101
|
Nicolson GL, Ash ME. Lipid Replacement Therapy: a natural medicine approach to replacing damaged lipids in cellular membranes and organelles and restoring function. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1838:1657-79. [PMID: 24269541 DOI: 10.1016/j.bbamem.2013.11.010] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2013] [Revised: 10/30/2013] [Accepted: 11/09/2013] [Indexed: 12/14/2022]
Abstract
Lipid Replacement Therapy, the use of functional oral supplements containing cell membrane phospholipids and antioxidants, has been used to replace damaged, usually oxidized, membrane glycerophospholipids that accumulate during aging and in various clinical conditions in order to restore cellular function. This approach differs from other dietary and intravenous phospholipid interventions in the composition of phospholipids and their defense against oxidation during storage, ingestion, digestion and uptake as well as the use of protective molecules that noncovalently complex with phospholipid micelles and prevent their enzymatic and bile disruption. Once the phospholipids have been taken in by transport processes, they are protected by several natural mechanisms involving lipid receptors, transport and carrier molecules and circulating cells and lipoproteins until their delivery to tissues and cells where they can again be transferred to intracellular membranes by specific and nonspecific transport systems. Once delivered to membrane sites, they naturally replace and stimulate removal of damaged membrane lipids. Various chronic clinical conditions are characterized by membrane damage, mainly oxidative but also enzymatic, resulting in loss of cellular function. This is readily apparent in mitochondrial inner membranes where oxidative damage to phospholipids like cardiolipin and other molecules results in loss of trans-membrane potential, electron transport function and generation of high-energy molecules. Recent clinical trials have shown the benefits of Lipid Replacement Therapy in restoring mitochondrial function and reducing fatigue in aged subjects and patients with a variety of clinical diagnoses that are characterized by loss of mitochondrial function and include fatigue as a major symptom. This Article is Part of a Special Issue Entitled: Membrane Structure and Function: Relevance in the Cell's Physiology, Pathology and Therapy.
Collapse
Affiliation(s)
- Garth L Nicolson
- Department of Molecular Pathology, The Institute for Molecular Medicine, Huntington Beach, CA 92649, USA.
| | - Michael E Ash
- Clinical Education, Newton Abbot, Devon TQ12 4SG, UK
| |
Collapse
|
102
|
Cornelius C, Trovato Salinaro A, Scuto M, Fronte V, Cambria MT, Pennisi M, Bella R, Milone P, Graziano A, Crupi R, Cuzzocrea S, Pennisi G, Calabrese V. Cellular stress response, sirtuins and UCP proteins in Alzheimer disease: role of vitagenes. IMMUNITY & AGEING 2013; 10:41. [PMID: 24498895 PMCID: PMC3842652 DOI: 10.1186/1742-4933-10-41] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Accepted: 09/11/2013] [Indexed: 11/14/2022]
Abstract
Alzheimer’s Disease (AD) is a neurodegenerative disorder affecting up to one third of individuals reaching the age of 80. Different integrated responses exist in the brain to detect oxidative stress which is controlled by several genes termed Vitagenes. Vitagenes encode for cytoprotective heat shock proteins (Hsp), as well as thioredoxin, sirtuins and uncouple proteins (UCPs). In the present study we evaluate stress response mechanisms in plasma and lymphocytes of AD patients, as compared to controls, in order to provide evidence of an imbalance of oxidant/antioxidant mechanisms and oxidative damage in AD patients and the possible protective role of vitagenes. We found that the levels of Sirt-1 and Sirt-2 in AD lymphocytes were significantly higher than in control subjects. Interestingly, analysis of plasma showed in AD patients increased expression of Trx, a finding associated with reduced expression of UCP1, as compared to control group. This finding can open up new neuroprotective strategies, as molecules inducing this defense mechanisms can represent a therapeutic target to minimize the deleterious consequences associated to oxidative stress, such as in brain aging and neurodegenerative disorders.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Vittorio Calabrese
- Department of Biomedical Sciences, University of Catania, Catania, Italy.
| |
Collapse
|
103
|
Montgomery MK, Osborne B, Brown SHJ, Small L, Mitchell TW, Cooney GJ, Turner N. Contrasting metabolic effects of medium- versus long-chain fatty acids in skeletal muscle. J Lipid Res 2013; 54:3322-33. [PMID: 24078708 DOI: 10.1194/jlr.m040451] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Dietary intake of long-chain fatty acids (LCFAs) plays a causative role in insulin resistance and risk of diabetes. Whereas LCFAs promote lipid accumulation and insulin resistance, diets rich in medium-chain fatty acids (MCFAs) have been associated with increased oxidative metabolism and reduced adiposity, with few deleterious effects on insulin action. The molecular mechanisms underlying these differences between dietary fat subtypes are poorly understood. To investigate this further, we treated C2C12 myotubes with various LCFAs (16:0, 18:1n9, and 18:2n6) and MCFAs (10:0 and 12:0), as well as fed mice diets rich in LCFAs or MCFAs, and investigated fatty acid-induced changes in mitochondrial metabolism and oxidative stress. MCFA-treated cells displayed less lipid accumulation, increased mitochondrial oxidative capacity, and less oxidative stress than LCFA-treated cells. These changes were associated with improved insulin action in MCFA-treated myotubes. MCFA-fed mice exhibited increased energy expenditure, reduced adiposity, and better glucose tolerance compared with LCFA-fed mice. Dietary MCFAs increased respiration in isolated mitochondria, with a simultaneous reduction in reactive oxygen species generation, and subsequently low oxidative damage. Collectively our findings indicate that in contrast to LCFAs, MCFAs increase the intrinsic respiratory capacity of mitochondria without increasing oxidative stress. These effects potentially contribute to the beneficial metabolic actions of dietary MCFAs.
Collapse
Affiliation(s)
- Magdalene K Montgomery
- Department of Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | | | | | | | | | | | | |
Collapse
|
104
|
Muise ES, Souza S, Chi A, Tan Y, Zhao X, Liu F, Dallas-yang Q, Wu M, Sarr T, Zhu L, Guo H, Li Z, Li W, Hu W, Jiang G, Paweletz CP, Hendrickson RC, Thompson JR, Mu J, Berger JP, Mehmet H. Downstream signaling pathways in mouse adipose tissues following acute in vivo administration of fibroblast growth factor 21. PLoS One 2013; 8:e73011. [PMID: 24039848 PMCID: PMC3765203 DOI: 10.1371/journal.pone.0073011] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Accepted: 07/22/2013] [Indexed: 01/19/2023] Open
Abstract
FGF21 is a novel secreted protein with robust anti-diabetic, anti-obesity, and anti-atherogenic activities in preclinical species. In the current study, we investigated the signal transduction pathways downstream of FGF21 following acute administration of the growth factor to mice. Focusing on adipose tissues, we identified FGF21-mediated downstream signaling events and target engagement biomarkers. Specifically, RNA profiling of adipose tissues and phosphoproteomic profiling of adipocytes, following FGF21 treatment revealed several specific changes in gene expression and post-translational modifications, specifically phosphorylation, in several relevant proteins. Affymetrix microarray analysis of white adipose tissues isolated from both C57BL/6 (fed either regular chow or HFD) and db/db mice identified over 150 robust potential RNA transcripts and over 50 potential secreted proteins that were changed greater than 1.5 fold by FGF21 acutely. Phosphoprofiling analysis identified over 130 phosphoproteins that were modulated greater than 1.5 fold by FGF21 in 3T3-L1 adipocytes. Bioinformatic analysis of the combined gene and phosphoprotein profiling data identified a number of known metabolic pathways such as glucose uptake, insulin receptor signaling, Erk/Mapk signaling cascades, and lipid metabolism. Moreover, a number of novel events with hitherto unknown links to FGF21 signaling were observed at both the transcription and protein phosphorylation levels following treatment. We conclude that such a combined "omics" approach can be used not only to identify robust biomarkers for novel therapeutics but can also enhance our understanding of downstream signaling pathways; in the example presented here, novel FGF21-mediated signaling events in adipose tissue have been revealed that warrant further investigation.
