301
|
Goncalves RLS, Rothschild DE, Quinlan CL, Scott GK, Benz CC, Brand MD. Sources of superoxide/H2O2 during mitochondrial proline oxidation. Redox Biol 2014; 2:901-9. [PMID: 25184115 PMCID: PMC4143814 DOI: 10.1016/j.redox.2014.07.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 07/07/2014] [Indexed: 12/28/2022] Open
Abstract
p53 Inducible gene 6 (PIG6) encodes mitochondrial proline dehydrogenase (PRODH) and is up-regulated several fold upon p53 activation. Proline dehydrogenase is proposed to generate radicals that contribute to cancer cell apoptosis. However, there are at least 10 mitochondrial sites that can produce superoxide and/or H2O2, and it is unclear whether proline dehydrogenase generates these species directly, or instead drives production by other sites. Amongst six cancer cell lines, ZR75-30 human breast cancer cells had the highest basal proline dehydrogenase levels, and mitochondria isolated from ZR75-30 cells consumed oxygen and produced H2O2 with proline as sole substrate. Insects use proline oxidation to fuel flight, and mitochondria isolated from Drosophila melanogaster were even more active with proline as sole substrate than ZR75-30 mitochondria. Using mitochondria from these two models we identified the sites involved in formation of superoxide/H2O2 during proline oxidation. In mitochondria from Drosophila the main sites were respiratory complexes I and II. In mitochondria from ZR75-30 breast cancer cells the main sites were complex I and the oxoglutarate dehydrogenase complex. Even with combinations of substrates and respiratory chain inhibitors designed to minimize the contributions of other sites and maximize any superoxide/H2O2 production from proline dehydrogenase itself, there was no significant direct contribution of proline dehydrogenase to the observed H2O2 production. Thus proline oxidation by proline dehydrogenase drives superoxide/H2O2 production, but it does so mainly or exclusively by providing anaplerotic carbon for other mitochondrial dehydrogenases and not by producing superoxide/H2O2 directly. Proline dehydrogenase is thought to produce reactive oxygen species (ROS) in cancer cells and to promote apoptosis. Isolated mitochondria from Drosophila melanogaster and from a human breast cancer cell line oxidize proline producing superoxide/H2O2 at measurable rates. Proline oxidation drives superoxide/H2O2 production indirectly at other sites and it is unlikely that proline dehydrogenase produces superoxide/H2O2 itself. In Drosophila, superoxide/H2O2 arises from sites IF and IIF (the flavin sites from complexes I and II, respectively). In the breast cancer cell line the main sites are IF and OF (from the oxoglutarate dehydrogenase complex).
Collapse
Key Words
- A5, atpenin A5
- AT, aminotransferase
- Asp, asparate
- Cancer cell mitochondria
- Drosophila
- Electron transport chain
- GDH, glutamate dehydrogenase
- GSA, glutamic semi-aldehyde
- Hydrogen peroxide
- IF, flavin of complex I
- IIF, flavin of complex II
- IIIQo, quinone binding site on the outer/cytosolic face of complex III
- OF, Flavin of the oxoglutarate dehydrogenase complex
- OGDH, 2-oxoglutarate dehydrogenase complex
- Oxa, oxaloacetate
- P5C, Δ1-pyrroline-5-carboxylate
- PIG6, proline dehydrogenase inducible gene 6
- PRODH, proline dehydrogenase
- Proline dehydrogenase (PRODH)
- ROS, reactive oxygen species
- Reactive oxygen species
- SCS, succinyl-CoA synthase
- Superoxide
- TCA, tricarboxylic acid
- oAB, o-aminobenzaldehyde
Collapse
Affiliation(s)
| | | | | | - Gary K Scott
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | | | - Martin D Brand
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| |
Collapse
|
302
|
Tyrrell DJ, Bharadwaj MS, Van Horn CG, Kritchevsky SB, Nicklas BJ, Molina AJA. Respirometric Profiling of Muscle Mitochondria and Blood Cells Are Associated With Differences in Gait Speed Among Community-Dwelling Older Adults. J Gerontol A Biol Sci Med Sci 2014; 70:1394-9. [PMID: 25030980 DOI: 10.1093/gerona/glu096] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 05/19/2014] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Gait speed provides an integrated measure of physical ability that is predictive of morbidity, disability, and mortality in older adults. Energy demands associated with walking suggest that mitochondrial bioenergetics may play a role in gait speed. Here, we examined the relationship between gait speed and skeletal muscle mitochondrial bioenergetics, and further evaluated whether blood-based bioenergetic profiling might have similar associations with gait speed. METHODS Participants in this study were comprised of two subsets (n = 17 per subset) and were overweight/obese (body mass index, 30.9 ± 2.37), well-functioning, community-dwelling older adults (69.1 ± 3.69 years) without major comorbidity. Gait speeds were calculated from a fast-paced 400 m walk test. Respiratory control ratios were measured from mitochondria isolated from leg skeletal muscle biopsies from one subset. Maximal respiration and spare respiratory capacity were measured from peripheral blood mononuclear cells from the other subset. RESULTS Individual differences in gait speed correlated directly with respiratory control ratio of mitochondria isolated from skeletal muscle (r = .536, p = .027) and with both maximal respiration and spare respiratory capacity of peripheral blood mononuclear cells (r = .585 and p = .014; r = .609 and p = .009, respectively). CONCLUSIONS The bioenergetic profile of mitochondria isolated from skeletal muscle is associated with gait speed in older adults. Blood-based bioenergetic profiling is also associated with gait speed and may provide an alternative measure of mitochondrial function.
Collapse
Affiliation(s)
- Daniel J Tyrrell
- Sticht Center on Aging and Department of Internal Medicine, Section on Gerontology and Geriatrics, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Manish S Bharadwaj
- Sticht Center on Aging and Department of Internal Medicine, Section on Gerontology and Geriatrics, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Cynthia G Van Horn
- Sticht Center on Aging and Department of Internal Medicine, Section on Gerontology and Geriatrics, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Stephen B Kritchevsky
- Sticht Center on Aging and Department of Internal Medicine, Section on Gerontology and Geriatrics, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Barbara J Nicklas
- Sticht Center on Aging and Department of Internal Medicine, Section on Gerontology and Geriatrics, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Anthony J A Molina
- Sticht Center on Aging and Department of Internal Medicine, Section on Gerontology and Geriatrics, Wake Forest School of Medicine, Winston-Salem, North Carolina.
| |
Collapse
|
303
|
Jimenez AG, Williams JB. Cellular metabolic rates from primary dermal fibroblast cells isolated from birds of different body masses. Comp Biochem Physiol A Mol Integr Physiol 2014; 176:41-8. [PMID: 25038299 DOI: 10.1016/j.cbpa.2014.07.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 06/14/2014] [Accepted: 07/09/2014] [Indexed: 11/19/2022]
Abstract
The rate of metabolism is the speed at which organisms use energy, an integration of energy transformations within the body; it governs biological processes that influence rates of growth and reproduction. Progress at understanding functional linkages between whole organism metabolic rate and underlying mechanisms that influence its magnitude has been slow despite the central role this issue plays in evolutionary and physiological ecology. Previous studies that have attempted to relate how cellular processes translate into whole-organism physiology have done so over a range of body masses of subjects. However, the data still remains controversial when observing metabolic rates at the cellular level. To bridge the gap between these ideas, we examined cellular metabolic rate of primary dermal fibroblasts isolated from 49 species of birds representing a 32,000-fold range in body masses to test the hypothesis that metabolic rate of cultured cells scales with body size. We used a Seahorse XF-96 Extracellular flux analyzer to measure cellular respiration in fibroblasts. Additionally, we measured fibroblast size and mitochondrial content. We found no significant correlation between cellular metabolic rate, cell size, or mitochondrial content and body mass. Additionally, there was a significant relationship between cellular basal metabolic rate and proton leak in these cells. We conclude that metabolic rate of cells isolated in culture does not scale with body mass, but cellular metabolic rate is correlated to growth rate in birds.
Collapse
Affiliation(s)
- Ana Gabriela Jimenez
- Department of Evolution, Ecology and Organismal Biology, Ohio State University, 318 W. 12th Ave., Columbus, OH 43210, USA.
| | - Joseph B Williams
- Department of Evolution, Ecology and Organismal Biology, Ohio State University, 318 W. 12th Ave., Columbus, OH 43210, USA
| |
Collapse
|
304
|
Beauchamp B, Ghosh S, Dysart MW, Kanaan GN, Chu A, Blais A, Rajamanickam K, Tsai EC, Patti ME, Harper ME. Low birth weight is associated with adiposity, impaired skeletal muscle energetics and weight loss resistance in mice. Int J Obes (Lond) 2014; 39:702-11. [PMID: 25091727 PMCID: PMC4326251 DOI: 10.1038/ijo.2014.120] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 06/07/2014] [Accepted: 06/28/2014] [Indexed: 01/29/2023]
Abstract
BACKGROUND In utero undernutrition is associated with obesity and insulin resistance, although its effects on skeletal muscle remain poorly defined. Therefore, in the current study we explored the effects of in utero food restriction on muscle energy metabolism in mice. METHODS We used an experimental mouse model system of maternal undernutrition during late pregnancy to examine offspring from undernourished dams (U) and control offspring from ad libitum-fed dams (C). Weight loss of 10-week-old offspring on a 4-week 40% calorie-restricted diet was also followed. Experimental approaches included bioenergetic analyses in isolated mitochondria, intact (permeabilized) muscle and at the whole body level. RESULTS U have increased adiposity and decreased glucose tolerance compared to C. Strikingly, when U are put on a 40% calorie-restricted diet they lose half as much weight as calorie-restricted controls. Mitochondria from muscle overall from U had decreased coupled (state 3) and uncoupled (state 4) respiration and increased maximal respiration compared to C. Mitochondrial yield was lower in U than C. In permeabilized fiber preparations from mixed fiber-type muscle, U had decreased mitochondrial content and decreased adenylate-free leak respiration, fatty acid oxidative capacity and state 3 respiratory capacity through complex I. Fiber maximal oxidative phosphorylation capacity did not differ between U and C but was decreased with calorie restriction. CONCLUSIONS Our results reveal that in utero undernutrition alters metabolic physiology through a profound effect on skeletal muscle energetics and blunts response to a hypocaloric diet in adulthood. We propose that mitochondrial dysfunction links undernutrition in utero with metabolic disease in adulthood.
Collapse
Affiliation(s)
- B Beauchamp
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - S Ghosh
- Cardiovascular & Metabolic Disorders Program, Duke-NUS Graduate Medical School, Singapore, Singapore
| | - M W Dysart
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - G N Kanaan
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - A Chu
- 1] Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada [2] Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - A Blais
- 1] Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada [2] Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - K Rajamanickam
- Division of Neurosurgery, Department of Surgery, Faculty of Medicine, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - E C Tsai
- Division of Neurosurgery, Department of Surgery, Faculty of Medicine, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - M-E Patti
- Division of Integrative Physiology and Metabolism, Joslin Diabetes Center, Boston, MA, USA
| | - M-E Harper
- 1] Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada [2] Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
305
|
Region specific mitochondrial impairment in mice with widespread overexpression of alpha-synuclein. Neurobiol Dis 2014; 70:204-13. [PMID: 25016198 DOI: 10.1016/j.nbd.2014.06.017] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 06/17/2014] [Accepted: 06/22/2014] [Indexed: 11/21/2022] Open
Abstract
Parkinson's disease (PD) is characterized by the progressive degeneration of nigrostriatal dopaminergic neurons leading to motor deficits. The mechanisms underlying the preferential vulnerability of nigrostriatal dopaminergic neurons in PD remain poorly understood. Recent evidence supports a role for mitochondrial dysfunction and increased oxidative stress in PD pathogenesis. Genetic and pathological studies also point to alpha-synuclein as a critical factor in both familial and sporadic forms of the disease; alpha-synuclein pathology affects mitochondrial function but is widespread in PD brain, raising the question of its role in the greater vulnerability of nigrostriatal neurons in PD. We have examined mitochondrial function and oxidative damage in mice overexpressing human wild type alpha-synuclein broadly throughout the nervous system under the Thy1 promoter (Thy1-aSyn mice) between 4 and 8months of age. Similar levels of alpha-synuclein accumulation in mitochondria were detected in the ventral midbrain, striatum and cortex of Thy1-aSyn mice. However, analysis of mitochondrial respiration using Seahorse XF analyzer showed defects in mitochondrial respiratory complexes I, II, IV and V specifically in the midbrain, and IV and V in the striatum, of Thy1-aSyn mice compared to wild type littermates; mitochondrial complex I activity assay by ELISA confirmed a 40% inhibition specifically in the ventral midbrain. Mitochondrial dysfunction can contribute to oxidative stress and we observed a 40% increase in 4-hydroxynenal and 2-fold increase in malondialdehyde levels, indicative of a high level of lipid peroxidation, specifically in the ventral midbrain of Thy1-aSyn mice. The levels of peroxiredoxin 2, a neuronal antioxidant enzyme that is involved in removal of H2O2 and other toxic peroxides were decreased in the midbrain whereas its oxidized form increased 4-fold, suggesting that antioxidant defenses were compromised in this region. In contrast, peroxiredoxin 2 increased in the striatum and cortex, which may contribute to their protection in the presence of high levels of alpha-synuclein. Thus, in mice over-expressing alpha-synuclein, mitochondrial dysfunction occurred preferentially in nigrostriatal dopaminergic neurons many months before striatal dopamine loss occurs at 14months of age. This may contribute to a higher level of oxidative stress that overwhelms antioxidant defense in these neurons, leading to their increased vulnerability in PD.
Collapse
|
306
|
Hey-Mogensen M, Goncalves RLS, Orr AL, Brand MD. Production of superoxide/H2O2 by dihydroorotate dehydrogenase in rat skeletal muscle mitochondria. Free Radic Biol Med 2014; 72:149-55. [PMID: 24746616 DOI: 10.1016/j.freeradbiomed.2014.04.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 04/01/2014] [Accepted: 04/05/2014] [Indexed: 10/25/2022]
Abstract
Dehydrogenases that use ubiquinone as an electron acceptor, including complex I of the respiratory chain, complex II, and glycerol-3-phosphate dehydrogenase, are known to be direct generators of superoxide and/or H2O2. Dihydroorotate dehydrogenase oxidizes dihydroorotate to orotate and reduces ubiquinone to ubiquinol during pyrimidine metabolism, but it is unclear whether it produces superoxide and/or H2O2 directly or does so only indirectly from other sites in the electron transport chain. Using mitochondria isolated from rat skeletal muscle we establish that dihydroorotate oxidation leads to superoxide/H2O2 production at a fairly high rate of about 300pmol H2O2·min(-1)·mg protein(-1) when oxidation of ubiquinol is prevented and complex II is uninhibited. This H2O2 production is abolished by brequinar or leflunomide, known inhibitors of dihydroorotate dehydrogenase. Eighty percent of this rate is indirect, originating from site IIF of complex II, because it can be prevented by malonate or atpenin A5, inhibitors of complex II. In the presence of inhibitors of all known sites of superoxide/H2O2 production (rotenone to inhibit sites in complex I (site IQ and, indirectly, site IF), myxothiazol to inhibit site IIIQo in complex III, and malonate plus atpenin A5 to inhibit site IIF in complex II), dihydroorotate dehydrogenase generates superoxide/H2O2, at a small but significant rate (23pmol H2O2·min(-1)·mg protein(-1)), from the ubiquinone-binding site. We conclude that dihydroorotate dehydrogenase can generate superoxide and/or H2O2 directly at low rates and is also capable of indirect production at higher rates from other sites through its ability to reduce the ubiquinone pool.
