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/100000002, National Institutes of Health;
                Award ID: DK112751
                Award Recipient :
                The funders had no role in study design, data collection and interpretation, or the decision to submit the work for publication.
                Categories
                Research Article
                Cell Biology
                Human Biology and Medicine
                Custom metadata
                Skeletal muscle mitochondrial pyruvate carrier disruption increases muscle fatty acid oxidation and systemic glucose turnover that drive whole-body leanness.

                Life sciences
                mitochondrial pyruvate carrier,skeletal muscle,cori cycle,diabetes,obesity,glucose uptake,mouse

                Comments

                Comment on this article