Collapse
Affiliation(s)
- Eric S. Muise
- Discovery and Preclinical Sciences, Merck Research Laboratories, Merck Sharp & Dohme Corp., Whitehouse Station, New Jersey, United States of America
- * E-mail:
| | - Sandra Souza
- Discovery and Preclinical Sciences, Merck Research Laboratories, Merck Sharp & Dohme Corp., Whitehouse Station, New Jersey, United States of America
| | - An Chi
- Discovery and Preclinical Sciences, Merck Research Laboratories, Merck Sharp & Dohme Corp., Whitehouse Station, New Jersey, United States of America
| | - Yejun Tan
- Discovery and Preclinical Sciences, Merck Research Laboratories, Merck Sharp & Dohme Corp., Whitehouse Station, New Jersey, United States of America
| | - Xuemei Zhao
- Discovery and Preclinical Sciences, Merck Research Laboratories, Merck Sharp & Dohme Corp., Whitehouse Station, New Jersey, United States of America
| | - Franklin Liu
- Discovery and Preclinical Sciences, Merck Research Laboratories, Merck Sharp & Dohme Corp., Whitehouse Station, New Jersey, United States of America
| | - Qing Dallas-yang
- Discovery and Preclinical Sciences, Merck Research Laboratories, Merck Sharp & Dohme Corp., Whitehouse Station, New Jersey, United States of America
| | - Margaret Wu
- Discovery and Preclinical Sciences, Merck Research Laboratories, Merck Sharp & Dohme Corp., Whitehouse Station, New Jersey, United States of America
| | - Tim Sarr
- Discovery and Preclinical Sciences, Merck Research Laboratories, Merck Sharp & Dohme Corp., Whitehouse Station, New Jersey, United States of America
| | - Lan Zhu
- Discovery and Preclinical Sciences, Merck Research Laboratories, Merck Sharp & Dohme Corp., Whitehouse Station, New Jersey, United States of America
| | - Hongbo Guo
- Discovery and Preclinical Sciences, Merck Research Laboratories, Merck Sharp & Dohme Corp., Whitehouse Station, New Jersey, United States of America
| | - Zhihua Li
- Discovery and Preclinical Sciences, Merck Research Laboratories, Merck Sharp & Dohme Corp., Whitehouse Station, New Jersey, United States of America
| | - Wenyu Li
- Discovery and Preclinical Sciences, Merck Research Laboratories, Merck Sharp & Dohme Corp., Whitehouse Station, New Jersey, United States of America
| | - Weiwen Hu
- Discovery and Preclinical Sciences, Merck Research Laboratories, Merck Sharp & Dohme Corp., Whitehouse Station, New Jersey, United States of America
| | - Guoqiang Jiang
- Discovery and Preclinical Sciences, Merck Research Laboratories, Merck Sharp & Dohme Corp., Whitehouse Station, New Jersey, United States of America
| | - Cloud P. Paweletz
- Discovery and Preclinical Sciences, Merck Research Laboratories, Merck Sharp & Dohme Corp., Whitehouse Station, New Jersey, United States of America
| | - Ronald C. Hendrickson
- Discovery and Preclinical Sciences, Merck Research Laboratories, Merck Sharp & Dohme Corp., Whitehouse Station, New Jersey, United States of America
| | - John R. Thompson
- Discovery and Preclinical Sciences, Merck Research Laboratories, Merck Sharp & Dohme Corp., Whitehouse Station, New Jersey, United States of America
| | - James Mu
- Discovery and Preclinical Sciences, Merck Research Laboratories, Merck Sharp & Dohme Corp., Whitehouse Station, New Jersey, United States of America
| | - Joel P. Berger
- Discovery and Preclinical Sciences, Merck Research Laboratories, Merck Sharp & Dohme Corp., Whitehouse Station, New Jersey, United States of America
| | - Huseyin Mehmet
- Discovery and Preclinical Sciences, Merck Research Laboratories, Merck Sharp & Dohme Corp., Whitehouse Station, New Jersey, United States of America
| |
Collapse
|
105
|
Pung YF, Sam WJ, Hardwick JP, Yin L, Ohanyan V, Logan S, Di Vincenzo L, Chilian WM. The role of mitochondrial bioenergetics and reactive oxygen species in coronary collateral growth. Am J Physiol Heart Circ Physiol 2013; 305:H1275-80. [PMID: 23997092 DOI: 10.1152/ajpheart.00077.2013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Coronary collateral growth is a process involving coordination between growth factors expressed in response to ischemia and mechanical forces. Underlying this response is proliferation of vascular smooth muscle and endothelial cells, resulting in an enlargement in the caliber of arterial-arterial anastomoses, i.e., a collateral vessel, sometimes as much as an order of magnitude. An integral element of this cell proliferation is the process known as phenotypic switching in which cells of a particular phenotype, e.g., contractile vascular smooth muscle, must change their phenotype to proliferate. Phenotypic switching requires that protein synthesis occurs and different kinase signaling pathways become activated, necessitating energy to make the switch. Moreover, kinases, using ATP to phosphorylate their targets, have an energy requirement themselves. Mitochondria play a key role in the energy production that enables phenotypic switching, but under conditions where mitochondrial energy production is constrained, e.g., mitochondrial oxidative stress, this switch is impaired. In addition, we discuss the potential importance of uncoupling proteins as modulators of mitochondrial reactive oxygen species production and bioenergetics, as well as the role of AMP kinase as an energy sensor upstream of mammalian target of rapamycin, the master regulator of protein synthesis.
Collapse
Affiliation(s)
- Yuh Fen Pung
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio
| | | | | | | | | | | | | | | |
Collapse
|
106
|
Huber SM, Butz L, Stegen B, Klumpp D, Braun N, Ruth P, Eckert F. Ionizing radiation, ion transports, and radioresistance of cancer cells. Front Physiol 2013; 4:212. [PMID: 23966948 PMCID: PMC3743404 DOI: 10.3389/fphys.2013.00212] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 07/24/2013] [Indexed: 12/22/2022] Open
Abstract
The standard treatment of many tumor entities comprises fractionated radiation therapy which applies ionizing radiation to the tumor-bearing target volume. Ionizing radiation causes double-strand breaks in the DNA backbone that result in cell death if the number of DNA double-strand breaks exceeds the DNA repair capacity of the tumor cell. Ionizing radiation reportedly does not only act on the DNA in the nucleus but also on the plasma membrane. In particular, ionizing radiation-induced modifications of ion channels and transporters have been reported. Importantly, these altered transports seem to contribute to the survival of the irradiated tumor cells. The present review article summarizes our current knowledge on the underlying mechanisms and introduces strategies to radiosensitize tumor cells by targeting plasma membrane ion transports.
Collapse
Affiliation(s)
- Stephan M Huber
- Department of Radiation Oncology, University of Tübingen Tübingen, Germany
| | | | | | | | | | | | | |
Collapse
|
107
|
Anedda A, López-Bernardo E, Acosta-Iborra B, Saadeh Suleiman M, Landázuri MO, Cadenas S. The transcription factor Nrf2 promotes survival by enhancing the expression of uncoupling protein 3 under conditions of oxidative stress. Free Radic Biol Med 2013; 61:395-407. [PMID: 23597505 DOI: 10.1016/j.freeradbiomed.2013.04.007] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Revised: 04/01/2013] [Accepted: 04/05/2013] [Indexed: 10/27/2022]
Abstract
Uncoupling protein 3 (UCP3) is a member of the mitochondrial inner membrane carrier superfamily that modulates energy efficiency by catalyzing proton conductance and thus decreasing the production of superoxide anion. However, its role during oxidative stress and the underlying regulatory and molecular mechanisms remain poorly understood. We sought to investigate how UCP3 expression is regulated by oxidative stress and to evaluate the putative antioxidant role of this protein. H2O2 treatment increased UCP3 expression and the nuclear accumulation of the transcription factor Nrf2 in C2C12 and HL-1 cells. Nrf2 siRNA prevented H2O2-induced UCP3 expression, increasing oxidative stress and cell death. ChIP assays identified an antioxidant-response element (ARE) within the UCP3 promoter that bound Nrf2 after exposure to H2O2. Luciferase reporter experiments confirmed increased ARE activity in H2O2-treated HL-1 cells. Importantly, H2O2 increased the UCP3-mediated proton leak, suggesting a role for this protein in attenuating ROS-induced damage. Nrf2 nuclear accumulation and increased UCP3 protein were also detected in intact mouse heart subjected to a condition known to increase ROS generation. This is the first study to demonstrate that H2O2 augments UCP3 expression and it provides the first evidence of Nrf2 binding to the UCP3 promoter in response to oxidative challenge. These findings suggest that UCP3 functions as a member of the cellular antioxidant defense system that protects against oxidative stress in vivo. In conclusion, we have identified a novel regulatory process induced by an oxidative insult whereby the expression of the mitochondrial protein UCP3 is driven by the Nrf2 transcription factor, which decreases ROS production and prevents cell death.