Collapse
Affiliation(s)
- Martin Hey-Mogensen
- Buck Institute for Research on Aging, Novato, CA 94945, USA; Department of Biomedical Sciences, Center for Healthy Aging, Copenhagen University, Copenhagen, Denmark
| | | | - Adam L Orr
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Martin D Brand
- Buck Institute for Research on Aging, Novato, CA 94945, USA.
| |
Collapse
|
307
|
Rogers GW, Nadanaciva S, Swiss R, Divakaruni AS, Will Y. Assessment of fatty acid beta oxidation in cells and isolated mitochondria. ACTA ACUST UNITED AC 2014; 60:25.3.1-19. [PMID: 24865647 DOI: 10.1002/0471140856.tx2503s60] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Fatty acid beta oxidation is a major pathway of energy metabolism and occurs primarily in mitochondria. Drug-induced modulation of this pathway can cause adverse effects such as liver injury, or be beneficial for treating heart failure, type 2 diabetes, and obesity. Hence, in vitro assays that are able to identify compounds that affect fatty acid oxidation are of value for toxicity assessments, as well as for efficacy assessments. Here, we describe two high-throughput assays, one for assessing fatty acid oxidation in cells and the other for assessing fatty acid oxidation in isolated rat liver mitochondria. Both assays measure fatty acid-driven oxygen consumption and can be used for rapid and robust screening of compounds that modulate fatty acid oxidation.
Collapse
|
308
|
Divakaruni AS, Rogers GW, Murphy AN. Measuring Mitochondrial Function in Permeabilized Cells Using the Seahorse XF Analyzer or a Clark-Type Oxygen Electrode. ACTA ACUST UNITED AC 2014; 60:25.2.1-16. [PMID: 24865646 DOI: 10.1002/0471140856.tx2502s60] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Measurements of mitochondrial respiration in intact cells can help define metabolism and its dysregulation in fields such as cancer, metabolic disease, immunology, and neurodegeneration. Although cells can be offered various substrates in the assay medium, many cell types can oxidize stored pools of energy substrates. A general bioenergetic profile can therefore be obtained using intact cells, but the inability to control substrate provision to the mitochondria can restrict an in-depth, mechanistic understanding. Mitochondria can be isolated from intact cells, but the yield and quality of the end product is often poor and prone to subselection during isolation. Plasma membrane permeabilization of cells provides a solution to this challenge, allowing experimental control of the medium surrounding the mitochondria. This unit describes techniques to measure respiration in permeabilized adherent cells using a Seahorse XF Analyzer or permeabilized suspended cells in a Hansatech Oxygraph.
Collapse
Affiliation(s)
- Ajit S Divakaruni
- Department of Pharmacology, University of California, San Diego, La Jolla, California
| | | | | |
Collapse
|
309
|
Chin RM, Fu X, Pai MY, Vergnes L, Hwang H, Deng G, Diep S, Lomenick B, Meli VS, Monsalve GC, Hu E, Whelan SA, Wang JX, Jung G, Solis GM, Fazlollahi F, Kaweeteerawat C, Quach A, Nili M, Krall AS, Godwin HA, Chang HR, Faull KF, Guo F, Jiang M, Trauger SA, Saghatelian A, Braas D, Christofk HR, Clarke CF, Teitell MA, Petrascheck M, Reue K, Jung ME, Frand AR, Huang J. The metabolite α-ketoglutarate extends lifespan by inhibiting ATP synthase and TOR. Nature 2014; 510:397-401. [PMID: 24828042 DOI: 10.1038/nature13264] [Citation(s) in RCA: 430] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Accepted: 03/17/2014] [Indexed: 12/11/2022]
Abstract
Metabolism and ageing are intimately linked. Compared with ad libitum feeding, dietary restriction consistently extends lifespan and delays age-related diseases in evolutionarily diverse organisms. Similar conditions of nutrient limitation and genetic or pharmacological perturbations of nutrient or energy metabolism also have longevity benefits. Recently, several metabolites have been identified that modulate ageing; however, the molecular mechanisms underlying this are largely undefined. Here we show that α-ketoglutarate (α-KG), a tricarboxylic acid cycle intermediate, extends the lifespan of adult Caenorhabditis elegans. ATP synthase subunit β is identified as a novel binding protein of α-KG using a small-molecule target identification strategy termed drug affinity responsive target stability (DARTS). The ATP synthase, also known as complex V of the mitochondrial electron transport chain, is the main cellular energy-generating machinery and is highly conserved throughout evolution. Although complete loss of mitochondrial function is detrimental, partial suppression of the electron transport chain has been shown to extend C. elegans lifespan. We show that α-KG inhibits ATP synthase and, similar to ATP synthase knockdown, inhibition by α-KG leads to reduced ATP content, decreased oxygen consumption, and increased autophagy in both C. elegans and mammalian cells. We provide evidence that the lifespan increase by α-KG requires ATP synthase subunit β and is dependent on target of rapamycin (TOR) downstream. Endogenous α-KG levels are increased on starvation and α-KG does not extend the lifespan of dietary-restricted animals, indicating that α-KG is a key metabolite that mediates longevity by dietary restriction. Our analyses uncover new molecular links between a common metabolite, a universal cellular energy generator and dietary restriction in the regulation of organismal lifespan, thus suggesting new strategies for the prevention and treatment of ageing and age-related diseases.
Collapse
Affiliation(s)
- Randall M Chin
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Xudong Fu
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Melody Y Pai
- 1] Molecular Biology Institute, University of California Los Angeles, Los Angeles, California 90095, USA [2]
| | - Laurent Vergnes
- 1] Department of Human Genetics, University of California Los Angeles, Los Angeles, California 90095, USA [2]
| | - Heejun Hwang
- 1] Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California 90095, USA [2]
| | - Gang Deng
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Simon Diep
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Brett Lomenick
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Vijaykumar S Meli
- Department of Biological Chemistry, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Gabriela C Monsalve
- Department of Biological Chemistry, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Eileen Hu
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Stephen A Whelan
- Department of Surgery, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Jennifer X Wang
- Small Molecule Mass Spectrometry Facility, FAS Division of Science, Harvard University, Cambridge, Massachusetts 02138, USA
| | - Gwanghyun Jung
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Gregory M Solis
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Farbod Fazlollahi
- Pasarow Mass Spectrometry Laboratory, Department of Psychiatry and Biobehavioral Sciences and Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Chitrada Kaweeteerawat
- Department of Environmental Health Sciences, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Austin Quach
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Mahta Nili
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Abby S Krall
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Hilary A Godwin
- Department of Environmental Health Sciences, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Helena R Chang
- Department of Surgery, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Kym F Faull
- Pasarow Mass Spectrometry Laboratory, Department of Psychiatry and Biobehavioral Sciences and Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Feng Guo
- Department of Biological Chemistry, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Meisheng Jiang
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Sunia A Trauger
- Small Molecule Mass Spectrometry Facility, FAS Division of Science, Harvard University, Cambridge, Massachusetts 02138, USA
| | - Alan Saghatelian
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, USA
| | - Daniel Braas
- 1] Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California 90095, USA [2] UCLA Metabolomics Center, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Heather R Christofk
- 1] Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California 90095, USA [2] UCLA Metabolomics Center, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Catherine F Clarke
- 1] Molecular Biology Institute, University of California Los Angeles, Los Angeles, California 90095, USA [2] Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Michael A Teitell
- 1] Molecular Biology Institute, University of California Los Angeles, Los Angeles, California 90095, USA [2] Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Michael Petrascheck
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Karen Reue
- 1] Molecular Biology Institute, University of California Los Angeles, Los Angeles, California 90095, USA [2] Department of Human Genetics, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Michael E Jung
- 1] Molecular Biology Institute, University of California Los Angeles, Los Angeles, California 90095, USA [2] Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Alison R Frand
- Department of Biological Chemistry, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Jing Huang
- 1] Molecular Biology Institute, University of California Los Angeles, Los Angeles, California 90095, USA [2] Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California 90095, USA
| |
Collapse
|
310
|
Low abundance of the matrix arm of complex I in mitochondria predicts longevity in mice. Nat Commun 2014; 5:3837. [PMID: 24815183 PMCID: PMC4024759 DOI: 10.1038/ncomms4837] [Citation(s) in RCA: 143] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 04/09/2014] [Indexed: 01/19/2023] Open
Abstract
Mitochondrial function is an important determinant of the ageing process; however, the mitochondrial properties that enable longevity are not well understood. Here we show that optimal assembly of mitochondrial complex I predicts longevity in mice. Using an unbiased high-coverage high-confidence approach, we demonstrate that electron transport chain proteins, especially the matrix arm subunits of complex I, are decreased in young long-living mice, which is associated with improved complex I assembly, higher complex I-linked state 3 oxygen consumption rates and decreased superoxide production, whereas the opposite is seen in old mice. Disruption of complex I assembly reduces oxidative metabolism with concomitant increase in mitochondrial superoxide production. This is rescued by knockdown of the mitochondrial chaperone, prohibitin. Disrupted complex I assembly causes premature senescence in primary cells. We propose that lower abundance of free catalytic complex I components supports complex I assembly, efficacy of substrate utilization and minimal ROS production, enabling enhanced longevity.
Collapse
|
311
|
Diamanti J, Mezzetti B, Giampieri F, Alvarez-Suarez JM, Quiles JL, Gonzalez-Alonso A, Ramirez-Tortosa MDC, Granados-Principal S, Gonzáles-Paramás AM, Santos-Buelga C, Battino M. Doxorubicin-induced oxidative stress in rats is efficiently counteracted by dietary anthocyanin differently enriched strawberry (Fragaria × ananassa Duch.). JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2014; 62:3935-3943. [PMID: 24580025 DOI: 10.1021/jf405721d] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
This study investigated the effects of two different strawberry cultivars, Adria and Sveva, against doxorubicin (DOX)-induced toxicity in rats. A controlled dietary intervention was conducted over 16 weeks with four groups: (i) normal diet; (ii) normal diet + DOX injection; (iii) Adria supplementation + DOX injection; and (iv) Sveva supplementation + DOX injection. Sveva presented higher total antioxidant capacity value and phenol and and vitamin C levels than Adria, which in turn presented higher anthocyanin contents. DOX drastically increased lymphocyte DNA damage, liver biomarkers of protein and lipid oxidation, and mitochondrial ROS content and markedly decreased plasma retinol level, liver antioxidant enzymes, and mitochondrial functionality. After 2 months of strawberry supplementation, rats presented a significant reduction of DNA damage and ROS concentration and a significant improvement of oxidative stress biomarkers, antioxidant enzyme activities, and mitochondrial performance. These results suggest that strawberry supplementation can counteract DOX toxicity, confirming the potential health benefit of strawberry in vivo against oxidative stress.
Collapse
Affiliation(s)
- Jacopo Diamanti
- Department of Agriculture, Food and Environmental Science, Marche Polytechnic University , 60121 Ancona, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
312
|
Jimenez AG, Cooper-Mullin C, Anthony NB, Williams JB. Cellular metabolic rates in cultured primary dermal fibroblasts and myoblast cells from fast-growing and control Coturnix quail. Comp Biochem Physiol A Mol Integr Physiol 2014; 171:23-30. [DOI: 10.1016/j.cbpa.2014.02.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 02/06/2014] [Accepted: 02/06/2014] [Indexed: 12/30/2022]
|
313
|
Stauch KL, Purnell PR, Fox HS. Quantitative proteomics of synaptic and nonsynaptic mitochondria: insights for synaptic mitochondrial vulnerability. J Proteome Res 2014; 13:2620-36. [PMID: 24708184 PMCID: PMC4015687 DOI: 10.1021/pr500295n] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Synaptic mitochondria are essential for maintaining calcium homeostasis and producing ATP, processes vital for neuronal integrity and synaptic transmission. Synaptic mitochondria exhibit increased oxidative damage during aging and are more vulnerable to calcium insult than nonsynaptic mitochondria. Why synaptic mitochondria are specifically more susceptible to cumulative damage remains to be determined. In this study, the generation of a super-SILAC mix that served as an appropriate internal standard for mouse brain mitochondria mass spectrometry based analysis allowed for the quantification of the proteomic differences between synaptic and nonsynaptic mitochondria isolated from 10-month-old mice. We identified a total of 2260 common proteins between synaptic and nonsynaptic mitochondria of which 1629 were annotated as mitochondrial. Quantitative proteomic analysis of the proteins common between synaptic and nonsynaptic mitochondria revealed significant differential expression of 522 proteins involved in several pathways including oxidative phosphorylation, mitochondrial fission/fusion, calcium transport, and mitochondrial DNA replication and maintenance. In comparison to nonsynaptic mitochondria, synaptic mitochondria exhibited increased age-associated mitochondrial DNA deletions and decreased bioenergetic function. These findings provide insights into synaptic mitochondrial susceptibility to damage.
Collapse
Affiliation(s)
- Kelly L Stauch
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center , 985800 Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | | | | |
Collapse
|
314
|
Mailloux RJ, Xuan JY, McBride S, Maharsy W, Thorn S, Holterman CE, Kennedy CRJ, Rippstein P, deKemp R, da Silva J, Nemer M, Lou M, Harper ME. Glutaredoxin-2 is required to control oxidative phosphorylation in cardiac muscle by mediating deglutathionylation reactions. J Biol Chem 2014; 289:14812-28. [PMID: 24727547 DOI: 10.1074/jbc.m114.550574] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Glutaredoxin-2 (Grx2) modulates the activity of several mitochondrial proteins in cardiac tissue by catalyzing deglutathionylation reactions. However, it remains uncertain whether Grx2 is required to control mitochondrial ATP output in heart. Here, we report that Grx2 plays a vital role modulating mitochondrial energetics and heart physiology by mediating the deglutathionylation of mitochondrial proteins. Deletion of Grx2 (Grx2(-/-)) decreased ATP production by complex I-linked substrates to half that in wild type (WT) mitochondria. Decreased respiration was associated with increased complex I glutathionylation diminishing its activity. Tissue glucose uptake was concomitantly increased. Mitochondrial ATP output and complex I activity could be recovered by restoring the redox environment to that favoring the deglutathionylated states of proteins. Grx2(-/-) hearts also developed left ventricular hypertrophy and fibrosis, and mice became hypertensive. Mitochondrial energetics from Grx2 heterozygotes (Grx2(+/-)) were also dysfunctional, and hearts were hypertrophic. Intriguingly, Grx2(+/-) mice were far less hypertensive than Grx2(-/-) mice. Thus, Grx2 plays a vital role in modulating mitochondrial metabolism in cardiac muscle, and Grx2 deficiency leads to pathology. As mitochondrial ATP production was restored by the addition of reductants, these findings may be relevant to novel redox-related therapies in cardiac disease.
Collapse
Affiliation(s)
- Ryan J Mailloux
- From the Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Jian Ying Xuan
- From the Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Skye McBride
- From the Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Wael Maharsy
- From the Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Stephanie Thorn
- the University of Ottawa Heart Institute, Ottawa, Ontario K1Y 4W7, Canada
| | - Chet E Holterman
- the Kidney Research Centre, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, Ontario K1H 8L6, Canada, and
| | - Christopher R J Kennedy
- the Kidney Research Centre, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, Ontario K1H 8L6, Canada, and
| | - Peter Rippstein
- the University of Ottawa Heart Institute, Ottawa, Ontario K1Y 4W7, Canada
| | - Robert deKemp
- the University of Ottawa Heart Institute, Ottawa, Ontario K1Y 4W7, Canada
| | - Jean da Silva
- the University of Ottawa Heart Institute, Ottawa, Ontario K1Y 4W7, Canada
| | - Mona Nemer
- From the Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Marjorie Lou
- the Center of Redox Biology and School of Veterinary Medicine and Biomedical Sciences, University of Nebraska at Lincoln, Lincoln, Nebraska 68583-0903
| | - Mary-Ellen Harper
- From the Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada,
| |
Collapse
|
315
|
Khiati S, Dalla Rosa I, Sourbier C, Ma X, Rao VA, Neckers LM, Zhang H, Pommier Y. Mitochondrial topoisomerase I (top1mt) is a novel limiting factor of doxorubicin cardiotoxicity. Clin Cancer Res 2014; 20:4873-81. [PMID: 24714774 DOI: 10.1158/1078-0432.ccr-13-3373] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Doxorubicin is one of the most effective chemotherapeutic agents. However, up to 30% of the patients treated with doxorubicin suffer from congestive heart failure. The mechanism of doxorubicin cardiotoxicity is likely multifactorial and most importantly, the genetic factors predisposing to doxorubicin cardiotoxicity are unknown. On the basis of the fact that mtDNA lesions and mitochondrial dysfunctions have been found in human hearts exposed to doxorubicin and that mitochondrial topoisomerase 1 (Top1mt) specifically controls mtDNA homeostasis, we hypothesized that Top1mt knockout (KO) mice might exhibit hypersensitivity to doxorubicin. EXPERIMENTAL DESIGN Wild-type (WT) and KO Top1mt mice were treated once a week with 4 mg/kg doxorubicin for 8 weeks. Heart tissues were analyzed one week after the last treatment. RESULTS Genetic inactivation of Top1mt in mice accentuates mtDNA copy number loss and mtDNA damage in heart tissue following doxorubicin treatment. Top1mt KO mice also fail to maintain respiratory chain protein production and mitochondrial cristae ultrastructure organization. These mitochondrial defects result in decreased O2 consumption, increased reactive oxygen species production, and enhanced heart muscle damage in animals treated with doxorubicin. Accordingly, Top1mt KO mice die within 45 days after the last doxorubicin injection, whereas the WT mice survive. CONCLUSIONS Our results provide evidence that Top1mt, which is conserved across vertebrates, is critical for cardiac tolerance to doxorubicin and adaptive response to doxorubicin cardiotoxicity. They also suggest the potential of Top1mt single-nucleotide polymorphisms testing to investigate patient susceptibility to doxorubicin-induced cardiotoxicity.
Collapse
Affiliation(s)
- Salim Khiati
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology
| | - Ilaria Dalla Rosa
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology
| | - Carole Sourbier
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute
| | - Xuefei Ma
- Laboratory of Molecular Cardiology, National Heart, Lung, and Blood Institute, NIH; and
| | - V Ashutosh Rao
- Center for Drug Evaluation and Research, Food and Drug Administration, Bethesda, Maryland
| | - Leonard M Neckers
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute
| | - Hongliang Zhang
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology
| | - Yves Pommier
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology;
| |
Collapse
|
316
|
Sun K, Kusminski CM, Luby-Phelps K, Spurgin SB, An YA, Wang QA, Holland WL, Scherer PE. Brown adipose tissue derived VEGF-A modulates cold tolerance and energy expenditure. Mol Metab 2014; 3:474-83. [PMID: 24944907 PMCID: PMC4060212 DOI: 10.1016/j.molmet.2014.03.010] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 03/23/2014] [Accepted: 03/24/2014] [Indexed: 12/30/2022] Open
Abstract
We recently reported that local overexpression of VEGF-A in white adipose tissue (WAT) protects against diet-induced obesity and metabolic dysfunction. The observation that VEGF-A induces a “brown adipose tissue (BAT)-like” phenotype in WAT prompted us to further explore the direct function of VEGF-A in BAT. We utilized a doxycycline (Dox)-inducible, brown adipocyte-specific VEGF-A transgenic overexpression model to assess direct effects of VEGF-A in BAT in vivo. We observed that BAT-specific VEGF-A expression increases vascularization and up-regulates expression of both UCP1 and PGC-1α in BAT. As a result, the transgenic mice show increased thermogenesis during chronic cold exposure. In diet-induced obese mice, introducing VEGF-A locally in BAT rescues capillary rarefaction, ameliorates brown adipocyte dysfunction, and improves deleterious effects on glucose and lipid metabolism caused by a high-fat diet challenge. These results demonstrate a direct positive role of VEGF-A in the activation and expansion of BAT.
Collapse
Affiliation(s)
- Kai Sun
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Christine M. Kusminski
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Kate Luby-Phelps
- Department of Cell Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Stephen B. Spurgin
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Yu A. An
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Qiong A. Wang
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - William L. Holland
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Philipp E. Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
- Department of Cell Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
- Corresponding author. Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-8549, USA. Tel.: +1 214 648 8715; fax: +1 214 648 8720.
| |
Collapse
|
317
|
Šileikytė J, Blachly-Dyson E, Sewell R, Carpi A, Menabò R, Di Lisa F, Ricchelli F, Bernardi P, Forte M. Regulation of the mitochondrial permeability transition pore by the outer membrane does not involve the peripheral benzodiazepine receptor (Translocator Protein of 18 kDa (TSPO)). J Biol Chem 2014; 289:13769-81. [PMID: 24692541 DOI: 10.1074/jbc.m114.549634] [Citation(s) in RCA: 162] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Translocator protein of 18 kDa (TSPO) is a highly conserved, ubiquitous protein localized in the outer mitochondrial membrane, where it is thought to play a key role in the mitochondrial transport of cholesterol, a key step in the generation of steroid hormones. However, it was first characterized as the peripheral benzodiazepine receptor because it appears to be responsible for high affinity binding of a number of benzodiazepines to non-neuronal tissues. Ensuing studies have employed natural and synthetic ligands to assess the role of TSPO function in a number of natural and pathological circumstances. Largely through the use of these compounds and biochemical associations, TSPO has been proposed to play a role in the mitochondrial permeability transition pore (PTP), which has been associated with cell death in many human pathological conditions. Here, we critically assess the role of TSPO in the function of the PTP through the generation of mice in which the Tspo gene has been conditionally eliminated. Our results show that 1) TSPO plays no role in the regulation or structure of the PTP, 2) endogenous and synthetic ligands of TSPO do not regulate PTP activity through TSPO, 3) outer mitochondrial membrane regulation of PTP activity occurs though a mechanism that does not require TSPO, and 4) hearts lacking TSPO are as sensitive to ischemia-reperfusion injury as hearts from control mice. These results call into question a wide variety of studies implicating TSPO in a number of pathological processes through its actions on the PTP.
Collapse
Affiliation(s)
- Justina Šileikytė
- From the Consiglio Nazionale delle Ricerche Institute of Neuroscience and Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, I-35121 Padova, Italy
| | | | - Randall Sewell
- the Vollum Institute, Oregon Health and Sciences University, Portland, Oregon 97239, and
| | - Andrea Carpi
- From the Consiglio Nazionale delle Ricerche Institute of Neuroscience and Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, I-35121 Padova, Italy
| | - Roberta Menabò
- From the Consiglio Nazionale delle Ricerche Institute of Neuroscience and Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, I-35121 Padova, Italy
| | - Fabio Di Lisa
- From the Consiglio Nazionale delle Ricerche Institute of Neuroscience and Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, I-35121 Padova, Italy
| | - Fernanda Ricchelli
- the Consiglio Nazionale delle Ricerche Institute of Biomedical Technologies at the Department of Biology, University of Padova, I-35121 Padova, Italy
| | - Paolo Bernardi
- From the Consiglio Nazionale delle Ricerche Institute of Neuroscience and Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, I-35121 Padova, Italy,
| | - Michael Forte
- the Vollum Institute, Oregon Health and Sciences University, Portland, Oregon 97239, and
| |
Collapse
|
318
|
Mishra P, Carelli V, Manfredi G, Chan DC. Proteolytic cleavage of Opa1 stimulates mitochondrial inner membrane fusion and couples fusion to oxidative phosphorylation. Cell Metab 2014; 19:630-41. [PMID: 24703695 PMCID: PMC4018240 DOI: 10.1016/j.cmet.2014.03.011] [Citation(s) in RCA: 342] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 12/03/2013] [Accepted: 01/27/2014] [Indexed: 12/29/2022]
Abstract
Mitochondrial fusion is essential for maintenance of mitochondrial function. The mitofusin GTPases control mitochondrial outer membrane fusion, whereas the dynamin-related GTPase Opa1 mediates inner membrane fusion. We show that mitochondrial inner membrane fusion is tuned by the level of oxidative phosphorylation (OXPHOS), whereas outer membrane fusion is insensitive. Consequently, cells from patients with pathogenic mtDNA mutations show a selective defect in mitochondrial inner membrane fusion. In elucidating the molecular mechanism of OXPHOS-stimulated fusion, we uncover that real-time proteolytic processing of Opa1 stimulates mitochondrial inner membrane fusion. OXPHOS-stimulated mitochondrial fusion operates through Yme1L, which cleaves Opa1 more efficiently under high OXPHOS conditions. Engineered cleavage of Opa1 is sufficient to mediate inner membrane fusion, regardless of respiratory state. Proteolytic cleavage therefore stimulates the membrane fusion activity of Opa1, and this feature is exploited to dynamically couple mitochondrial fusion to cellular metabolism.
Collapse
Affiliation(s)
- Prashant Mishra
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Valerio Carelli
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Department of Biomedical and NeuroMotor Sciences, University of Bologna, Via Ugo Foscolo 7, 40123 Bologna, Italy
| | - Giovanni Manfredi
- Departments of Neurology and Neuroscience, Weill Medical College of Cornell University, 1300 York Avenue, A501, New York, NY 10065, USA
| | - David C Chan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA; Howard Hughes Medical Institute, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
319
|
Ruiz LM, Jensen EL, Bustos RI, Argüelloa G, Gutierrez-Garcia R, González M, Hernández C, Paredes R, Simon F, Riedel C, Ferrick D, Elorza AA. Adaptive responses of mitochondria to mild copper deprivation involve changes in morphology, OXPHOS remodeling and bioenergetics. J Cell Physiol 2014; 229:607-19. [PMID: 24446197 DOI: 10.1002/jcp.24484] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 09/30/2013] [Indexed: 12/23/2022]
Abstract
Copper is an essential cofactor of complex IV of the electron transfer chain, and it is directly involved in the generation of mitochondrial membrane potential. Its deficiency induces the formation of ROS, large mitochondria and anemia. Thus, there is a connection between copper metabolism and bioenergetics, mitochondrial dynamics and erythropoiesis. Copper depletion might end in cellular apoptosis or necrosis. However, before entering into those irreversible processes, mitochondria may execute a series of adaptive responses. Mitochondrial adaptive responses (MAR) may involve multiple and diverse mechanisms for preserving cell life, such as mitochondrial dynamics, OXPHOS remodeling and bioenergetics output. In this study, a mild copper deficiency was produced in an animal model through intraperitoneal injections of bathocuproine disulfonate in order to study the MAR. Under these conditions, a new type of mitochondrial morphology was discovered in the liver. Termed the "butternut squash" mitochondria, it coexisted with normal and swollen mitochondria. Western blot analyses of mitochondrial dynamics proteins showed an up-regulation of MFN-2 and OPA1 fusion proteins. Furthermore, isolated liver mitochondria displayed OXPHOS remodeling through a decrease in supercomplex activity with a concomitant increase at an individual level of complexes I and IV, higher respiratory rates at complex I and II levels, higher oligomycin-insensitive respiration, and lower respiratory control ratio values when compared to the control group. As expected, total ATP and ATP/ADP values were not significantly different, since animal's health was not compromised. As a whole, these results describe a compensatory and adaptive response of metabolism and bioenergetics under copper deprivation.
Collapse
Affiliation(s)
- Lina María Ruiz
- Center for Biomedical Research, Faculty of Biological Sciences and Faculty of Medicine, Universidad Andres Bello, Santiago, Chile
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
320
|
Luo J, Abdallah BG, Wolken GG, Arriaga EA, Ros A. Insulator-based dielectrophoresis of mitochondria. BIOMICROFLUIDICS 2014; 8:021801. [PMID: 24959306 PMCID: PMC4056684 DOI: 10.1063/1.4866852] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 01/28/2014] [Indexed: 05/03/2023]
Abstract
Isolated mitochondria display a wide range of sizes plausibly resulting from the coexistence of subpopulations, some of which may be associated with disease or aging. Strategies to separate subpopulations are needed to study the importance of these organelles in cellular functions. Here, insulator-based dielectrophoresis (iDEP) was exploited to provide a new dimension of organelle separation. The dielectrophoretic properties of isolated Fischer 344 (F344) rat semimembranosus muscle mitochondria and C57BL/6 mouse hepatic mitochondria in low conductivity buffer (0.025-0.030 S/m) at physiological pH (7.2-7.4) were studied using polydimethylsiloxane (PDMS) microfluidic devices. First, direct current (DC) and alternating current (AC) of 0-50 kHz with potentials of 0-3000 V applied over a channel length of 1 cm were separately employed to generate inhomogeneous electric fields and establish that mitochondria exhibit negative DEP (nDEP). DEP trapping potential thresholds at 0-50 kHz were also determined to be weakly dependent on applied frequency and were generally above 200 V. Second, we demonstrated a separation scheme using DC potentials <100 V to perform the first size-based iDEP sorting of mitochondria. Samples of isolated mitochondria with heterogeneous sizes (150 nm-2 μm diameters) were successfully separated into sub-micron fractions, indicating the ability to isolate mitochondria into populations based on their size.