Collapse
Affiliation(s)
- Andrea Anedda
- Servicio de Inmunología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IP), 28006 Madrid, Spain
| | - Elia López-Bernardo
- Servicio de Inmunología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IP), 28006 Madrid, Spain
| | - Bárbara Acosta-Iborra
- Servicio de Inmunología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IP), 28006 Madrid, Spain; Unidad de Investigación, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria Princesa (IP), 28009 Madrid, Spain
| | | | - Manuel O Landázuri
- Servicio de Inmunología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IP), 28006 Madrid, Spain; Unidad de Investigación, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria Princesa (IP), 28009 Madrid, Spain
| | - Susana Cadenas
- Servicio de Inmunología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IP), 28006 Madrid, Spain; Departamento de Biología Molecular, Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain.
| |
Collapse
|
108
|
Echtay KS, Brand MD. 4-Hydroxy-2-nonenal and uncoupling proteins: an approach for regulation of mitochondrial ROS production. Redox Rep 2013; 12:26-9. [PMID: 17263904 DOI: 10.1179/135100007x162158] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
One factor that has the potential to regulate reactive oxygen species (ROS) generation is the mild uncoupling of oxidative phosphorylation, i.e. proton (H(+)) leak across the mitochondrial inner membrane. Proton leak has been shown to attenuate ROS generation, whereas ROS and their derivatives (such as superoxide and hydroxynonenal) have been shown to induce H(+) leak through uncoupling proteins (UCPs). This suggests the existence of a feedback loop between ROS and H(+) leak mediated through UCPs. Although the physiological functions of the new UCPs, such as UCP2 and UCP3, are still not established, extensive data support the idea that these mitochondrial carrier proteins are involved in the control of ROS generation. The molecular basis of both ROS generation and hydroxynonenal-induced uncoupling through UCPs is reviewed and the consequences of their interaction for protection against excessive ROS production at the expense of energy production is discussed.
Collapse
Affiliation(s)
- Karim S Echtay
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Tripoli, Lebanon.
| | | |
Collapse
|
109
|
Thrush AB, Dent R, McPherson R, Harper ME. Implications of mitochondrial uncoupling in skeletal muscle in the development and treatment of obesity. FEBS J 2013; 280:5015-29. [DOI: 10.1111/febs.12399] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Revised: 06/03/2013] [Accepted: 06/17/2013] [Indexed: 12/13/2022]
Affiliation(s)
- A. Brianne Thrush
- Department of Biochemistry, Microbiology and Immunology; Faculty of Medicine; University of Ottawa; Ontario; Canada
| | - Robert Dent
- Ottawa Hospital Weight Management Clinic; Ottawa Hospital; Ontario; Canada
| | | | - Mary-Ellen Harper
- Department of Biochemistry, Microbiology and Immunology; Faculty of Medicine; University of Ottawa; Ontario; Canada
| |
Collapse
|
110
|
Perez-Leighton CE, Billington CJ, Kotz CM. Orexin modulation of adipose tissue. Biochim Biophys Acta Mol Basis Dis 2013; 1842:440-5. [PMID: 23791983 DOI: 10.1016/j.bbadis.2013.06.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2013] [Revised: 05/23/2013] [Accepted: 06/05/2013] [Indexed: 10/26/2022]
Abstract
The orexins are neuropeptides with critical functions in the central nervous system. These neuropeptides have important roles in energy balance and obesity, and therefore on the accumulation of adipose tissue. Rodents lacking orexins, typically through genetic knockouts, experience increased weight gain and accumulation of adipose tissue. Evidence indicates that the lack of the orexins increase adiposity as a result of decreased energy expenditure, principally through a reduction of physical activity. Different lines of evidence suggest that other mechanisms are likely also in play, and neural influences on both white and brown adipose tissues remain to be fully and functionally defined. In addition, the orexin peptides and their receptors are expressed in adipose tissue, with little available information as to their significance. This review summarizes our current understanding of how the orexin peptides affect adipose tissue. We provide a brief introduction to the physiology of orexins and their effects on white and brown adipose tissues in the context of energy balance. We conclude this review by integrating this information in the context of the known physiology of the orexins. This article is part of a Special Issue entitled: Modulation of Adipose Tissue in Health and Disease.
Collapse
Affiliation(s)
- Claudio E Perez-Leighton
- Veterans Health Care System, GRECC, One Veterans Drive, Minneapolis, MN 55417, USA; University of Minnesota, MN Obesity Center, 1334 Eckles Avenue, St Paul, MN 55108, USA; Center for Integrative Medicine and Innovative Sciences, Facultad de Medicina, Universidad Andres Bello, Echaurren 183, Santiago, 8370071, Chile.
| | - Charles J Billington
- Veterans Health Care System, Endocrinology, One Veterans Drive, Minneapolis, MN 55417, USA; University of Minnesota, MN Obesity Center, 1334 Eckles Avenue, St Paul, MN 55108, USA; University of Minnesota, Graduate Program in Nutrition, 1334 Eckles Avenue, St Paul, MN 55108, USA
| | - Catherine M Kotz
- Veterans Health Care System, GRECC, One Veterans Drive, Minneapolis, MN 55417, USA; University of Minnesota, MN Obesity Center, 1334 Eckles Avenue, St Paul, MN 55108, USA; University of Minnesota, Department of Food Science and Nutrition, 1334 Eckles Avenue, St Paul, MN 55108, USA; University of Minnesota, Graduate Program in Nutrition, 1334 Eckles Avenue, St Paul, MN 55108, USA; University of Minnesota, Graduate Program in Neuroscience, USA
| |
Collapse
|
111
|
Nagai N, Habuchi H, Sugaya N, Nakamura M, Imamura T, Watanabe H, Kimata K. Involvement of heparan sulfate 6-O-sulfation in the regulation of energy metabolism and the alteration of thyroid hormone levels in male mice. Glycobiology 2013; 23:980-92. [PMID: 23690091 DOI: 10.1093/glycob/cwt037] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Here, we report that male heparan sulfate 6-O-sulfotransferase-2 (Hs6st2) knockout mice showed increased body weight in an age-dependent manner even when fed with a normal diet and showed a phenotype of impaired glucose metabolism and insulin resistance. Quantitative reverse transcription-polymerase chain reaction (RT-PCR) analysis showed that the expression of mitochondrial uncoupling proteins Ucp1 and Ucp3 was reduced in the interscapular brown adipose tissue (BAT) of male Hs6st2 knockout mice, suggesting reduced energy metabolism. The serum level of thyroid-stimulating hormone was significantly higher and that of thyroxine was lower in the knockout mice. When cultures of brown adipocytes from wild-type and Hs6st2 knockout mice isolated and differentiated in vitro were treated with FGF19 (fibroblast growth factor 19) or FGF21 in the presence or the absence of heparitinase I, phosphorylation of p42/p44 mitogen-activated protein (MAP) kinase was reduced. Heparan sulfate (HS) 6-O-sulfation was reduced not only in BAT but also in the thyroid tissue of the knockout mice. Thus, 6-O-sulfation in HS seems to play an important role in mediating energy metabolism by controlling thyroid hormone levels and signals from the FGF19 subfamily proteins, and the alteration of the HS composition may result in metabolic syndrome phenotypes such as altered glucose and insulin tolerance.
Collapse
Affiliation(s)
- Naoko Nagai
- Institute for Molecular Science of Medicine, Aichi Medical University, Nagakute, Aichi, Japan
| | | | | | | | | | | | | |
Collapse
|
112
|
Perrino C, Schiattarella GG, Sannino A, Pironti G, Petretta MP, Cannavo A, Gargiulo G, Ilardi F, Magliulo F, Franzone A, Carotenuto G, Serino F, Altobelli GG, Cimini V, Cuocolo A, Lombardi A, Goglia F, Indolfi C, Trimarco B, Esposito G. Genetic deletion of uncoupling protein 3 exaggerates apoptotic cell death in the ischemic heart leading to heart failure. J Am Heart Assoc 2013; 2:e000086. [PMID: 23688674 PMCID: PMC3698767 DOI: 10.1161/jaha.113.000086] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND Uncoupling protein 3 (ucp3) is a member of the mitochondrial anion carrier superfamily of proteins uncoupling mitochondrial respiration. In this study, we investigated the effects of ucp3 genetic deletion on mitochondrial function and cell survival under low oxygen conditions in vitro and in vivo. METHODS AND RESULTS To test the effects of ucp3 deletion in vitro, murine embryonic fibroblasts and adult cardiomyocytes were isolated from wild-type (WT, n=67) and ucp3 knockout mice (ucp3(-/-), n=70). To test the effects of ucp3 genetic deletion in vivo, myocardial infarction (MI) was induced by permanent coronary artery ligation in WT and ucp3(-/-) mice. Compared with WT, ucp3(-/-) murine embryonic fibroblasts and cardiomyocytes exhibited mitochondrial dysfunction and increased mitochondrial reactive oxygen species generation and apoptotic cell death under hypoxic conditions in vitro (terminal deoxynucleotidyl transferase-dUTP nick end labeling-positive nuclei: WT hypoxia, 70.3 ± 1.2%; ucp3(-/-) hypoxia, 85.3 ± 0.9%; P<0.05). After MI, despite similar areas at risk in the 2 groups, ucp3(-/-) hearts demonstrated a significantly larger infarct size compared with WT (infarct area/area at risk: WT, 48.2 ± 3.7%; ucp3(-/-), 65.0 ± 2.9%; P<0.05). Eight weeks after MI, cardiac function was significantly decreased in ucp3(-/-) mice compared with WT (fractional shortening: WT MI, 42.7 ± 3.1%; ucp3(-/-) MI, 24.4 ± 2.9; P<0.05), and this was associated with heightened apoptotic cell death (terminal deoxynucleotidyl transferase-dUTP nick end labeling-positive nuclei: WT MI, 0.7 ± 0.04%; ucp3(-/-) MI, 1.1 ± 0.09%, P<0.05). CONCLUSIONS Our data indicate that ucp3 levels regulate reactive oxygen species levels and cell survival during hypoxia, modulating infarct size in the ischemic heart.