Collapse
Affiliation(s)
- Jinghui Luo
- Department of Chemistry and Biochemistry, Arizona State University, Tempe, Arizona 85287, USA
| | - Bahige G Abdallah
- Department of Chemistry and Biochemistry, Arizona State University, Tempe, Arizona 85287, USA
| | - Gregory G Wolken
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Edgar A Arriaga
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Alexandra Ros
- Department of Chemistry and Biochemistry, Arizona State University, Tempe, Arizona 85287, USA
| |
Collapse
|
321
|
Sridharan V, Aykin-Burns N, Tripathi P, Krager KJ, Sharma SK, Moros EG, Corry PM, Nowak G, Hauer-Jensen M, Boerma M. Radiation-induced alterations in mitochondria of the rat heart. Radiat Res 2014; 181:324-34. [PMID: 24568130 PMCID: PMC4029615 DOI: 10.1667/rr13452.1] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Radiation therapy for the treatment of thoracic cancers may be associated with radiation-induced heart disease (RIHD), especially in long-term cancer survivors. Mechanisms by which radiation causes heart disease are largely unknown. To identify potential long-term contributions of mitochondria in the development of radiation-induced heart disease, we examined the time course of effects of irradiation on cardiac mitochondria. In this study, Sprague-Dawley male rats received image-guided local X irradiation of the heart with a single dose ranging from 3-21 Gy. Two weeks after irradiation, left ventricular mitochondria were isolated to assess the dose-dependency of the mitochondrial permeability transition pore (mPTP) opening in a mitochondrial swelling assay. At time points from 6 h to 9 months after a cardiac dose of 21 Gy, the following analyses were performed: left ventricular Bax and Bcl-2 protein levels; apoptosis; mitochondrial inner membrane potential and mPTP opening; mitochondrial mass and expression of mitophagy mediators Parkin and PTEN induced putative kinase-1 (PINK-1); mitochondrial respiration and protein levels of succinate dehydrogenase A (SDHA); and the 70 kDa subunit of complex II. Local heart irradiation caused a prolonged increase in Bax/Bcl-2 ratio and induced apoptosis between 6 h and 2 weeks. The mitochondrial membrane potential was reduced until 2 weeks, and the calcium-induced mPTP opening was increased from 6 h up to 9 months. An increased mitochondrial mass together with unaltered levels of Parkin suggested that mitophagy did not occur. Lastly, we detected a significant decrease in succinate-driven state 2 respiration in isolated mitochondria from 2 weeks up to 9 months after irradiation, coinciding with reduced mitochondrial levels of succinate dehydrogenase A. Our results suggest that local heart irradiation induces long-term changes in cardiac mitochondrial membrane functions, levels of SDH and state 2 respiration. At any time after exposure to radiation, cardiac mitochondria are more prone to mPTP opening. Future studies will determine whether this makes the heart more susceptible to secondary stressors such as calcium overload or ischemia/reperfusion.
Collapse
Affiliation(s)
- Vijayalakshmi Sridharan
- University of Arkansas for Medical Sciences, Department of Pharmaceutical Sciences, Division of Radiation Health, Little Rock, Arkansas
| | - Nukhet Aykin-Burns
- University of Arkansas for Medical Sciences, Department of Pharmaceutical Sciences, Division of Radiation Health, Little Rock, Arkansas
| | - Preeti Tripathi
- University of Arkansas for Medical Sciences, Department of Pharmaceutical Sciences, Division of Radiation Health, Little Rock, Arkansas
| | - Kimberly J. Krager
- University of Arkansas for Medical Sciences, Department of Pharmaceutical Sciences, Division of Radiation Health, Little Rock, Arkansas
| | - Sunil K. Sharma
- University of Arkansas for Medical Sciences, Department of Radiation Oncology, Little Rock, Arkansas
| | - Eduardo G. Moros
- Moffitt Cancer Center and Research Institute, Department of Radiation Oncology, Tampa, Florida
| | - Peter M. Corry
- University of Arkansas for Medical Sciences, Department of Radiation Oncology, Little Rock, Arkansas
| | - Grazyna Nowak
- University of Arkansas for Medical Sciences, Department of Pharmaceutical Sciences, Little Rock, Arkansas
| | - Martin Hauer-Jensen
- University of Arkansas for Medical Sciences, Department of Pharmaceutical Sciences, Division of Radiation Health, Little Rock, Arkansas
- Surgical Service, Central Arkansas Veterans Healthcare System, Little Rock, Arkansas
| | - Marjan Boerma
- University of Arkansas for Medical Sciences, Department of Pharmaceutical Sciences, Division of Radiation Health, Little Rock, Arkansas
| |
Collapse
|
322
|
Novel inhibitors of mitochondrial sn-glycerol 3-phosphate dehydrogenase. PLoS One 2014; 9:e89938. [PMID: 24587137 PMCID: PMC3933693 DOI: 10.1371/journal.pone.0089938] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 01/28/2014] [Indexed: 11/19/2022] Open
Abstract
Mitochondrial sn-glycerol 3-phosphate dehydrogenase (mGPDH) is a ubiquinone-linked enzyme in the mitochondrial inner membrane best characterized as part of the glycerol phosphate shuttle that transfers reducing equivalents from cytosolic NADH into the mitochondrial electron transport chain. Despite the widespread expression of mGPDH and the availability of mGPDH-null mice, the physiological role of this enzyme remains poorly defined in many tissues, likely because of compensatory pathways for cytosolic regeneration of NAD+ and mechanisms for glycerol phosphate metabolism. Here we describe a novel class of cell-permeant small-molecule inhibitors of mGPDH (iGP) discovered through small-molecule screening. Structure-activity analysis identified a core benzimidazole-phenyl-succinamide structure as being essential to inhibition of mGPDH while modifications to the benzimidazole ring system modulated both potency and off-target effects. Live-cell imaging provided evidence that iGPs penetrate cellular membranes. Two compounds (iGP-1 and iGP-5) were characterized further to determine potency and selectivity and found to be mixed inhibitors with IC50 and Ki values between ∼1–15 µM. These novel mGPDH inhibitors are unique tools to investigate the role of glycerol 3-phosphate metabolism in both isolated and intact systems.
Collapse
|
323
|
Baltgalvis KA, White K, Li W, Claypool MD, Lang W, Alcantara R, Singh BK, Friera AM, McLaughlin J, Hansen D, McCaughey K, Nguyen H, Smith IJ, Godinez G, Shaw SJ, Goff D, Singh R, Markovtsov V, Sun TQ, Jenkins Y, Uy G, Li Y, Pan A, Gururaja T, Lau D, Park G, Hitoshi Y, Payan DG, Kinsella TM. Exercise performance and peripheral vascular insufficiency improve with AMPK activation in high-fat diet-fed mice. Am J Physiol Heart Circ Physiol 2014; 306:H1128-45. [PMID: 24561866 DOI: 10.1152/ajpheart.00839.2013] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Intermittent claudication is a form of exercise intolerance characterized by muscle pain during walking in patients with peripheral artery disease (PAD). Endothelial cell and muscle dysfunction are thought to be important contributors to the etiology of this disease, but a lack of preclinical models that incorporate these elements and measure exercise performance as a primary end point has slowed progress in finding new treatment options for these patients. We sought to develop an animal model of peripheral vascular insufficiency in which microvascular dysfunction and exercise intolerance were defining features. We further set out to determine if pharmacological activation of 5'-AMP-activated protein kinase (AMPK) might counteract any of these functional deficits. Mice aged on a high-fat diet demonstrate many functional and molecular characteristics of PAD, including the sequential development of peripheral vascular insufficiency, increased muscle fatigability, and progressive exercise intolerance. These changes occur gradually and are associated with alterations in nitric oxide bioavailability. Treatment of animals with an AMPK activator, R118, increased voluntary wheel running activity, decreased muscle fatigability, and prevented the progressive decrease in treadmill exercise capacity. These functional performance benefits were accompanied by improved mitochondrial function, the normalization of perfusion in exercising muscle, increased nitric oxide bioavailability, and decreased circulating levels of the endogenous endothelial nitric oxide synthase inhibitor asymmetric dimethylarginine. These data suggest that aged, obese mice represent a novel model for studying exercise intolerance associated with peripheral vascular insufficiency, and pharmacological activation of AMPK may be a suitable treatment for intermittent claudication associated with PAD.
Collapse
|
324
|
Brite/beige fat and UCP1 - is it thermogenesis? BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2014; 1837:1075-82. [PMID: 24530356 DOI: 10.1016/j.bbabio.2014.02.008] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 01/22/2014] [Accepted: 02/10/2014] [Indexed: 12/17/2022]
Abstract
The presence of two distinct types of adipose tissue, which have opposing functions, has been known for decades. White adipose tissue (WAT) is the main tissue of energy storage, while brown adipose tissue (BAT) dissipates energy as heat and is required for non-shivering thermoregulation. In the last few years, a third type of adipocyte was identified, termed the brite ("brown and white") or beige adipocyte. Their physiological control and role, however, are not fully clarified. Brite/beige adipocytes have a positive impact on systemic metabolism that is generally explained by the thermogenesis of brite/beige adipocytes; although thermogenesis has not been directly measured but is mostly inferred by gene expression data of typical thermogenic genes such as uncoupling protein 1 (UCP1). Here we critically review functional evidence for the thermogenic potential of brite/beige adipocytes, leading to the conclusion that direct measurements of brite/beige adipocyte bioenergetics, beyond gene regulation, are pivotal to quantify their thermogenic potential. In particular, we exemplified that the massive induction of UCP1 mRNA during the browning of isolated subcutaneous adipocytes in vitro is not reflected in significant alterations of cellular bioenergetics. Herein, we demonstrate that increases in mitochondrial respiration in response to beta-adrenergic stimulus can be independent of UCP1. Using HEK293 cells expressing UCP1, we show how to directly assess UCP1 function by adequate activation in intact cells. Finally, we provide a guide on the interpretation of UCP1 activity and the pitfalls by solely using respiration measurements. The functional analysis of beige adipocyte bioenergetics will assist to delineate the impact of browning on thermogenesis, possibly elucidating additional physiological roles and its contribution to systemic metabolism, highlighting possible avenues for future research. This article is part of a Special Issue entitled: 18th European Bioenergetic Conference.
Collapse
|
325
|
Mitochondrial oxygen consumption deficits in skeletal muscle isolated from an Alzheimer's disease-relevant murine model. BMC Neurosci 2014; 15:24. [PMID: 24524276 PMCID: PMC3930757 DOI: 10.1186/1471-2202-15-24] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 02/04/2014] [Indexed: 11/10/2022] Open
Abstract
Background Age is considered a primary risk factor for neurodegenerative diseases including Alzheimer’s disease (AD). It is also now well understood that mitochondrial function declines with age. Mitochondrial deficits have been previously assessed in brain from both human autopsy tissue and disease-relevant transgenic mice. Recently it has been recognized that abnormalities of muscle may be an intrinsic aspect of AD and might contribute to the pathophysiology. However, deficits in mitochondrial function have yet to be clearly assessed in tissues outside the central nervous system (CNS). In the present study, we utilized a well-characterized AD-relevant transgenic mouse strain to assess mitochondrial respiratory deficits in both brain and muscle. In addition to mitochondrial function, we assessed levels of transgene-derived amyloid precursor protein (APP) in homogenates isolated from brain and muscle of these AD-relevant animals. Results We now demonstrate that skeletal muscles isolated from these animals have differential levels of mutant full-length APP depending on muscle type. Additionally, isolated muscle fibers from young transgenic mice (3 months) have significantly decreased maximal mitochondrial oxygen consumption capacity compared to non-transgenic, age-matched mice, with similar deficits to those previously described in brain. Conclusions This is the first study to directly examine mitochondrial function in skeletal muscle from an AD-relevant transgenic murine model. As with brain, these deficits in muscle are an early event, occurring prior to appearance of amyloid plaques.
Collapse
|
326
|
Liu K, Ji K, Guo L, Wu W, Lu H, Shan P, Yan C. Mesenchymal stem cells rescue injured endothelial cells in an in vitro ischemia-reperfusion model via tunneling nanotube like structure-mediated mitochondrial transfer. Microvasc Res 2014; 92:10-8. [PMID: 24486322 DOI: 10.1016/j.mvr.2014.01.008] [Citation(s) in RCA: 257] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Revised: 01/22/2014] [Accepted: 01/22/2014] [Indexed: 12/31/2022]
Abstract
Mesenchymal stem cells can be used as a novel treatment of ischemic vascular disease; however, their therapeutic effect and mechanism of action require further evaluation. Mitochondrial dysfunction has core functions in ischemia-reperfusion injury of the microvascular network. A recent discovery has shown that intercellular communication using tunneling nanotubes can transfer mitochondria between adjacent cells. This study aimed to investigate the tunneling nanotube mechanisms that might be involved in stem cell-mediated mitochondrial rescue of injured vascular endothelial cells. Using laser scanning confocal microscopy, mitochondrial transfer via a tunneling nanotube-like structure was detected between mesenchymal stem cells and human umbilical vein endothelial cells. Oxygen glucose deprivation and reoxygenation were performed on human umbilical vein endothelial cells, which induced mitochondrial transfer through tunneling nanotube-like structures to become frequent and almost unidirectional from mesenchymal stem cells to injured endothelial cells, thereby resulting in the rescue of aerobic respiration and protection of endothelial cells from apoptosis. We found that the formation of tunneling nanotube-like structures might represent a defense and rescue mechanism through phosphatidylserines exposed on the surface of apoptotic endothelial cells and stem cell recognition. Our data provided evidence that stem cells can rescue damaged vascular endothelial cells through a mechanism that has not yet been identified.
Collapse
Affiliation(s)
- Kaiming Liu
- Department of Geriatrics, Qilu Hospital of Shandong University, Jinan, Shandong, China; Department of Neurology, Qilu Hospital of Shandong University, Jinan, Shandong, China; Key Laboratory for Experimental Teratology of the Ministry of Education, Brain Science Research Institute, Shandong University, Jinan, Shandong, China
| | - Kunqian Ji
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, Shandong, China; Key Laboratory for Experimental Teratology of the Ministry of Education, Brain Science Research Institute, Shandong University, Jinan, Shandong, China
| | - Liang Guo
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Wei Wu
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Huixia Lu
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Peiyan Shan
- Department of Geriatrics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Chuanzhu Yan
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, Shandong, China; Key Laboratory for Experimental Teratology of the Ministry of Education, Brain Science Research Institute, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
327
|
Jimenez AG, Van Brocklyn J, Wortman M, Williams JB. Cellular metabolic rate is influenced by life-history traits in tropical and temperate birds. PLoS One 2014; 9:e87349. [PMID: 24498080 PMCID: PMC3907555 DOI: 10.1371/journal.pone.0087349] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 12/19/2013] [Indexed: 11/20/2022] Open
Abstract
In general, tropical birds have a “slow pace of life,” lower rates of whole-animal metabolism and higher survival rates, than temperate species. A fundamental challenge facing physiological ecologists is the understanding of how variation in life-history at the whole-organism level might be linked to cellular function. Because tropical birds have lower rates of whole-animal metabolism, we hypothesized that cells from tropical species would also have lower rates of cellular metabolism than cells from temperate species of similar body size and common phylogenetic history. We cultured primary dermal fibroblasts from 17 tropical and 17 temperate phylogenetically-paired species of birds in a common nutritive and thermal environment and then examined basal, uncoupled, and non-mitochondrial cellular O2 consumption (OCR), proton leak, and anaerobic glycolysis (extracellular acidification rates [ECAR]), using an XF24 Seahorse Analyzer. We found that multiple measures of metabolism in cells from tropical birds were significantly lower than their temperate counterparts. Basal and uncoupled cellular metabolism were 29% and 35% lower in cells from tropical birds, respectively, a decrease closely aligned with differences in whole-animal metabolism between tropical and temperate birds. Proton leak was significantly lower in cells from tropical birds compared with cells from temperate birds. Our results offer compelling evidence that whole-animal metabolism is linked to cellular respiration as a function of an animal’s life-history evolution. These findings are consistent with the idea that natural selection has uniquely fashioned cells of long-lived tropical bird species to have lower rates of metabolism than cells from shorter-lived temperate species.
Collapse
Affiliation(s)
- Ana Gabriela Jimenez
- Department of Evolution, Ecology and Organismal Biology, Ohio State University, Columbus, Ohio, United States of America
- * E-mail:
| | - James Van Brocklyn
- Department of Evolution, Ecology and Organismal Biology, Ohio State University, Columbus, Ohio, United States of America
| | - Matthew Wortman
- Cancer Institute, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Joseph B. Williams
- Department of Evolution, Ecology and Organismal Biology, Ohio State University, Columbus, Ohio, United States of America
| |
Collapse
|
328
|
Salabei JK, Gibb AA, Hill BG. Comprehensive measurement of respiratory activity in permeabilized cells using extracellular flux analysis. Nat Protoc 2014; 9:421-38. [PMID: 24457333 DOI: 10.1038/nprot.2014.018] [Citation(s) in RCA: 233] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Extracellular flux (XF) analysis has become a mainstream method for measuring mitochondrial function in cells and tissues. Although this technique is commonly used to measure bioenergetics in intact cells, we outline here a detailed XF protocol for measuring respiration in permeabilized cells. Cells are permeabilized using saponin (SAP), digitonin (DIG) or recombinant perfringolysin O (rPFO) (XF-plasma membrane permeabilizer (PMP) reagent), and they are provided with specific substrates to measure complex I- or complex II-mediated respiratory activity, complex III+IV respiratory activity or complex IV activity. Medium- and long-chain acylcarnitines or glutamine may also be provided for measuring fatty acid (FA) oxidation or glutamine oxidation, respectively. This protocol uses a minimal number of cells compared with other protocols and does not require isolation of mitochondria. The results are highly reproducible, and mitochondria remain well coupled. Collectively, this protocol provides comprehensive and detailed information regarding mitochondrial activity and efficiency, and, after preparative steps, it takes 6-8 h to complete.
Collapse
Affiliation(s)
- Joshua K Salabei
- Diabetes and Obesity Center, Institute of Molecular Cardiology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Andrew A Gibb
- 1] Diabetes and Obesity Center, Institute of Molecular Cardiology, University of Louisville School of Medicine, Louisville, Kentucky, USA. [2] Department of Physiology and Biophysics, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Bradford G Hill
- 1] Diabetes and Obesity Center, Institute of Molecular Cardiology, University of Louisville School of Medicine, Louisville, Kentucky, USA. [2] Department of Physiology and Biophysics, University of Louisville School of Medicine, Louisville, Kentucky, USA. [3] Department of Biochemistry and Molecular Biology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| |
Collapse
|
329
|
Kim C, Patel P, Gouvin LM, Brown ML, Khalil A, Henchey EM, Heuck AP, Yadava N. Comparative Analysis of the Mitochondrial Physiology of Pancreatic β Cells. ACTA ACUST UNITED AC 2014; 3:110. [PMID: 25309834 DOI: 10.4172/2167-7662.1000110] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The mitochondrial metabolism of β cells is thought to be highly specialized. Its direct comparison with other cells using isolated mitochondria is limited by the availability of islets/β cells in sufficient quantity. In this study, we have compared mitochondrial metabolism of INS1E/β cells with other cells in intact and permeabilized states. To selectively permeabilize the plasma membrane, we have evaluated the use of perfringolysin-O (PFO) in conjunction with microplate-based respirometry. PFO is a protein that binds membranes based on a threshold level of active cholesterol. Therefore, unless active cholesterol reaches a threshold level in mitochondria, they are expected to remain untouched by PFO. Cytochrome c sensitivity tests showed that in PFO-permeabilized cells, the mitochondrial integrity was completely preserved. Our data show that a time-dependent decline of the oligomycin-insensitive respiration observed in INS1E cells was due to a limitation in substrate supply to the respiratory chain. We predict that it is linked with the β cell-specific metabolism involving metabolites shuttling between the cytoplasm and mitochondria. In permeabilized β cells, the Complex l-dependent respiration was either transient or absent because of the inefficient TCA cycle. The TCA cycle insufficiency was confirmed by analysis of the CO2 evolution. This may be linked with lower levels of NAD+, which is required as a co-factor for CO2 producing reactions of the TCA cycle. β cells showed comparable OxPhos and respiratory capacities that were not affected by the inorganic phosphate (Pi) levels in the respiration medium. They showed lower ADP-stimulation of the respiration on different substrates. We believe that this study will significantly enhance our understanding of the β cell mitochondrial metabolism.
Collapse
Affiliation(s)
- Chul Kim
- Pioneer Valley Life Sciences Institute, Springfield, MA, USA
| | - Pinal Patel
- Pioneer Valley Life Sciences Institute, Springfield, MA, USA
| | - Lindsey M Gouvin
- Departments of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, MA, USA
| | - Melissa L Brown
- Pioneer Valley Life Sciences Institute, Springfield, MA, USA
| | - Ahmed Khalil
- Department of Biology, University of Massachusetts, Amherst, MA, USA
| | | | - Alejandro P Heuck
- Departments of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, MA, USA
| | - Nagendra Yadava
- Pioneer Valley Life Sciences Institute, Springfield, MA, USA ; Department of Biology, University of Massachusetts, Amherst, MA, USA ; Division of Endocrinology, Diabetes & Metabolism at Baystate Medical Center of Tufts University School of Medicine, Springfield, MA, USA
| |
Collapse
|
330
|
Franko A, Baris OR, Bergschneider E, von Toerne C, Hauck SM, Aichler M, Walch AK, Wurst W, Wiesner RJ, Johnston ICD, de Angelis MH. Efficient isolation of pure and functional mitochondria from mouse tissues using automated tissue disruption and enrichment with anti-TOM22 magnetic beads. PLoS One 2013; 8:e82392. [PMID: 24349272 PMCID: PMC3861405 DOI: 10.1371/journal.pone.0082392] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Accepted: 11/01/2013] [Indexed: 12/13/2022] Open
Abstract
To better understand molecular mechanisms regulating changes in metabolism, as observed e.g. in diabetes or neuronal disorders, the function of mitochondria needs to be precisely determined. The usual isolation methods such as differential centrifugation result in isolates of highly variable quality and quantity. To fulfill the need of a reproducible isolation method from solid tissues, which is suitable to handle parallel samples simultaneously, we developed a protocol based on anti-TOM22 (translocase of outer mitochondrial membrane 22 homolog) antibody-coupled magnetic beads. To measure oxygen consumption rate in isolated mitochondria from various mouse tissues, a traditional Clark electrode and the high-throughput XF Extracellular Flux Analyzer were used. Furthermore, Western blots, transmission electron microscopic and proteomic studies were performed to analyze the purity and integrity of the mitochondrial preparations. Mitochondrial fractions isolated from liver, brain and skeletal muscle by anti-TOM22 magnetic beads showed oxygen consumption capacities comparable to previously reported values and little contamination with other organelles. The purity and quality of isolated mitochondria using anti-TOM22 magnetic beads was compared to traditional differential centrifugation protocol in liver and the results indicated an obvious advantage of the magnetic beads method compared to the traditional differential centrifugation technique.
Collapse
Affiliation(s)
- Andras Franko
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Olivier R. Baris
- Institute of Vegetative Physiology, Medical Faculty, University of Köln, Köln, Germany
| | | | - Christine von Toerne
- Research Unit Protein Science, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Stefanie M. Hauck
- Research Unit Protein Science, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Michaela Aichler
- Research Unit Analytical Pathology, Institute of Pathology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Axel K. Walch
- Research Unit Analytical Pathology, Institute of Pathology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München, Technische Universität München, Neuherberg, Germany
- Max Planck Institute of Psychiatry, Munich, Germany
- Technische Universität München, Lehrstuhl für Entwicklungsgenetik, c/o Helmholtz Zentrum München, Neuherberg, Germany
- DZNE – Deutsches Zentrum fuer Neurodegenerative Erkrankungen, Site Munich, Germany
| | - Rudolf J. Wiesner
- Institute of Vegetative Physiology, Medical Faculty, University of Köln, Köln, Germany
- Center for Molecular Medicine (CMMC), University of Köln, Köln, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Ageing-associated Diseases (CECAD), University of Köln, Köln, Germany
| | | | - Martin Hrabĕ de Angelis
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Technische Universität München, WZW - Center of Life and Food Science Weihenstephan, Chair of Experimental Genetics, Freising-Weihenstephan, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- * E-mail:
| |
Collapse
|
331
|
Jenkins Y, Sun TQ, Markovtsov V, Foretz M, Li W, Nguyen H, Li Y, Pan A, Uy G, Gross L, Baltgalvis K, Yung SL, Gururaja T, Kinoshita T, Owyang A, Smith IJ, McCaughey K, White K, Godinez G, Alcantara R, Choy C, Ren H, Basile R, Sweeny DJ, Xu X, Issakani SD, Carroll DC, Goff DA, Shaw SJ, Singh R, Boros LG, Laplante MA, Marcotte B, Kohen R, Viollet B, Marette A, Payan DG, Kinsella TM, Hitoshi Y. AMPK activation through mitochondrial regulation results in increased substrate oxidation and improved metabolic parameters in models of diabetes. PLoS One 2013; 8:e81870. [PMID: 24339975 PMCID: PMC3855387 DOI: 10.1371/journal.pone.0081870] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 10/19/2013] [Indexed: 12/28/2022] Open
Abstract
Modulation of mitochondrial function through inhibiting respiratory complex I activates a key sensor of cellular energy status, the 5'-AMP-activated protein kinase (AMPK). Activation of AMPK results in the mobilization of nutrient uptake and catabolism for mitochondrial ATP generation to restore energy homeostasis. How these nutrient pathways are affected in the presence of a potent modulator of mitochondrial function and the role of AMPK activation in these effects remain unclear. We have identified a molecule, named R419, that activates AMPK in vitro via complex I inhibition at much lower concentrations than metformin (IC50 100 nM vs 27 mM, respectively). R419 potently increased myocyte glucose uptake that was dependent on AMPK activation, while its ability to suppress hepatic glucose production in vitro was not. In addition, R419 treatment of mouse primary hepatocytes increased fatty acid oxidation and inhibited lipogenesis in an AMPK-dependent fashion. We have performed an extensive metabolic characterization of its effects in the db/db mouse diabetes model. In vivo metabolite profiling of R419-treated db/db mice showed a clear upregulation of fatty acid oxidation and catabolism of branched chain amino acids. Additionally, analyses performed using both 13C-palmitate and 13C-glucose tracers revealed that R419 induces complete oxidation of both glucose and palmitate to CO2 in skeletal muscle, liver, and adipose tissue, confirming that the compound increases mitochondrial function in vivo. Taken together, our results show that R419 is a potent inhibitor of complex I and modulates mitochondrial function in vitro and in diabetic animals in vivo. R419 may serve as a valuable molecular tool for investigating the impact of modulating mitochondrial function on nutrient metabolism in multiple tissues and on glucose and lipid homeostasis in diabetic animal models.
Collapse
Affiliation(s)
- Yonchu Jenkins
- Rigel Pharmaceuticals, Inc., South San Francisco, California, United States of America
| | - Tian-Qiang Sun
- Rigel Pharmaceuticals, Inc., South San Francisco, California, United States of America
| | - Vadim Markovtsov
- Rigel Pharmaceuticals, Inc., South San Francisco, California, United States of America
| | - Marc Foretz
- Inserm, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris cité, Paris, France
| | - Wei Li
- Rigel Pharmaceuticals, Inc., South San Francisco, California, United States of America
| | - Henry Nguyen
- Rigel Pharmaceuticals, Inc., South San Francisco, California, United States of America
| | - Yingwu Li
- Rigel Pharmaceuticals, Inc., South San Francisco, California, United States of America
| | - Alison Pan
- Rigel Pharmaceuticals, Inc., South San Francisco, California, United States of America
| | - Gerald Uy
- Rigel Pharmaceuticals, Inc., South San Francisco, California, United States of America
| | - Lisa Gross
- Rigel Pharmaceuticals, Inc., South San Francisco, California, United States of America
| | - Kristen Baltgalvis
- Rigel Pharmaceuticals, Inc., South San Francisco, California, United States of America
| | - Stephanie L. Yung
- Rigel Pharmaceuticals, Inc., South San Francisco, California, United States of America
| | - Tarikere Gururaja
- Rigel Pharmaceuticals, Inc., South San Francisco, California, United States of America
| | - Taisei Kinoshita
- Rigel Pharmaceuticals, Inc., South San Francisco, California, United States of America
| | - Alexander Owyang
- Rigel Pharmaceuticals, Inc., South San Francisco, California, United States of America
| | - Ira J. Smith
- Rigel Pharmaceuticals, Inc., South San Francisco, California, United States of America
| | - Kelly McCaughey
- Rigel Pharmaceuticals, Inc., South San Francisco, California, United States of America
| | - Kathy White
- Rigel Pharmaceuticals, Inc., South San Francisco, California, United States of America
| | - Guillermo Godinez
- Rigel Pharmaceuticals, Inc., South San Francisco, California, United States of America
| | - Raniel Alcantara
- Rigel Pharmaceuticals, Inc., South San Francisco, California, United States of America
| | - Carmen Choy
- Rigel Pharmaceuticals, Inc., South San Francisco, California, United States of America
| | - Hong Ren
- Rigel Pharmaceuticals, Inc., South San Francisco, California, United States of America
| | - Rachel Basile
- Rigel Pharmaceuticals, Inc., South San Francisco, California, United States of America
| | - David J. Sweeny
- Rigel Pharmaceuticals, Inc., South San Francisco, California, United States of America
| | - Xiang Xu
- Rigel Pharmaceuticals, Inc., South San Francisco, California, United States of America
| | - Sarkiz D. Issakani
- Rigel Pharmaceuticals, Inc., South San Francisco, California, United States of America
| | - David C. Carroll
- Rigel Pharmaceuticals, Inc., South San Francisco, California, United States of America
| | - Dane A. Goff
- Rigel Pharmaceuticals, Inc., South San Francisco, California, United States of America
| | - Simon J. Shaw
- Rigel Pharmaceuticals, Inc., South San Francisco, California, United States of America
| | - Rajinder Singh
- Rigel Pharmaceuticals, Inc., South San Francisco, California, United States of America
| | - Laszlo G. Boros
- SiDMAP, LLC, Los Angeles, California, United States of America
- Department of Pediatrics, Los Angeles Biomedical Research Institute (LABIOMED) at the Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Marc-André Laplante
- Department of Medicine, Faculty of Medicine, Cardiology Axis of the Institut Universitaire de Cardiologie et de Pneumologie de Québec (Hôpital Laval), Québec, Québec, Canada
| | - Bruno Marcotte
- Department of Medicine, Faculty of Medicine, Cardiology Axis of the Institut Universitaire de Cardiologie et de Pneumologie de Québec (Hôpital Laval), Québec, Québec, Canada
| | - Rita Kohen
- Department of Medicine, Faculty of Medicine, Cardiology Axis of the Institut Universitaire de Cardiologie et de Pneumologie de Québec (Hôpital Laval), Québec, Québec, Canada
| | - Benoit Viollet
- Inserm, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris cité, Paris, France
| | - André Marette
- Department of Medicine, Faculty of Medicine, Cardiology Axis of the Institut Universitaire de Cardiologie et de Pneumologie de Québec (Hôpital Laval), Québec, Québec, Canada
| | - Donald G. Payan
- Rigel Pharmaceuticals, Inc., South San Francisco, California, United States of America
| | - Todd M. Kinsella
- Rigel Pharmaceuticals, Inc., South San Francisco, California, United States of America
| | - Yasumichi Hitoshi
- Rigel Pharmaceuticals, Inc., South San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
332
|
Orr AL, Ashok D, Sarantos MR, Shi T, Hughes RE, Brand MD. Inhibitors of ROS production by the ubiquinone-binding site of mitochondrial complex I identified by chemical screening. Free Radic Biol Med 2013; 65:1047-1059. [PMID: 23994103 PMCID: PMC4321955 DOI: 10.1016/j.freeradbiomed.2013.08.170] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 08/12/2013] [Accepted: 08/16/2013] [Indexed: 12/21/2022]
Abstract
Mitochondrial production of reactive oxygen species is often considered an unavoidable consequence of aerobic metabolism and currently cannot be manipulated without perturbing oxidative phosphorylation. Antioxidants are widely used to suppress effects of reactive oxygen species after formation, but they can never fully prevent immediate effects at the sites of production. To identify site-selective inhibitors of mitochondrial superoxide/H2O2 production that do not interfere with mitochondrial energy metabolism, we developed a robust small-molecule screen and secondary profiling strategy. We describe the discovery and characterization of a compound (N-cyclohexyl-4-(4-nitrophenoxy)benzenesulfonamide; CN-POBS) that selectively inhibits superoxide/H2O2 production from the ubiquinone-binding site of complex I (site I(Q)) with no effects on superoxide/H2O2 production from other sites or on oxidative phosphorylation. Structure/activity studies identified a core structure that is important for potency and selectivity for site I(Q). By employing CN-POBS in mitochondria respiring on NADH-generating substrates, we show that site I(Q) does not produce significant amounts of superoxide/H2O2 during forward electron transport on glutamate plus malate. Our screening platform promises to facilitate further discovery of direct modulators of mitochondrially derived oxidative damage and advance our ability to understand and manipulate mitochondrial reactive oxygen species production under both normal and pathological conditions.