Collapse
Affiliation(s)
- Cinzia Perrino
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy (C.P., G.G.S., A.S., G.P., M.P.P., A.C., G.G., F.I., F.M., A.F., G.C., F.S., G.G.A., V.C., A.C., B.T., G.E.)
| | - Gabriele G. Schiattarella
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy (C.P., G.G.S., A.S., G.P., M.P.P., A.C., G.G., F.I., F.M., A.F., G.C., F.S., G.G.A., V.C., A.C., B.T., G.E.)
| | - Anna Sannino
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy (C.P., G.G.S., A.S., G.P., M.P.P., A.C., G.G., F.I., F.M., A.F., G.C., F.S., G.G.A., V.C., A.C., B.T., G.E.)
| | - Gianluigi Pironti
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy (C.P., G.G.S., A.S., G.P., M.P.P., A.C., G.G., F.I., F.M., A.F., G.C., F.S., G.G.A., V.C., A.C., B.T., G.E.)
| | - Maria Piera Petretta
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy (C.P., G.G.S., A.S., G.P., M.P.P., A.C., G.G., F.I., F.M., A.F., G.C., F.S., G.G.A., V.C., A.C., B.T., G.E.)
| | - Alessandro Cannavo
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy (C.P., G.G.S., A.S., G.P., M.P.P., A.C., G.G., F.I., F.M., A.F., G.C., F.S., G.G.A., V.C., A.C., B.T., G.E.)
| | - Giuseppe Gargiulo
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy (C.P., G.G.S., A.S., G.P., M.P.P., A.C., G.G., F.I., F.M., A.F., G.C., F.S., G.G.A., V.C., A.C., B.T., G.E.)
| | - Federica Ilardi
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy (C.P., G.G.S., A.S., G.P., M.P.P., A.C., G.G., F.I., F.M., A.F., G.C., F.S., G.G.A., V.C., A.C., B.T., G.E.)
| | - Fabio Magliulo
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy (C.P., G.G.S., A.S., G.P., M.P.P., A.C., G.G., F.I., F.M., A.F., G.C., F.S., G.G.A., V.C., A.C., B.T., G.E.)
| | - Anna Franzone
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy (C.P., G.G.S., A.S., G.P., M.P.P., A.C., G.G., F.I., F.M., A.F., G.C., F.S., G.G.A., V.C., A.C., B.T., G.E.)
| | - Giuseppe Carotenuto
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy (C.P., G.G.S., A.S., G.P., M.P.P., A.C., G.G., F.I., F.M., A.F., G.C., F.S., G.G.A., V.C., A.C., B.T., G.E.)
| | - Federica Serino
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy (C.P., G.G.S., A.S., G.P., M.P.P., A.C., G.G., F.I., F.M., A.F., G.C., F.S., G.G.A., V.C., A.C., B.T., G.E.)
| | - Giovanna G. Altobelli
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy (C.P., G.G.S., A.S., G.P., M.P.P., A.C., G.G., F.I., F.M., A.F., G.C., F.S., G.G.A., V.C., A.C., B.T., G.E.)
| | - Vincenzo Cimini
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy (C.P., G.G.S., A.S., G.P., M.P.P., A.C., G.G., F.I., F.M., A.F., G.C., F.S., G.G.A., V.C., A.C., B.T., G.E.)
| | - Alberto Cuocolo
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy (C.P., G.G.S., A.S., G.P., M.P.P., A.C., G.G., F.I., F.M., A.F., G.C., F.S., G.G.A., V.C., A.C., B.T., G.E.)
| | - Assunta Lombardi
- Department of Biology, Federico II University, Naples, Italy (A.L.)
| | - Fernando Goglia
- Department of Biology Sciences, Geology and Environment, Sannio University, Benevento, Italy (F.G.)
| | - Ciro Indolfi
- Department of Cardiology, Magna Graecia University, Catanzaro, Italy (C.I.)
| | - Bruno Trimarco
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy (C.P., G.G.S., A.S., G.P., M.P.P., A.C., G.G., F.I., F.M., A.F., G.C., F.S., G.G.A., V.C., A.C., B.T., G.E.)
| | - Giovanni Esposito
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy (C.P., G.G.S., A.S., G.P., M.P.P., A.C., G.G., F.I., F.M., A.F., G.C., F.S., G.G.A., V.C., A.C., B.T., G.E.)
- Correspondence to: Giovanni Esposito, MD, PhD, or Cinzia Perrino, MD, PhD, Division of Cardiology, Federico II University, Via Pansini 5, 80131 Naples, Italy. E‐mail: ,
| |
Collapse
|
113
|
Uncoupling protein 2 -866G/A and uncoupling protein 3 -55C/T polymorphisms in young South African Indian coronary artery disease patients. Gene 2013; 524:79-83. [PMID: 23639961 DOI: 10.1016/j.gene.2013.04.048] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Revised: 04/03/2013] [Accepted: 04/04/2013] [Indexed: 12/11/2022]
Abstract
BACKGROUND Uncoupling proteins (UCPs) 2 and 3 play an important role in the regulation of oxidative stress which contributes to chronic inflammation. Promoter polymorphisms of these genes have been linked to chronic diseases including heart disease and type II diabetes mellitus in several populations. This is the first investigation of the UCP2 -866G/A rs659366 and UCP3 -55C/T rs1800849 polymorphisms in young South African (SA) Indians with coronary artery disease (CAD). METHODS A total of 300 subjects were recruited into this study of which 100 were SA Indian males with CAD, 100 age- (range 24-45 years), gender- and race-matched controls and 100 age-matched black SA males. The frequency of the UCP2 -866G/A and UPC3 -55C/T genotypes was assessed by polymerase chain reaction restriction fragment length polymorphism (PCR-RFLP). RESULTS The heterozygous UCP2 -866G/A and homozygous UCP3 -55C/C genotypes occurred at highest frequency in CAD patients (60% and 64%, respectively) compared to SA Indian controls (52% and 63%) and SA Black controls (50% and 58%). The UCP2 -886G/A (OR=1.110; 95% CI=0.7438-1.655; p=0.6835) and UCP3 -55C/T (OR=0.788; 95% CI=0.482-1.289; p=0.382) polymorphisms did not influence the risk of CAD. The rare homozygous UCP3 -55T/T genotype was associated with highest fasting glucose (11.87 ± 3.7 mmol/L vs. C/C:6.11 ± 0.27 mmol/L and C/T:6.48 ± 0.57 mmol/L, p=0.0025), HbA1c (10.05 ± 2.57% vs. C/C:6.44 ± 0.21% and C/T:6.76 ± 0.35%, p=0.0006) and triglycerides (6.47 ± 1.7 mmol/L vs. C/C:2.33 ± 0.17 mmol/L and C/T:2.06 ± 0.25 mmol/L, p<0.0001) in CAD patients. CONCLUSION The frequency of the UCP2 -866G/A and UCP3 -55C/T polymorphisms was similar in our SA Indian and SA Black groups. The presence of the UCP2 -866G/A and UCP3 -55C/T polymorphisms does not influence the risk of CAD in young South African Indian CAD patients.