Collapse
Affiliation(s)
- Adam L Orr
- Buck Institute for Research on Aging, Novato, CA 94945, USA.
| | - Deepthi Ashok
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | | | - Tong Shi
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | | | - Martin D Brand
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| |
Collapse
|
333
|
Kenwood BM, Weaver JL, Bajwa A, Poon IK, Byrne FL, Murrow BA, Calderone JA, Huang L, Divakaruni AS, Tomsig JL, Okabe K, Lo RH, Cameron Coleman G, Columbus L, Yan Z, Saucerman JJ, Smith JS, Holmes JW, Lynch KR, Ravichandran KS, Uchiyama S, Santos WL, Rogers GW, Okusa MD, Bayliss DA, Hoehn KL. Identification of a novel mitochondrial uncoupler that does not depolarize the plasma membrane. Mol Metab 2013; 3:114-23. [PMID: 24634817 DOI: 10.1016/j.molmet.2013.11.005] [Citation(s) in RCA: 168] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 11/15/2013] [Accepted: 11/15/2013] [Indexed: 11/29/2022] Open
Abstract
Dysregulation of oxidative phosphorylation is associated with increased mitochondrial reactive oxygen species production and some of the most prevalent human diseases including obesity, cancer, diabetes, neurodegeneration, and heart disease. Chemical 'mitochondrial uncouplers' are lipophilic weak acids that transport protons into the mitochondrial matrix via a pathway that is independent of ATP synthase, thereby uncoupling nutrient oxidation from ATP production. Mitochondrial uncouplers also lessen the proton motive force across the mitochondrial inner membrane and thereby increase the rate of mitochondrial respiration while decreasing production of reactive oxygen species. Thus, mitochondrial uncouplers are valuable chemical tools that enable the measurement of maximal mitochondrial respiration and they have been used therapeutically to decrease mitochondrial reactive oxygen species production. However, the most widely used protonophore uncouplers such as carbonyl cyanide p-trifluoromethoxyphenylhydrazone (FCCP) and 2,4-dinitrophenol have off-target activity at other membranes that lead to a range of undesired effects including plasma membrane depolarization, mitochondrial inhibition, and cytotoxicity. These unwanted properties interfere with the measurement of mitochondrial function and result in a narrow therapeutic index that limits their usefulness in the clinic. To identify new mitochondrial uncouplers that lack off-target activity at the plasma membrane we screened a small molecule chemical library. Herein we report the identification and validation of a novel mitochondrial protonophore uncoupler (2-fluorophenyl){6-[(2-fluorophenyl)amino](1,2,5-oxadiazolo[3,4-e]pyrazin-5-yl)}amine, named BAM15, that does not depolarize the plasma membrane. Compared to FCCP, an uncoupler of equal potency, BAM15 treatment of cultured cells stimulates a higher maximum rate of mitochondrial respiration and is less cytotoxic. Furthermore, BAM15 is bioactive in vivo and dose-dependently protects mice from acute renal ischemic-reperfusion injury. From a technical standpoint, BAM15 represents an effective new tool that allows the study of mitochondrial function in the absence of off-target effects that can confound data interpretation. From a therapeutic perspective, BAM15-mediated protection from ischemia-reperfusion injury and its reduced toxicity will hopefully reignite interest in pharmacological uncoupling for the treatment of the myriad of diseases that are associated with altered mitochondrial function.
Collapse
Key Words
- ANT, adenine nucleotide translocase
- Bioenergetics
- CCCP
- DNP
- ECAR, extracellular acidification rate
- FCCP
- FCCP, carbonyl cyanide p-trifluoromethoxyphenylhydrazone
- Ischemia
- Mitochondria
- OCR, oxygen consumption rate
- ROS, reactive oxygen species
- TCA cycle, tricarboxylic acid cycle
- TMPD, N,N,N′,N′-tetramethyl-p-phenylenediamine dihydrochloride
- TMRM, tetramethylrhodamine
Collapse
Affiliation(s)
- Brandon M Kenwood
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Janelle L Weaver
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Amandeep Bajwa
- Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Ivan K Poon
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Frances L Byrne
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Beverley A Murrow
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Joseph A Calderone
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, VA 24061, USA
| | - Liping Huang
- Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Ajit S Divakaruni
- Department of Pharmacology, University of California at San Diego, La Jolla, CA 92093, USA
| | - Jose L Tomsig
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | | | - Ryan H Lo
- Department of Chemistry, University of Virginia, Charlottesville, VA 22908, USA
| | - G Cameron Coleman
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Linda Columbus
- Department of Chemistry, University of Virginia, Charlottesville, VA 22908, USA
| | - Zhen Yan
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA ; Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA ; Department of Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA
| | - Jeffrey J Saucerman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA ; Department of Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA
| | - Jeffrey S Smith
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Jeffrey W Holmes
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA ; Department of Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA
| | - Kevin R Lynch
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Kodi S Ravichandran
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | | | - Webster L Santos
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, VA 24061, USA
| | | | - Mark D Okusa
- Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Douglas A Bayliss
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Kyle L Hoehn
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA ; Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA ; Department of Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA ; Emily Couric Clinical Cancer Center, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
334
|
Peek CB, Affinati AH, Ramsey KM, Kuo HY, Yu W, Sena LA, Ilkayeva O, Marcheva B, Kobayashi Y, Omura C, Levine DC, Bacsik DJ, Gius D, Newgard CB, Goetzman E, Chandel NS, Denu JM, Mrksich M, Bass J. Circadian clock NAD+ cycle drives mitochondrial oxidative metabolism in mice. Science 2013; 342:1243417. [PMID: 24051248 PMCID: PMC3963134 DOI: 10.1126/science.1243417] [Citation(s) in RCA: 465] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Circadian clocks are self-sustained cellular oscillators that synchronize oxidative and reductive cycles in anticipation of the solar cycle. We found that the clock transcription feedback loop produces cycles of nicotinamide adenine dinucleotide (NAD(+)) biosynthesis, adenosine triphosphate production, and mitochondrial respiration through modulation of mitochondrial protein acetylation to synchronize oxidative metabolic pathways with the 24-hour fasting and feeding cycle. Circadian control of the activity of the NAD(+)-dependent deacetylase sirtuin 3 (SIRT3) generated rhythms in the acetylation and activity of oxidative enzymes and respiration in isolated mitochondria, and NAD(+) supplementation restored protein deacetylation and enhanced oxygen consumption in circadian mutant mice. Thus, circadian control of NAD(+) bioavailability modulates mitochondrial oxidative function and organismal metabolism across the daily cycles of fasting and feeding.
Collapse
Affiliation(s)
- Clara Bien Peek
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Alison H. Affinati
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Kathryn Moynihan Ramsey
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Hsin-Yu Kuo
- Department of Chemistry, Northwestern University, Evanston, IL 60208, USA
- Howard Hughes Medical Institute, Northwestern University, Evanston, IL 60208, USA
| | - Wei Yu
- Department of Biomolecular Chemistry and Wisconsin Institute for Discovery, University of Wisconsin, Madison, WI 53715, USA
| | - Laura A. Sena
- Deparment of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Olga Ilkayeva
- Sarah W. Stedman Nutrition and Metabolism Center, Departments of Pharmacology and Cancer Biology and Medicine, Duke University Medical Center, Durham, NC 27705, USA
| | - Biliana Marcheva
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Yumiko Kobayashi
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Chiaki Omura
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Daniel C. Levine
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - David J. Bacsik
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - David Gius
- Department of Radiation Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Christopher B. Newgard
- Sarah W. Stedman Nutrition and Metabolism Center, Departments of Pharmacology and Cancer Biology and Medicine, Duke University Medical Center, Durham, NC 27705, USA
| | - Eric Goetzman
- Department of Pediatrics, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Navdeep S. Chandel
- Deparment of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - John M. Denu
- Department of Biomolecular Chemistry and Wisconsin Institute for Discovery, University of Wisconsin, Madison, WI 53715, USA
| | - Milan Mrksich
- Department of Chemistry, Northwestern University, Evanston, IL 60208, USA
- Howard Hughes Medical Institute, Northwestern University, Evanston, IL 60208, USA
| | - Joseph Bass
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| |
Collapse
|
335
|
Sew YS, Ströher E, Holzmann C, Huang S, Taylor NL, Jordana X, Millar AH. Multiplex micro-respiratory measurements of Arabidopsis tissues. THE NEW PHYTOLOGIST 2013; 200:922-932. [PMID: 23834713 DOI: 10.1111/nph.12394] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Accepted: 05/29/2013] [Indexed: 05/08/2023]
Abstract
Researchers often want to study the respiratory properties of individual parts of plants in response to a range of treatments. Arabidopsis is an obvious model for this work; however, because of its size, it represents a challenge for gas exchange measurements of respiration. The combination of micro-respiratory technologies with multiplex assays has the potential to bridge this gap, and make measurements possible in this model plant species. We show the adaptation of the commercial technology used for mammalian cell respiration analysis to study three critical tissues of interest: leaf sections, root tips and seeds. The measurement of respiration in single leaf discs has allowed the age dependence of the respiration rate in Arabidopsis leaves across the rosette to be observed. The oxygen consumption of single root tips from plate-grown seedlings shows the enhanced respiration of root tips and their time-dependent susceptibility to salinity. The monitoring of single Arabidopsis seeds shows the kinetics of respiration over 48 h post-imbibition, and the effect of the phytohormones gibberellic acid (GA3 ) and abscisic acid (ABA) on respiration during seed germination. These studies highlight the potential for multiplexed micro-respiratory assays to study oxygen consumption in Arabidopsis tissues, and open up new possibilities to screen and study mutants and to identify differences in ecotypes or populations of different plant species.
Collapse
Affiliation(s)
- Yun Shin Sew
- ARC Centre of Excellence in Plant Energy Biology, The University of Western Australia, Bayliss Building M316, 35 Stirling Highway, Crawley, WA, 6009, Australia
- Centre for Comparative Analysis of Biomolecular Networks (CABiN), The University of Western Australia, Bayliss Building M316, 35 Stirling Highway, Crawley, WA, 6009, Australia
| | - Elke Ströher
- ARC Centre of Excellence in Plant Energy Biology, The University of Western Australia, Bayliss Building M316, 35 Stirling Highway, Crawley, WA, 6009, Australia
- Centre for Comparative Analysis of Biomolecular Networks (CABiN), The University of Western Australia, Bayliss Building M316, 35 Stirling Highway, Crawley, WA, 6009, Australia
| | - Cristián Holzmann
- ARC Centre of Excellence in Plant Energy Biology, The University of Western Australia, Bayliss Building M316, 35 Stirling Highway, Crawley, WA, 6009, Australia
- Millenium Nucleus in Plant Functional Genomics, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidád Católica de Chile, Casilla 114-D, Santiago, Chile
| | - Shaobai Huang
- ARC Centre of Excellence in Plant Energy Biology, The University of Western Australia, Bayliss Building M316, 35 Stirling Highway, Crawley, WA, 6009, Australia
- Centre for Comparative Analysis of Biomolecular Networks (CABiN), The University of Western Australia, Bayliss Building M316, 35 Stirling Highway, Crawley, WA, 6009, Australia
| | - Nicolas L Taylor
- ARC Centre of Excellence in Plant Energy Biology, The University of Western Australia, Bayliss Building M316, 35 Stirling Highway, Crawley, WA, 6009, Australia
- Centre for Comparative Analysis of Biomolecular Networks (CABiN), The University of Western Australia, Bayliss Building M316, 35 Stirling Highway, Crawley, WA, 6009, Australia
| | - Xavier Jordana
- Millenium Nucleus in Plant Functional Genomics, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidád Católica de Chile, Casilla 114-D, Santiago, Chile
| | - A Harvey Millar
- ARC Centre of Excellence in Plant Energy Biology, The University of Western Australia, Bayliss Building M316, 35 Stirling Highway, Crawley, WA, 6009, Australia
- Centre for Comparative Analysis of Biomolecular Networks (CABiN), The University of Western Australia, Bayliss Building M316, 35 Stirling Highway, Crawley, WA, 6009, Australia
| |
Collapse
|
336
|
Billiard J, Dennison JB, Briand J, Annan RS, Chai D, Colón M, Dodson CS, Gilbert SA, Greshock J, Jing J, Lu H, McSurdy-Freed JE, Orband-Miller LA, Mills GB, Quinn CJ, Schneck JL, Scott GF, Shaw AN, Waitt GM, Wooster RF, Duffy KJ. Quinoline 3-sulfonamides inhibit lactate dehydrogenase A and reverse aerobic glycolysis in cancer cells. Cancer Metab 2013; 1:19. [PMID: 24280423 PMCID: PMC4178217 DOI: 10.1186/2049-3002-1-19] [Citation(s) in RCA: 163] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 08/27/2013] [Indexed: 12/13/2022] Open
Abstract
Background Most normal cells in the presence of oxygen utilize glucose for mitochondrial oxidative phosphorylation. In contrast, many cancer cells rapidly convert glucose to lactate in the cytosol, a process termed aerobic glycolysis. This glycolytic phenotype is enabled by lactate dehydrogenase (LDH), which catalyzes the inter-conversion of pyruvate and lactate. The purpose of this study was to identify and characterize potent and selective inhibitors of LDHA. Methods High throughput screening and lead optimization were used to generate inhibitors of LDHA enzymatic activity. Effects of these inhibitors on metabolism were evaluated using cell-based lactate production, oxygen consumption, and 13C NMR spectroscopy assays. Changes in comprehensive metabolic profile, cell proliferation, and apoptosis were assessed upon compound treatment. Results 3-((3-carbamoyl-7-(3,5-dimethylisoxazol-4-yl)-6-methoxyquinolin-4-yl) amino) benzoic acid was identified as an NADH-competitive LDHA inhibitor. Lead optimization yielded molecules with LDHA inhibitory potencies as low as 2 nM and 10 to 80-fold selectivity over LDHB. Molecules in this family rapidly and profoundly inhibited lactate production rates in multiple cancer cell lines including hepatocellular and breast carcinomas. Consistent with selective inhibition of LDHA, the most sensitive breast cancer cell lines to lactate inhibition in hypoxic conditions were cells with low expression of LDHB. Our inhibitors increased rates of oxygen consumption in hepatocellular carcinoma cells at doses up to 3 microM, while higher concentrations directly inhibited mitochondrial function. Analysis of more than 500 metabolites upon LDHA inhibition in Snu398 cells revealed that intracellular concentrations of glycolysis and citric acid cycle intermediates were increased, consistent with enhanced Krebs cycle activity and blockage of cytosolic glycolysis. Treatment with these compounds also potentiated PKM2 activity and promoted apoptosis in Snu398 cells. Conclusions Rapid chemical inhibition of LDHA by these quinoline 3-sulfonamids led to profound metabolic alterations and impaired cell survival in carcinoma cells making it a compelling strategy for treating solid tumors that rely on aerobic glycolysis for survival.
Collapse
Affiliation(s)
- Julia Billiard
- Cancer Metabolism DPU, GlaxoSmithKline, Collegeville PA, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
337
|
Hyperoxia decreases glycolytic capacity, glycolytic reserve and oxidative phosphorylation in MLE-12 cells and inhibits complex I and II function, but not complex IV in isolated mouse lung mitochondria. PLoS One 2013; 8:e73358. [PMID: 24023862 PMCID: PMC3759456 DOI: 10.1371/journal.pone.0073358] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Accepted: 07/26/2013] [Indexed: 01/01/2023] Open
Abstract
High levels of oxygen (hyperoxia) are frequently used in critical care units and in conditions of respiratory insufficiencies in adults, as well as in infants. However, hyperoxia has been implicated in a number of pulmonary disorders including bronchopulmonary dysplasia (BPD) and adult respiratory distress syndrome (ARDS). Hyperoxia increases the generation of reactive oxygen species (ROS) in the mitochondria that could impair the function of the mitochondrial electron transport chain. We analyzed lung mitochondrial function in hyperoxia using the XF24 analyzer (extracellular flux) and optimized the assay for lung epithelial cells and mitochondria isolated from lungs of mice. Our data show that hyperoxia decreases basal oxygen consumption rate (OCR), spare respiratory capacity, maximal respiration and ATP turnover in MLE-12 cells. There was significant decrease in glycolytic capacity and glycolytic reserve in MLE-12 cells exposed to hyperoxia. Using mitochondria isolated from lungs of mice exposed to hyperoxia or normoxia we have shown that hyperoxia decreased the basal, state 3 and state3 μ (respiration in an uncoupled state) respirations. Further, using substrate or inhibitor of a specific complex we show that the OCR via complex I and II, but not complex IV was decreased, demonstrating that complexes I and II are specific targets of hyperoxia. Further, the activities of complex I (NADH dehydrogenase, NADH-DH) and complex II (succinate dehydrogenase, SDH) were decreased in hyperoxia, but the activity of complex IV (cytochrome oxidase, COX) remains unchanged. Taken together, our study show that hyperoxia impairs glycolytic and mitochondrial energy metabolism in in tact cells, as well as in lungs of mice by selectively inactivating components of electron transport system.
Collapse
|
338
|
Adjeitey CNK, Mailloux RJ, Dekemp RA, Harper ME. Mitochondrial uncoupling in skeletal muscle by UCP1 augments energy expenditure and glutathione content while mitigating ROS production. Am J Physiol Endocrinol Metab 2013; 305:E405-15. [PMID: 23757405 PMCID: PMC3742851 DOI: 10.1152/ajpendo.00057.2013] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Enhancement of proton leaks in muscle tissue represents a potential target for obesity treatment. In this study, we examined the bioenergetic and physiological implications of increased proton leak in skeletal muscle. To induce muscle-specific increases in proton leak, we used mice that selectively express uncoupling protein-1 (UCP1) in skeletal muscle tissue. UCP1 expression in muscle mitochondria was ∼13% of levels in brown adipose tissue (BAT) mitochondria and caused increased GDP-sensitive proton leak. This was associated with an increase in whole body energy expenditure and a decrease in white adipose tissue content. Muscle UCP1 activity had divergent effects on mitochondrial ROS emission and glutathione levels compared with BAT. UCP1 in muscle increased total mitochondrial glutathione levels ∼7.6 fold. Intriguingly, unlike in BAT mitochondria, leak through UCP1 in muscle controlled mitochondrial ROS emission. Inhibition of UCP1 with GDP in muscle mitochondria increased ROS emission ∼2.8-fold relative to WT muscle mitochondria. GDP had no impact on ROS emission from BAT mitochondria from either genotype. Collectively, these findings indicate that selective induction of UCP1-mediated proton leak in muscle can increase whole body energy expenditure and decrease adiposity. Moreover, ectopic UCP1 expression in skeletal muscle can control mitochondrial ROS emission, while it apparently plays no such role in its endogenous tissue, brown fat.
Collapse
Affiliation(s)
- Cyril Nii-Klu Adjeitey
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | | | | | | |
Collapse
|
339
|
Still AJ, Floyd BJ, Hebert AS, Bingman CA, Carson JJ, Gunderson DR, Dolan BK, Grimsrud PA, Dittenhafer-Reed KE, Stapleton DS, Keller MP, Westphall MS, Denu JM, Attie AD, Coon JJ, Pagliarini DJ. Quantification of mitochondrial acetylation dynamics highlights prominent sites of metabolic regulation. J Biol Chem 2013; 288:26209-26219. [PMID: 23864654 DOI: 10.1074/jbc.m113.483396] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Lysine acetylation is rapidly becoming established as a key post-translational modification for regulating mitochondrial metabolism. Nonetheless, distinguishing regulatory sites from among the thousands identified by mass spectrometry and elucidating how these modifications alter enzyme function remain primary challenges. Here, we performed multiplexed quantitative mass spectrometry to measure changes in the mouse liver mitochondrial acetylproteome in response to acute and chronic alterations in nutritional status, and integrated these data sets with our compendium of predicted Sirt3 targets. These analyses highlight a subset of mitochondrial proteins with dynamic acetylation sites, including acetyl-CoA acetyltransferase 1 (Acat1), an enzyme central to multiple metabolic pathways. We performed in vitro biochemistry and molecular modeling to demonstrate that acetylation of Acat1 decreases its activity by disrupting the binding of coenzyme A. Collectively, our data reveal an important new target of regulatory acetylation and provide a foundation for investigating the role of select mitochondrial protein acetylation sites in mediating acute and chronic metabolic transitions.
Collapse
Affiliation(s)
| | | | - Alexander S Hebert
- Chemistry, and; Genome Center of Wisconsin, University of Wisconsin-Madison, Madison, Wisconsin 53706
| | | | | | | | | | | | | | | | | | - Michael S Westphall
- Genome Center of Wisconsin, University of Wisconsin-Madison, Madison, Wisconsin 53706
| | | | | | - Joshua J Coon
- Chemistry, and; Genome Center of Wisconsin, University of Wisconsin-Madison, Madison, Wisconsin 53706; Biomolecular Chemistry and
| | | |
Collapse
|
340
|
Wills LP, Beeson GC, Trager RE, Lindsey CC, Beeson CC, Peterson YK, Schnellmann RG. High-throughput respirometric assay identifies predictive toxicophore of mitochondrial injury. Toxicol Appl Pharmacol 2013; 272:490-502. [PMID: 23811330 DOI: 10.1016/j.taap.2013.06.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 06/14/2013] [Accepted: 06/18/2013] [Indexed: 11/29/2022]
Abstract
Many environmental chemicals and drugs negatively affect human health through deleterious effects on mitochondrial function. Currently there is no chemical library of mitochondrial toxicants, and no reliable methods for predicting mitochondrial toxicity. We hypothesized that discrete toxicophores defined by distinct chemical entities can identify previously unidentified mitochondrial toxicants. We used a respirometric assay to screen 1760 compounds (5 μM) from the LOPAC and ChemBridge DIVERSet libraries. Thirty-one of the assayed compounds decreased uncoupled respiration, a stress test for mitochondrial dysfunction, prior to a decrease in cell viability and reduced the oxygen consumption rate in isolated mitochondria. The mitochondrial toxicants were grouped by chemical similarity and two clusters containing four compounds each were identified. Cheminformatic analysis of one of the clusters identified previously uncharacterized mitochondrial toxicants from the ChemBridge DIVERSet. This approach will enable the identification of mitochondrial toxicants and advance the prediction of mitochondrial toxicity for both drug discovery and risk assessment.
Collapse
|
341
|
Das KC, Muniyappa H. Age-dependent mitochondrial energy dynamics in the mice heart: role of superoxide dismutase-2. Exp Gerontol 2013; 48:947-59. [PMID: 23806974 PMCID: PMC4045457 DOI: 10.1016/j.exger.2013.06.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Revised: 06/07/2013] [Accepted: 06/11/2013] [Indexed: 12/02/2022]
Abstract
The aging process alters cardiac physiology, decreases the number of cardiomyocytes and alters the energy metabolism. Mitochondrial dysfunction in aging is believed to cause these functional and phenotypic changes in the heart. Although precise understanding of alterations of mitochondrial respiration in aging is necessary to manage heart diseases in the elderly population conflicting data on the function of specific complex of electron transport chain of the heart mitochondria limits the intervention process. We have addressed these issues using the assay of mitochondrial coupling and electron flow to assess specific functional defects in mitochondria isolated from young or aged mice. Our results demonstrate that cardiac mitochondria from older mice utilize oxygen at a decreased rate via complex I, II or IV compared to younger mice. We further show that mitochondrial function decreases in young Sod2+/− mice heart compared to young wildtype mice. However, the mitochondrial function remains unchanged in older Sod2+/− mice heart compared to younger Sod2+/− mice heart. Further, the oxygen consumption remains similar in old wildtype mice and old Sod2+/− mice heart mitochondria. The expression and activity of Sod2 in young or old Sod2+/− mice heart remain unchanged. These data demonstrate that decreased oxygen utilization in older age could have resulted in decreased mitochondrial ROS-mediated oxidative damage requiring less Sod2 for protection against mitochondrial oxidative stress in older wildtype or older Sod2+/− mice.
Collapse
Affiliation(s)
- Kumuda C Das
- Department of Anesthesiology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| | | |
Collapse
|
342
|
McQuaker SJ, Quinlan CL, Caldwell ST, Brand MD, Hartley RC. A prototypical small-molecule modulator uncouples mitochondria in response to endogenous hydrogen peroxide production. Chembiochem 2013; 14:993-1000. [PMID: 23640856 PMCID: PMC3743171 DOI: 10.1002/cbic.201300115] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Indexed: 12/31/2022]
Abstract
A high membrane potential across the mitochondrial inner membrane leads to the production of the reactive oxygen species (ROS) implicated in aging and age-related diseases. A prototypical drug for the correction of this type of mitochondrial dysfunction is presented. MitoDNP-SUM accumulates in mitochondria in response to the membrane potential due to its mitochondria-targeting alkyltriphenylphosphonium (TPP) cation and is uncaged by endogenous hydrogen peroxide to release the mitochondrial uncoupler, 2,4-dinitrophenol (DNP). DNP is known to reduce the high membrane potential responsible for the production of ROS. The approach potentially represents a general method for the delivery of drugs to the mitochondrial matrix through mitochondria targeting and H(2)O(2)-induced uncaging.
Collapse
Affiliation(s)
- Stephen J McQuaker
- WestChem School of Chemistry, University of GlasgowGlasgow, G12 8QQ (UK) E-mail:
| | - Casey L Quinlan
- Buck Institute for Research on Aging8001 Redwood Boulevard, Novato, California 94945 (USA)
| | - Stuart T Caldwell
- WestChem School of Chemistry, University of GlasgowGlasgow, G12 8QQ (UK) E-mail:
| | - Martin D Brand
- Buck Institute for Research on Aging8001 Redwood Boulevard, Novato, California 94945 (USA)
| | - Richard C Hartley
- WestChem School of Chemistry, University of GlasgowGlasgow, G12 8QQ (UK) E-mail:
| |
Collapse
|
343
|
Wiley SE, Andreyev AY, Divakaruni AS, Karisch R, Perkins G, Wall EA, van der Geer P, Chen YF, Tsai TF, Simon MI, Neel BG, Dixon JE, Murphy AN. Wolfram Syndrome protein, Miner1, regulates sulphydryl redox status, the unfolded protein response, and Ca2+ homeostasis. EMBO Mol Med 2013; 5:904-18. [PMID: 23703906 PMCID: PMC3779451 DOI: 10.1002/emmm.201201429] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2012] [Revised: 03/26/2013] [Accepted: 03/27/2013] [Indexed: 01/21/2023] Open
Abstract
Miner1 is a redox-active 2Fe2S cluster protein. Mutations in Miner1 result in Wolfram Syndrome, a metabolic disease associated with diabetes, blindness, deafness, and a shortened lifespan. Embryonic fibroblasts from Miner1(-/-) mice displayed ER stress and showed hallmarks of the unfolded protein response. In addition, loss of Miner1 caused a depletion of ER Ca(2+) stores, a dramatic increase in mitochondrial Ca(2+) load, increased reactive oxygen and nitrogen species, an increase in the GSSG/GSH and NAD(+)/NADH ratios, and an increase in the ADP/ATP ratio consistent with enhanced ATP utilization. Furthermore, mitochondria in fibroblasts lacking Miner1 displayed ultrastructural alterations, such as increased cristae density and punctate morphology, and an increase in O2 consumption. Treatment with the sulphydryl anti-oxidant N-acetylcysteine reversed the abnormalities in the Miner1 deficient cells, suggesting that sulphydryl reducing agents should be explored as a treatment for this rare genetic disease.