Collapse
|
114
|
Muller F. The nature and mechanism of superoxide production by the electron transport chain: Its relevance to aging. J Am Aging Assoc 2013; 23:227-53. [PMID: 23604868 DOI: 10.1007/s11357-000-0022-9] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Most biogerontologists agree that oxygen (and nitrogen) free radicals play a major role in the process of aging. The evidence strongly suggests that the electron transport chain, located in the inner mitochondrial membrane, is the major source of reactive oxygen species in animal cells. It has been reported that there exists an inverse correlation between the rate of superoxide/hydrogen peroxide production by mitochondria and the maximum longevity of mammalian species. However, no correlation or most frequently an inverse correlation exists between the amount of antioxidant enzymes and maximum longevity. Although overexpression of the antioxidant enzymes SOD1 and CAT (as well as SOD1 alone) have been successful at extending maximum lifespan in Drosophila, this has not been the case in mice. Several labs have overexpressed SOD1 and failed to see a positive effect on longevity. An explanation for this failure is that there is some level of superoxide damage that is not preventable by SOD, such as that initiated by the hydroperoxyl radical inside the lipid bilayer, and that accumulation of this damage is responsible for aging. I therefore suggest an alternative approach to testing the free radical theory of aging in mammals. Instead of trying to increase the amount of antioxidant enzymes, I suggest using molecular biology/transgenics to decrease the rate of superoxide production, which in the context of the free radical theory of aging would be expected to increase longevity. This paper aims to summarize what is known about the nature and mechanisms of superoxide production and what genes are involved in controlling the rate of superoxide production.
Collapse
Affiliation(s)
- F Muller
- Laboratory of David M. Kramer, Institute of Biological Chemistry, Washington State University, Pullman, WA 99164 USA
| |
Collapse
|
115
|
Ma CJ, Nie AF, Zhang ZJ, Zhang ZG, Du L, Li XY, Ning G. Genipin stimulates glucose transport in C2C12 myotubes via an IRS-1 and calcium-dependent mechanism. J Endocrinol 2013; 216:353-62. [PMID: 23257267 DOI: 10.1530/joe-11-0473] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Genipin, a compound derived from Gardenia jasminoides Ellis fruits, has been used over the years in traditional Chinese medicine to treat symptoms of type 2 diabetes. However, the molecular basis for its antidiabetic effect has not been fully revealed. In this study, we investigated the effects of genipin on glucose uptake and signaling pathways in C(2)C(12) myotubes. Our study demonstrates that genipin stimulated glucose uptake in a time- and dose-dependent manner. The maximal effect was achieved at 2 h with a concentration of 10 μM. In myotubes, genipin promoted glucose transporter 4 (GLUT4) translocation to the cell surface, which was observed by analyzing their distribution in subcellular membrane fraction, and increased the phosphorylation of insulin receptor substrate-1 (IRS-1), AKT, and GSK3β. Meanwhile, genipin increased ATP levels, closed K(ATP) channels, and then increased the concentration of calcium in the cytoplasm in C(2)C(12) myotubes. Genipin-stimulated glucose uptake could be blocked by both the PI3-K inhibitor wortmannin and calcium chelator EGTA. Moreover, genipin increases the level of reactive oxygen species and ATP in C(2)C(12) myotubes. These results suggest that genipin activates IRS-1, PI3-K, and downstream signaling pathway and increases concentrations of calcium, resulting in GLUT4 translocation and glucose uptake increase in C(2)C(12) myotubes.
Collapse
Affiliation(s)
- Chan-Juan Ma
- Shanghai Key Laboratory of Endocrine Tumor, Shanghai Jiao-Tong University School of Medicine, Shanghai Institute of Endocrinology and Metabolism, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Rui-Jin Hospital, 197 Rui-Jin 2nd Road, Shanghai 200025, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
116
|
Quintens R, Singh S, Lemaire K, De Bock K, Granvik M, Schraenen A, Vroegrijk IOCM, Costa V, Van Noten P, Lambrechts D, Lehnert S, Van Lommel L, Thorrez L, De Faudeur G, Romijn JA, Shelton JM, Scorrano L, Lijnen HR, Voshol PJ, Carmeliet P, Mammen PPA, Schuit F. Mice deficient in the respiratory chain gene Cox6a2 are protected against high-fat diet-induced obesity and insulin resistance. PLoS One 2013; 8:e56719. [PMID: 23460811 PMCID: PMC3584060 DOI: 10.1371/journal.pone.0056719] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 01/14/2013] [Indexed: 01/07/2023] Open
Abstract
Oxidative phosphorylation in mitochondria is responsible for 90% of ATP synthesis in most cells. This essential housekeeping function is mediated by nuclear and mitochondrial genes encoding subunits of complex I to V of the respiratory chain. Although complex IV is the best studied of these complexes, the exact function of the striated muscle-specific subunit COX6A2 is still poorly understood. In this study, we show that Cox6a2-deficient mice are protected against high-fat diet-induced obesity, insulin resistance and glucose intolerance. This phenotype results from elevated energy expenditure and a skeletal muscle fiber type switch towards more oxidative fibers. At the molecular level we observe increased formation of reactive oxygen species, constitutive activation of AMP-activated protein kinase, and enhanced expression of uncoupling proteins. Our data indicate that COX6A2 is a regulator of respiratory uncoupling in muscle and we demonstrate that a novel and direct link exists between muscle respiratory chain activity and diet-induced obesity/insulin resistance.
Collapse
Affiliation(s)
- Roel Quintens
- Gene Expression Unit, Department of Molecular and Cellular Medicine, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Sarvjeet Singh
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Katleen Lemaire
- Gene Expression Unit, Department of Molecular and Cellular Medicine, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Katrien De Bock
- Vesalius Research Center, Katholieke Universiteit Leuven, Leuven, Belgium
- Vesalius Research Center, Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium
| | - Mikaela Granvik
- Gene Expression Unit, Department of Molecular and Cellular Medicine, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Anica Schraenen
- Gene Expression Unit, Department of Molecular and Cellular Medicine, Katholieke Universiteit Leuven, Leuven, Belgium
| | | | - Veronica Costa
- Department of Cell Physiology and Metabolism, University of Geneva, Geneve, Switzerland
| | - Pieter Van Noten
- Physical Activity and Health Laboratory, Biomedical Kinesiology Department, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Dennis Lambrechts
- Department of Metallurgy and Materials Engineering, KU Leuven, Leuven, Belgium
| | - Stefan Lehnert
- Gene Expression Unit, Department of Molecular and Cellular Medicine, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Leentje Van Lommel
- Gene Expression Unit, Department of Molecular and Cellular Medicine, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Lieven Thorrez
- Gene Expression Unit, Department of Molecular and Cellular Medicine, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Geoffroy De Faudeur
- Gene Expression Unit, Department of Molecular and Cellular Medicine, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Johannes Anthonius Romijn
- Department of Endocrinology and Metabolic Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - John Michael Shelton
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Luca Scorrano
- Department of Cell Physiology and Metabolism, University of Geneva, Geneve, Switzerland
| | - Henri Roger Lijnen
- Center for Molecular and Vascular Biology, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Peter Jacobus Voshol
- Department of Endocrinology and Metabolic Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter Carmeliet
- Vesalius Research Center, Katholieke Universiteit Leuven, Leuven, Belgium
- Vesalius Research Center, Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium
| | - Pradeep Puthenveetil Abraham Mammen
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Frans Schuit
- Gene Expression Unit, Department of Molecular and Cellular Medicine, Katholieke Universiteit Leuven, Leuven, Belgium
- * E-mail:
| |
Collapse
|
117
|
Crosstalk between Oxidative Stress and SIRT1: Impact on the Aging Process. Int J Mol Sci 2013; 14:3834-59. [PMID: 23434668 PMCID: PMC3588074 DOI: 10.3390/ijms14023834] [Citation(s) in RCA: 322] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Revised: 01/25/2013] [Accepted: 01/29/2013] [Indexed: 12/16/2022] Open
Abstract
Increased oxidative stress has been associated with the aging process. However, recent studies have revealed that a low-level oxidative stress can even extend the lifespan of organisms. Reactive oxygen species (ROS) are important signaling molecules, e.g., being required for autophagic degradation. SIRT1, a class III protein deacetylase, is a crucial cellular survival protein, which is also involved in combatting oxidative stress. For instance, SIRT1 can stimulate the expression of antioxidants via the FoxO pathways. Moreover, in contrast to ROS, SIRT1 inhibits NF-κB signaling which is a major inducer of inflammatory responses, e.g., with inflammasome pathway. Recent studies have demonstrated that an increased level of ROS can both directly and indirectly control the activity of SIRT1 enzyme. For instance, ROS can inhibit SIRT1 activity by evoking oxidative modifications on its cysteine residues. Decreased activity of SIRT1 enhances the NF-κB signaling, which supports inflammatory responses. This crosstalk between the SIRT1 and ROS signaling provokes in a context-dependent manner a decline in autophagy and a low-grade inflammatory phenotype, both being common hallmarks of ageing. We will review the major mechanisms controlling the signaling balance between the ROS production and SIRT1 activity emphasizing that this crosstalk has a crucial role in the regulation of the aging process.
Collapse
|
118
|
Shi Y, Pulliam DA, Liu Y, Hamilton RT, Jernigan AL, Bhattacharya A, Sloane LB, Qi W, Chaudhuri A, Buffenstein R, Ungvari Z, Austad SN, Van Remmen H. Reduced mitochondrial ROS, enhanced antioxidant defense, and distinct age-related changes in oxidative damage in muscles of long-lived Peromyscus leucopus. Am J Physiol Regul Integr Comp Physiol 2013; 304:R343-55. [PMID: 23325454 DOI: 10.1152/ajpregu.00139.2012] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Comparing biological processes in closely related species with divergent life spans is a powerful approach to study mechanisms of aging. The oxidative stress hypothesis of aging predicts that longer-lived species would have lower reactive oxygen species (ROS) generation and/or an increased antioxidant capacity, resulting in reduced oxidative damage with age than in shorter-lived species. In this study, we measured ROS generation in the young adult animals of the long-lived white-footed mouse, Peromyscus leucopus (maximal life span potential, MLSP = 8 yr) and the common laboratory mouse, Mus musculus (C57BL/6J strain; MLSP = 3.5 yr). Consistent with the hypothesis, our results show that skeletal muscle mitochondria from adult P. leucopus produce less ROS (superoxide and hydrogen peroxide) compared with M. musculus. Additionally, P. leucopus has an increase in the activity of antioxidant enzymes superoxide dismutase 1, catalase, and glutathione peroxidase 1 at young age. P. leucopus compared with M. musculus display low levels of lipid peroxidation (isoprostanes) throughout life; however, P. leucopus although having elevated protein carbonyls at a young age, the accrual of protein oxidation with age is minimal in contrast to the linear increase in M. musculus. Altogether, the results from young animals are in agreement with the predictions of the oxidative stress hypothesis of aging with the exception of protein carbonyls. Nonetheless, the age-dependent increase in protein carbonyls is more pronounced in short-lived M. musculus, which supports enhanced protein homeostasis in long-lived P. leucopus.
Collapse
Affiliation(s)
- Yun Shi
- Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX 78245, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
119
|
Montez P, Vázquez-Medina JP, Rodríguez R, Thorwald MA, Viscarra JA, Lam L, Peti-Peterdi J, Nakano D, Nishiyama A, Ortiz RM. Angiotensin receptor blockade recovers hepatic UCP2 expression and aconitase and SDH activities and ameliorates hepatic oxidative damage in insulin resistant rats. Endocrinology 2012; 153:5746-59. [PMID: 23087176 PMCID: PMC3512060 DOI: 10.1210/en.2012-1390] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Metabolic syndrome (MetS) is commonly associated with elevated renin-angiotensin system, oxidative stress, and steatohepatitis with down-regulation of uncoupling proteins (UCPs). However, the mechanisms linking renin-angiotensin system, steatosis, and UCP2 to hepatic oxidative damage during insulin resistance are not described. To test the hypothesis that angiotensin receptor activation contributes to decreased hepatic UCP2 expression and aconitase activity and to increased oxidative damage after increased glucose intake in a model of MetS, lean and obese Long Evans rats (n = 10/group) were randomly assigned to the following groups: 1) untreated Long Evans Tokushima Otsuka (lean, strain control), 2) untreated Otsuka Long Evans Tokushima Fatty (OLETF) (MetS model), 3) OLETF + angiotensin receptor blocker (ARB) (10 mg olmesartan/kg·d × 6 wk), 4) OLETF + high glucose (HG) (5% in drinking water × 6 wk), and 5) OLETF + ARB + HG (ARB/HG × 6 wk). HG increased body mass (37%), plasma triglycerides (TGs) (35%), plasma glycerol (87%), plasma free fatty acids (28%), and hepatic nitrotyrosine (74%). ARB treatment in HG decreased body mass (12%), plasma TG (15%), plasma glycerol (23%), plasma free fatty acids (14%), and hepatic TG content (42%), suggesting that angiotensin receptor type 1 (AT1) activation and increased adiposity contribute to the development of obesity-related dyslipidemia. ARB in HG also decreased hepatic nitrotyrosine and increased hepatic UCP2 expression (59%) and aconitase activity (40%), as well as antioxidant enzyme activities (50-120%), suggesting that AT1 activation also contributes to protein oxidation, impaired lipid metabolism, and antioxidant metabolism in the liver. Thus, in addition to promoting obesity-related hypertension, AT1 activation may also impair lipid metabolism and antioxidant capacity, resulting in steatosis via decreased UCP2 and tricarboxylic acid cycle activity.
Collapse
Affiliation(s)
- Priscilla Montez
- Department of Molecular and Cellular Biology, University of California, Merced, CA 95343, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
120
|
Befroy DE, Rothman DL, Petersen KF, Shulman GI. ³¹P-magnetization transfer magnetic resonance spectroscopy measurements of in vivo metabolism. Diabetes 2012; 61:2669-78. [PMID: 23093656 PMCID: PMC3478545 DOI: 10.2337/db12-0558] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Magnetic resonance spectroscopy offers a broad range of noninvasive analytical methods for investigating metabolism in vivo. Of these, the magnetization-transfer (MT) techniques permit the estimation of the unidirectional fluxes associated with metabolic exchange reactions. Phosphorus (³¹P) MT measurements can be used to examine the bioenergetic reactions of the creatine-kinase system and the ATP synthesis/hydrolysis cycle. Observations from our group and others suggest that the inorganic phosphate (P(i)) → ATP flux in skeletal muscle may be modulated by certain conditions, including aging, insulin resistance, and diabetes, and may reflect inherent alterations in mitochondrial metabolism. However, such effects on the P(i) → ATP flux are not universally observed under conditions in which mitochondrial function, assessed by other techniques, is impaired, and recent articles have raised concerns about the absolute magnitude of the measured reaction rates. As the application of ³¹P-MT techniques becomes more widespread, this article reviews the methodology and outlines our experience with its implementation in a variety of models in vivo. Also discussed are potential limitations of the technique, complementary methods for assessing oxidative metabolism, and whether the P(i) → ATP flux is a viable biomarker of metabolic function in vivo.
Collapse
Affiliation(s)
- Douglas E Befroy
- Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, CT, USA.
| | | | | | | |
Collapse
|
121
|
Mostyn A, Bos PM, Litten JC, Laws J, Symonds ME, Clarke L. Differential effects of thyroid hormone manipulation and beta adrenoceptor agonist administration on uncoupling protein mRNA abundance in adipose tissue and thermoregulation in neonatal pigs. Organogenesis 2012; 4:182-7. [PMID: 19279731 DOI: 10.4161/org.4.3.6505] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2008] [Accepted: 05/31/2008] [Indexed: 01/28/2023] Open
Abstract
We have shown that there is significant disparity in the expression of uncoupling proteins (UCP) 2 and 3 between modern-commercial and ancient-Meishan porcine genotypes, commercial pigs also have higher plasma triiodothyronine (T(3)) in on the first day of life. T(3) and the sympathetic nervous system are both known to regulate UCPs in rodents and humans; their role in regulating these proteins in the pig is unknown. This study examined whether thyroid hormone manipulation or administration of a selective beta3 adrenoceptor agonist (ZD) influenced plasma hormones, colonic temperature and UCP expression in adipose tissue of two breeds of pig. To mimic the differences observed in thyroid hormone status, piglets from Meishan and commercial litters were randomly assigned to control (1 ml/kg water), T(3) (10 mg/kg) (Meishan only), methimazole (a commonly used antithyroid drug) (50 mg/kg) (commercial only) or ZD (10 mg/kg) oral administration for the first 4 days of postnatal life. Adipose tissue UCP2/3 mRNA abundance was measured on day 4 using PCR. T(3) administration raised plasma T(3) concentrations and increased colonic temperature on day 4. UCP3 mRNA abundance was higher in Meishan, than commercial piglets (p = 0.042) and was downregulated following T(3) administration (p = 0.014). Irrespective of genotype, ZD increased UCP2 mRNA abundance (Meishan p = 0.05, commercial p = 0.03). Expression of neither UCP2 nor 3 was related to colonic temperature, regardless of treatment. In conclusion, we have demonstrated a dissociation between thyroid hormones and the sympathetic nervous system in the regulation of UCPs in porcine adipose tissue. We have also suggested that expression of adipose tissue UCP2 and 3 are not related to body temperature in piglets.