Collapse
Affiliation(s)
- Sandra E Wiley
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
344
|
Singh IN, Gilmer LK, Miller DM, Cebak JE, Wang JA, Hall ED. Phenelzine mitochondrial functional preservation and neuroprotection after traumatic brain injury related to scavenging of the lipid peroxidation-derived aldehyde 4-hydroxy-2-nonenal. J Cereb Blood Flow Metab 2013; 33:593-9. [PMID: 23321786 PMCID: PMC3618398 DOI: 10.1038/jcbfm.2012.211] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Phenelzine (PZ) is a scavenger of the lipid peroxidation (LP)-derived reactive aldehyde 4-hydroxynonenal (4-HNE) due to its hydrazine functional group, which can covalently react with 4-HNE. In this study, we first examined the ability of PZ to prevent the respiratory depressant effects of 4-HNE on normal isolated brain cortical mitochondria. Second, in rats subjected to controlled cortical impact traumatic brain injury (CCI-TBI), we evaluated PZ (10 mg/kg subcutaneously at 15 minutes after CCI-TBI) to attenuate 3-hour post-TBI mitochondrial respiratory dysfunction, and in separate animals, to improve cortical tissue sparing at 14 days. While 4-HNE exposure inhibited mitochondrial complex I and II respiration in a concentration-dependent manner, pretreatment with equimolar concentrations of PZ antagonized these effects. Western blot analysis demonstrated a PZ decrease in 4-HNE in mitochondrial proteins. Mitochondria isolated from peri-contusional brain tissue of CCI-TBI rats treated with vehicle at 15 minutes after injury showed a 37% decrease in the respiratory control ratio (RCR) relative to noninjured mitochondria. In PZ-treated rats, RCR suppression was prevented (P<0.05 versus vehicle). In another cohort, PZ administration increased spared cortical tissue from 86% to 97% (P<0.03). These results suggest that PZ's neuroprotective effect is due to mitochondrial protection by scavenging of LP-derived 4-HNE.
Collapse
Affiliation(s)
- Indrapal N Singh
- Spinal Cord and Brain Injury Research Center (SCoBIRC), Department of Anatomy and Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | | | | | | | | | | |
Collapse
|
345
|
Abstract
A growing body of research is investigating the potential contribution of mitochondrial function to the etiology of type 2 diabetes. Numerous in vitro, in situ, and in vivo methodologies are available to examine various aspects of mitochondrial function, each requiring an understanding of their principles, advantages, and limitations. This review provides investigators with a critical overview of the strengths, limitations and critical experimental parameters to consider when selecting and conducting studies on mitochondrial function. In vitro (isolated mitochondria) and in situ (permeabilized cells/tissue) approaches provide direct access to the mitochondria, allowing for study of mitochondrial bioenergetics and redox function under defined substrate conditions. Several experimental parameters must be tightly controlled, including assay media, temperature, oxygen concentration, and in the case of permeabilized skeletal muscle, the contractile state of the fibers. Recently developed technology now offers the opportunity to measure oxygen consumption in intact cultured cells. Magnetic resonance spectroscopy provides the most direct way of assessing mitochondrial function in vivo with interpretations based on specific modeling approaches. The continuing rapid evolution of these technologies offers new and exciting opportunities for deciphering the potential role of mitochondrial function in the etiology and treatment of diabetes.
Collapse
Affiliation(s)
- Christopher G R Perry
- School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada.
| | | | | | | |
Collapse
|
346
|
Ye F, Hoppel CL. Measuring oxidative phosphorylation in human skin fibroblasts. Anal Biochem 2013; 437:52-8. [PMID: 23462540 DOI: 10.1016/j.ab.2013.02.010] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Revised: 02/12/2013] [Accepted: 02/13/2013] [Indexed: 12/21/2022]
Abstract
An approach has been developed to quantitate oxidative phosphorylation in harvested human skin fibroblasts that have been permeabilized with digitonin. In protocol 1, state 3 rates are measured with complex I and II substrates, followed by uncoupled maximal oxidative capacity measured in the presence of these combined substrates as well as through complex IV. In protocol 2, state 3 rates are measured using palmitoylcarnitine to monitor fatty acid oxidation and duroquinol to assess the flux through complex III; uncoupled duroquinol oxidation measures maximal oxidative capacity through complex III. The activity of citrate synthase is determined in every experiment as a marker of the amount of mitochondria per chamber. Data are expressed on the basis of cell count (per million fibroblasts), of protein, or of citrate synthase activity. Cell growth conditions are optimized, and it is necessary to keep cultured cells from reaching confluency. Cultures in passages 3 to 10 show reproducible oxidative phosphorylation data. Based on the data from the 15 normal human skin fibroblast lines, we are evaluating the use of this approach to diagnose systemic mitochondrial disease and avoid issues associated with open skeletal muscle biopsy.
Collapse
Affiliation(s)
- Fang Ye
- Center for Mitochondrial Disease, Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | | |
Collapse
|
347
|
Cyr AR, Brown KE, McCormick ML, Coleman MC, Case AJ, Watts GS, Futscher BW, Spitz DR, Domann FE. Maintenance of mitochondrial genomic integrity in the absence of manganese superoxide dismutase in mouse liver hepatocytes. Redox Biol 2013; 1:172-7. [PMID: 24024150 PMCID: PMC3757676 DOI: 10.1016/j.redox.2013.01.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 12/21/2012] [Accepted: 01/02/2013] [Indexed: 12/21/2022] Open
Abstract
Manganese superoxide dismutase, encoded by the Sod2 gene, is a ubiquitously expressed mitochondrial antioxidant enzyme that is essential for mammalian life. Mice born with constitutive genetic knockout of Sod2 do not survive the neonatal stage, which renders the longitudinal study of the biochemical and metabolic effects of Sod2 loss difficult. However, multiple studies have demonstrated that tissue-specific knockout of Sod2 in murine liver yields no observable gross pathology or injury to the mouse. We hypothesized that Sod2 loss may have sub-pathologic effects on liver biology, including the acquisition of reactive oxygen species-mediated mitochondrial DNA mutations. To evaluate this, we established and verified a hepatocyte-specific knockout of Sod2 in C57/B6 mice using Cre-LoxP recombination technology. We utilized deep sequencing to identify possible mutations in Sod2−/− mitochondrial DNA as compared to wt, and both RT-PCR and traditional biochemical assays to evaluate baseline differences in redox-sensitive pathways in Sod2−/− hepatocytes. Surprisingly, no mutations in Sod2−/− mitochondrial DNA were detected despite measurable increases in dihydroethidium staining in situ and concomitant decreases in complex II activity indicative of elevated superoxide in the Sod2−/− hepatocytes. In contrast, numerous compensatory alterations in gene expression were identified that suggest hepatocytes have a remarkable capacity to adapt and overcome the loss of Sod2 through transcriptional means. Taken together, these results suggest that murine hepatocytes have a large reserve capacity to cope with the presence of additional mitochondrial reactive oxygen species.
Collapse
Affiliation(s)
- Anthony R. Cyr
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Carver College of Medicine and the Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Kyle E. Brown
- Department of Internal Medicine, Gastroenterology Division, Carver College of Medicine, The University of Iowa, Iowa City, IA 52242, USA
| | - Michael L. McCormick
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Carver College of Medicine and the Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Mitchell C. Coleman
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Carver College of Medicine and the Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Adam J. Case
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Carver College of Medicine and the Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| | - George S. Watts
- University of Arizona Cancer Center and Department of Pharmacology, University of Arizona, Tucson, AZ 85724, USA
| | - Bernard W. Futscher
- University of Arizona Cancer Center and Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ 85724, USA
| | - Douglas R. Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Carver College of Medicine and the Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Frederick E. Domann
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Carver College of Medicine and the Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
- Correspondence to: Free Radical and Radiation Biology Program, B180 Medical Laboratories, 500 Newton Road, The University of Iowa, Iowa City, IA 52242, USA. Tel.: +1 319 335 8019.
| |
Collapse
|
348
|
Wahl DR, Byersdorfer CA, Ferrara JLM, Opipari AW, Glick GD. Distinct metabolic programs in activated T cells: opportunities for selective immunomodulation. Immunol Rev 2013; 249:104-15. [PMID: 22889218 DOI: 10.1111/j.1600-065x.2012.01148.x] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
For several decades, it has been known that T-cell activation in vitro leads to increased glycolytic metabolism that fuels proliferation and effector function. Recently, this simple model has been complicated by the observation that different T-cell subsets differentially regulate fundamental metabolic pathways under the control of distinct molecular regulators. Although the majority of these data have been generated in vitro, several recent studies have documented the metabolism of T cells activated in vivo. Here, we review the recent data surrounding the differential regulation of metabolism by distinct T-cell subsets in vitro and in vivo and discuss how differential metabolic regulation might facilitate T-cell function vis-à-vis proliferation, survival, and energy production. We further discuss the important therapeutic implications of differential metabolism across T-cell subsets and review recent successes in exploiting lymphocyte metabolism to treat immune-mediated diseases.
Collapse
Affiliation(s)
- Daniel R Wahl
- Chemical Biology Doctoral Program, University of Michigan, Ann Arbor, MI 48109-1055, USA
| | | | | | | | | |
Collapse
|
349
|
Amigo I, Traba J, González-Barroso MM, Rueda CB, Fernández M, Rial E, Sánchez A, Satrústegui J, Del Arco A. Glucagon regulation of oxidative phosphorylation requires an increase in matrix adenine nucleotide content through Ca2+ activation of the mitochondrial ATP-Mg/Pi carrier SCaMC-3. J Biol Chem 2013; 288:7791-7802. [PMID: 23344948 DOI: 10.1074/jbc.m112.409144] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
It has been known for a long time that mitochondria isolated from hepatocytes treated with glucagon or Ca(2+)-mobilizing agents such as phenylephrine show an increase in their adenine nucleotide (AdN) content, respiratory activity, and calcium retention capacity (CRC). Here, we have studied the role of SCaMC-3/slc25a23, the mitochondrial ATP-Mg/Pi carrier present in adult mouse liver, in the control of mitochondrial AdN levels and respiration in response to Ca(2+) signals as a candidate target of glucagon actions. With the use of SCaMC-3 knock-out (KO) mice, we have found that the carrier is responsible for the accumulation of AdNs in liver mitochondria in a strictly Ca(2+)-dependent way with an S0.5 for Ca(2+) activation of 3.3 ± 0.9 μm. Accumulation of matrix AdNs allows a SCaMC-3-dependent increase in CRC. In addition, SCaMC-3-dependent accumulation of AdNs is required to acquire a fully active state 3 respiration in AdN-depleted liver mitochondria, although further accumulation of AdNs is not followed by increases in respiration. Moreover, glucagon addition to isolated hepatocytes increases oligomycin-sensitive oxygen consumption and maximal respiratory rates in cells derived from wild type, but not SCaMC-3-KO mice and glucagon administration in vivo results in an increase in AdN content, state 3 respiration and CRC in liver mitochondria in wild type but not in SCaMC-3-KO mice. These results show that SCaMC-3 is required for the increase in oxidative phosphorylation observed in liver mitochondria in response to glucagon and Ca(2+)-mobilizing agents, possibly by allowing a Ca(2+)-dependent accumulation of mitochondrial AdNs and matrix Ca(2+), events permissive for other glucagon actions.
Collapse
Affiliation(s)
- Ignacio Amigo
- Departmento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid-CSIC; Centro de Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER) 28049 Madrid
| | - Javier Traba
- Departmento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid-CSIC; Centro de Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER) 28049 Madrid
| | - M Mar González-Barroso
- Departamento de Medicina Celular y Molecular, Centro de Investigaciones Biológicas (CIB), Consejo Superior de Investigaciones Científicas 28040 Madrid
| | - Carlos B Rueda
- Departmento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid-CSIC; Centro de Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER) 28049 Madrid
| | - Margarita Fernández
- Departamento de Bioquímica y Biología Molecular II, Facultad de Farmacia, Universidad Complutense de Madrid 28040 Madrid
| | - Eduardo Rial
- Departamento de Medicina Celular y Molecular, Centro de Investigaciones Biológicas (CIB), Consejo Superior de Investigaciones Científicas 28040 Madrid
| | - Aránzazu Sánchez
- Departamento de Bioquímica y Biología Molecular II, Facultad de Farmacia, Universidad Complutense de Madrid 28040 Madrid
| | - Jorgina Satrústegui
- Departmento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid-CSIC; Centro de Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER) 28049 Madrid.
| | - Araceli Del Arco
- Área de Bioquímica, Centro Regional de Investigaciones Biomédicas, Facultad de Ciencias Ambientales y Bioquímica, Universidad de Castilla La Mancha (UCLM), Avda. Carlos III s/n, Toledo 45071, Spain.
| |
Collapse
|
350
|
Mailloux RJ, Xuan JY, Beauchamp B, Jui L, Lou M, Harper ME. Glutaredoxin-2 is required to control proton leak through uncoupling protein-3. J Biol Chem 2013; 288:8365-8379. [PMID: 23335511 DOI: 10.1074/jbc.m112.442905] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Glutathionylation has emerged as a key modification required for controlling protein function in response to changes in cell redox status. Recently, we showed that the glutathionylation state of uncoupling protein-3 (UCP3) modulates the leak of protons back into the mitochondrial matrix, thus controlling reactive oxygen species production. However, whether or not UCP3 glutathionylation is mediated enzymatically has remained unknown because previous work relied on the use of pharmacological agents, such as diamide, to alter the UCP3 glutathionylation state. Here, we demonstrate that glutaredoxin-2 (Grx2), a matrix oxidoreductase, is required to glutathionylate and inhibit UCP3. Analysis of bioenergetics in skeletal muscle mitochondria revealed that knock-out of Grx2 (Grx2(-/-)) increased proton leak in a UCP3-dependent manner. These effects were reversed using diamide, a glutathionylation catalyst. Importantly, the increased leak did not compromise coupled respiration. Knockdown of Grx2 augmented proton leak-dependent respiration in primary myotubes from wild type mice, an effect that was absent in UCP3(-/-) cells. These results confirm that Grx2 deactivates UCP3 by glutathionylation. To our knowledge, this is the first enzyme identified to regulate UCP3 by glutathionylation and is the first study on the role of Grx2 in the regulation of energy metabolism.
Collapse
Affiliation(s)
- Ryan J Mailloux
- Department of Biochemistry, Immunology, and Microbiology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Jian Ying Xuan
- Department of Biochemistry, Immunology, and Microbiology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Brittany Beauchamp
- Department of Biochemistry, Immunology, and Microbiology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Linda Jui
- Department of Biochemistry, Immunology, and Microbiology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Marjorie Lou
- Center of Redox Biology and School of Veterinary Medicine and Biomedical Sciences, University of Nebraska, Lincoln, Nebraska 68583
| | - Mary-Ellen Harper
- Department of Biochemistry, Immunology, and Microbiology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.
| |
Collapse
|