Collapse
Affiliation(s)
- Alison Mostyn
- Centre for Reproduction and Early Life; Institute of Clinical Research; University Hospital; Nottingham United Kingdom
| | | | | | | | | | | |
Collapse
|
122
|
Mitochondria-targeted antioxidants and metabolic modulators as pharmacological interventions to slow ageing. Biotechnol Adv 2012; 31:563-92. [PMID: 23022622 DOI: 10.1016/j.biotechadv.2012.09.005] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Revised: 09/19/2012] [Accepted: 09/21/2012] [Indexed: 02/07/2023]
Abstract
Populations in many nations today are rapidly ageing. This unprecedented demographic change represents one of the main challenges of our time. A defining property of the ageing process is a marked increase in the risk of mortality and morbidity with age. The incidence of cancer, cardiovascular and neurodegenerative diseases increases non-linearly, sometimes exponentially with age. One of the most important tasks in biogerontology is to develop interventions leading to an increase in healthy lifespan (health span), and a better understanding of basic mechanisms underlying the ageing process itself may lead to interventions able to delay or prevent many or even all age-dependent conditions. One of the putative basic mechanisms of ageing is age-dependent mitochondrial deterioration, closely associated with damage mediated by reactive oxygen species (ROS). Given the central role that mitochondria and mitochondrial dysfunction play not only in ageing but also in apoptosis, cancer, neurodegeneration and other age-related diseases there is great interest in approaches to protect mitochondria from ROS-mediated damage. In this review, we explore strategies of targeting mitochondria to reduce mitochondrial oxidative damage with the aim of preventing or delaying age-dependent decline in mitochondrial function and some of the resulting pathologies. We discuss mitochondria-targeted and -localized antioxidants (e.g.: MitoQ, SkQ, ergothioneine), mitochondrial metabolic modulators (e.g. dichloroacetic acid), and uncouplers (e.g.: uncoupling proteins, dinitrophenol) as well as some alternative future approaches for targeting compounds to the mitochondria, including advances from nanotechnology.
Collapse
|
123
|
Porter RK. Studies on the function and regulation of mitochondrial uncoupling proteins. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 748:171-84. [PMID: 22729858 DOI: 10.1007/978-1-4614-3573-0_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
Mitochondrial uncoupling proteins are members of the SLC25 family of solute carriers. Models of mitochondrial transporter function predict that uncoupling proteins are solute carriers. Evidence in the literature suggests that uncoupling proteins can transport protons, fatty acid anions, chloride anions, and recently the dicarboxylate succinate. Studies have also demonstrated that UCPs can be covalently modified and in some instances this covalent modification is needed to affect uncoupling function. The current evidence from functional analyses of mammalian uncoupling proteins is summarized in this chapter.
Collapse
Affiliation(s)
- Richard K Porter
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
124
|
Hesselink MKC, Mensink M, Schrauwen P. Human Uncoupling Protein-3 and Obesity: An Update. ACTA ACUST UNITED AC 2012; 11:1429-43. [PMID: 14694206 DOI: 10.1038/oby.2003.192] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The cloning of the uncoupling protein (UCP)1 homologs UCP2 and UCP3 has raised considerable interest in the mechanism. The expression of UCP3 mainly in skeletal muscle mitochondria and the potency of the skeletal muscle as a thermogenic organ made UCP3 an attractive target for studies toward manipulation of energy expenditure to fight disorders such as obesity and type 2 diabetes. Overexpressing UCP3 in mice resulted in lean, hyperphagic mice. However, the lack of an apparent phenotype in mice lacking UCP3 triggered the search for alternative functions of UCP3. The observation that fatty acid levels significantly affect UCP3 expression has given UCP3 a position in fatty acid handling and/or oxidation. Emerging data indicate that the primary physiological role of UCP3 may be the mitochondrial handling of fatty acids rather than the regulation of energy expenditure through thermogenesis. It has been proposed that UCP3 functions to export fatty acid anions away from the mitochondrial matrix. In doing so, fatty acids are exchanged with protons, explaining the uncoupling activity of UCP3. The exported fatty acid anions may originate from hydrolysis of fatty acid esters by a mitochondrial thioesterase, or they may have entered the mitochondria as nonesterified fatty acids by incorporating into and flip-flopping across the mitochondrial inner membrane. Regardless of the origin of the fatty acid anions, this putative function of UCP3 might be of great importance in protecting mitochondria against fatty acid accumulation and may help to maintain muscular fat oxidative capacity.
Collapse
Affiliation(s)
- Matthijs K C Hesselink
- Department of Movement Sciences, Maastricht University, 6200 MD Maastricht, The Netherlands.
| | | | | |
Collapse
|
125
|
Liu D, Huang L, Wang Y, Wang W, Wehrens XH, Belousova T, Abdelrahim M, DiMattia G, Sheikh-Hamad D. Human stanniocalcin-1 suppresses angiotensin II-induced superoxide generation in cardiomyocytes through UCP3-mediated anti-oxidant pathway. PLoS One 2012; 7:e36994. [PMID: 22693564 PMCID: PMC3365029 DOI: 10.1371/journal.pone.0036994] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Accepted: 04/11/2012] [Indexed: 01/30/2023] Open
Abstract
Rationale We have previously shown increased cardiac stanniocalcin-1 (STC1) in patients with idiopathic dilated cardiomyopathy. STC1 localizes to the inner mitochondrial membrane and transgenic over-expression of STC1 is associated with increased energy utilization. Objective We examined the hypothesis that STC1 uncouples mitochondrial oxidative phosphorylation - to suppress superoxide generation and modulate neurohormonal effects on cardiomyocytes. Methods and Results Compared to WT mouse heart, STC1 Tg heart displays: 2-fold higher uncoupling protein 3 (UCP3) levels, but no effect on UCP2 protein; 40% lower ATP levels; but similar activities of respiratory chain complexes I-IV. In cultured adult rat and freshly-isolated mouse cardiomyocytes, rSTC1 induces UCP3, but not UCP2. Treatment of cardiomyocytes with STC1 decreases mitochondrial membrane potential and suppresses baseline and angiotensin II (Ang II)-induced superoxide generation. Furthermore, baseline superoxide generation is higher in freshly-isolated adult UCP3−/− mouse cardiomyocytes compared to WT, suggesting an important role for UCP3 in regulating cardiomyocyte ROS under physiologic conditions. Treatment of UCP3−/− cardiomyocytes with rSTC1 failed to suppress superoxide generation, suggesting that the effects of STC1 on superoxide generation in cardiomyocytes are UCP3-dependent. Conclusion STC1 activates a novel anti-oxidant pathway in cardiac myocytes through induction of UCP3, and may play an important role in suppressing ROS in the heart under normal physiologic conditions and ameliorate the deleterious effects of Ang II-mediated cardiac injury. Importantly, our data point to a critical role for the mitochondria in regulating ROS generation in response to Ang II.
Collapse
Affiliation(s)
- Dajun Liu
- Division of Nephrology/Department of Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Luping Huang
- Division of Nephrology/Department of Medicine, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail: (DSH); (LH)
| | - Yanlin Wang
- Division of Nephrology/Department of Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Wei Wang
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Xander H.T. Wehrens
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Tatiana Belousova
- Division of Nephrology/Department of Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Maen Abdelrahim
- Cancer Research Institute, MD Anderson Cancer Center Orlando, Orlando, Florida, United States of America
| | - Gabriel DiMattia
- Departments of Oncology and Biochemistry, The University of Western Ontario, London, Ontario, Canada
| | - David Sheikh-Hamad
- Division of Nephrology/Department of Medicine, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail: (DSH); (LH)
| |
Collapse
|
126
|
Majerczak J, Korostynski M, Nieckarz Z, Szkutnik Z, Duda K, Zoladz JA. Endurance training decreases the non-linearity in the oxygen uptake-power output relationship in humans. Exp Physiol 2012; 97:386-99. [DOI: 10.1113/expphysiol.2011.062992] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
127
|
Dhamrait SS, Williams AG, Day SH, Skipworth J, Payne JR, World M, Humphries SE, Montgomery HE. Variation in the uncoupling protein 2 and 3 genes and human performance. J Appl Physiol (1985) 2012; 112:1122-7. [PMID: 22241057 DOI: 10.1152/japplphysiol.00766.2011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Uncoupling proteins 2 and 3 (UCP2 and UCP3) may negatively regulate mitochondrial ATP synthesis and, through this, influence human physical performance. However, human data relating to both these issues remain sparse. Examining the association of common variants in the UCP3/2 locus with performance phenotypes offers one means of investigation. The efficiency of skeletal muscle contraction, delta efficiency (DE), was assessed by cycle ergometry in 85 young, healthy, sedentary adults both before and after a period of endurance training. Of these, 58 were successfully genotyped for the UCP3-55C>T (rs1800849) and 61 for the UCP2-866G>A (rs659366) variant. At baseline, UCP genotype was unrelated to any physical characteristic, including DE. However, the UCP2-866G>A variant was independently and strongly associated with the DE response to physical training, with UCP2-866A allele carriers exhibiting a greater increase in DE with training (absolute change in DE of -0.2 ± 3.6% vs. 1.7 ± 2.8% vs. 2.3 ± 3.7% for GG vs. GA vs. AA, respectively; P = 0.02 for A allele carriers vs. GG homozygotes). In multivariate analysis, there was a significant interaction between UCP2-866G>A and UCP3-55C>T genotypes in determining changes in DE (adjusted R(2) = 0.137; P value for interaction = 0.003), which was independent of the effect of either single polymorphism or baseline characteristics. In conclusion, common genetic variation at the UCP3/2 gene locus is associated with training-related improvements in DE, an index of skeletal muscle performance. Such effects may be mediated through differences in the coupling of mitochondrial energy transduction in human skeletal muscle, but further mechanistic studies are required to delineate this potential role.
Collapse
Affiliation(s)
- Sukhbir S Dhamrait
- Centre for Cardiovascular Genetics, British Heart Foundation Laboratories, Royal Free & University College London Medical School, London.
| | | | | | | | | | | | | | | |
Collapse
|
128
|
Sluse FE. Uncoupling proteins: molecular, functional, regulatory, physiological and pathological aspects. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 942:137-56. [PMID: 22399421 DOI: 10.1007/978-94-007-2869-1_6] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Uncoupling proteins are a subfamily of the mitochondrial anion carrier family. They are widespread in the whole eukaryotic world with a few exceptions and present tissue specific isoforms in higher organisms. They mediate purine nucleotide-sensitive free fatty acid-activated proton inward flux through the inner mitochondrial membrane. This proton flux occurs at the expense of the proton motive force build up by the respiration and weakens the coupling between respiration and ATP synthesis. In this chapter we describe current and reliable knowledge of uncoupling proteins. A new methodology allowing study of their activity and regulation during phosphorylating respiration is described. It has entitled us to assert that all uncoupling proteins share common mechanisms of activation and regulation. This is of the utmost importance in order to understand the physiological roles of UCPs as well as their participation in pathological processes since every role of the UCPs in every cell is an integral part of their function and regulation. The central role of reduction level of ubiquinone in the control of their regulation is well-argued. Their potential and reliable roles in thermogenesis, reactive oxygen species prevention and energy flow are discussed as well as their role in some pathological disorders.
Collapse
Affiliation(s)
- Francis E Sluse
- Department of Life Sciences, University of Liege, Liege, Belgium.
| |
Collapse
|
129
|
Nukitrangsan N, Okabe T, Toda T, Inafuku M, Iwasaki H, Oku H. Effect of Peucedanum japonicum Thunb Extract on High-fat Diet-induced Obesity and Gene Expression in Mice. J Oleo Sci 2012; 61:89-101. [DOI: 10.5650/jos.61.89] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
130
|
Rose G, Crocco P, De Rango F, Montesanto A, Passarino G. Further support to the uncoupling-to-survive theory: the genetic variation of human UCP genes is associated with longevity. PLoS One 2011; 6:e29650. [PMID: 22216339 PMCID: PMC3246500 DOI: 10.1371/journal.pone.0029650] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2011] [Accepted: 12/02/2011] [Indexed: 12/26/2022] Open
Abstract
In humans Uncoupling Proteins (UCPs) are a group of five mitochondrial inner membrane transporters with variable tissue expression, which seem to function as regulators of energy homeostasis and antioxidants. In particular, these proteins uncouple respiration from ATP production, allowing stored energy to be released as heat. Data from experimental models have previously suggested that UCPs may play an important role on aging rate and lifespan. We analyzed the genetic variability of human UCPs in cohorts of subjects ranging between 64 and 105 years of age (for a total of 598 subjects), to determine whether specific UCP variability affects human longevity. Indeed, we found that the genetic variability of UCP2, UCP3 and UCP4 do affect the individual's chances of surviving up to a very old age. This confirms the importance of energy storage, energy use and modulation of ROS production in the aging process. In addition, given the different localization of these UCPs (UCP2 is expressed in various tissues including brain, hearth and adipose tissue, while UCP3 is expressed in muscles and Brown Adipose Tissue and UCP4 is expressed in neuronal cells), our results may suggest that the uncoupling process plays an important role in modulating aging especially in muscular and nervous tissues, which are indeed very responsive to metabolic alterations and are very important in estimating health status and survival in the elderly.
Collapse
Affiliation(s)
- Giuseppina Rose
- Department of Cell Biology, University of Calabria, Rende, Italy
| | - Paolina Crocco
- Department of Cell Biology, University of Calabria, Rende, Italy
| | | | | | | |
Collapse
|
131
|
Gamboa JL, Andrade FH. Muscle endurance and mitochondrial function after chronic normobaric hypoxia: contrast of respiratory and limb muscles. Pflugers Arch 2011; 463:327-38. [PMID: 22113781 DOI: 10.1007/s00424-011-1057-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Revised: 10/25/2011] [Accepted: 10/30/2011] [Indexed: 11/29/2022]
Abstract
Skeletal muscle adaptation to chronic hypoxia includes loss of oxidative capacity and decrease in fiber size. However, the diaphragm may adapt differently since its activity increases in response to hypoxia. Thus, we hypothesized that chronic hypoxia would not affect endurance, mitochondrial function, or fiber size in the mouse diaphragm. Adult male mice were kept in normoxia (control) or hypoxia (hypoxia, FIO(2) = 10%) for 4 weeks. After that time, muscles were collected for histological, biochemical, and functional analyses. Hypoxia soleus muscles fatigued faster (fatigue index higher in control, 21.5 ± 2.6% vs. 13.4 ± 2.4%, p < 0.05), but there was no difference between control and hypoxia diaphragm bundles. Mean fiber cross-sectional area was unchanged in hypoxia limb muscles, but it was 25% smaller in diaphragm (p < 0.001). Ratio of capillary length contact to fiber perimeter was significantly higher in hypoxia diaphragm (28.6 ± 1.2 vs. 49.3 ± 1.4, control and hypoxia, p < 0.001). Mitochondrial respiration rates in hypoxia limb muscles were lower: state 2 decreased 19%, state 3 31%, and state 4 18% vs. control, p < 0.05 for all comparisons. There were similar changes in hypoxia diaphragm: state 3 decreased 29% and state 4 17%, p < 0.05. After 4 weeks of hypoxia, limb muscle mitochondria had lower content of complex IV (cytochrome c oxidase), while diaphragm mitochondria had higher content of complexes IV and V (F (1)/F (0) ATP synthase) and less uncoupling protein 3 (UCP-3). These data demonstrate that diaphragm retains its endurance during chronic hypoxia, apparently due to a combination of morphometric changes and optimization of mitochondrial energy production.
Collapse
Affiliation(s)
- Jorge L Gamboa
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA.
| | | |
Collapse
|
132
|
Significance of uncoupling protein 3 in mitochondrial function upon mid- and long-term dietary high-fat exposure. FEBS Lett 2011; 585:4010-7. [PMID: 22115550 DOI: 10.1016/j.febslet.2011.11.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2011] [Revised: 10/07/2011] [Accepted: 11/08/2011] [Indexed: 02/07/2023]
Abstract
Uncoupling protein 3 (UCP3) may reduce mitochondrial ROS production, and thereby protect against mitochondrial dysfunction in skeletal muscle. UCP3 has been suggested to specifically fulfill this role under high-fat conditions. Here we show that UCP3 knockout mice indeed have elevated mitochondrial ROS production after short-term (8 weeks) high-fat feeding. After 26 weeks of high-fat feeding, UCP3 knockout mice exhibited reduced mitochondrial function as measured ex vivo in isolated mitochondria. In conclusion, these data suggest that UCP3 may have a role in the protection of mitochondria against lipid-induced mitochondrial dysfunction, but only after long-term exposure to high-fat.
Collapse
|
133
|
Ninomiya M, Shirabe K, Shimada M, Terashi T, Maehara Y. Role of UCP2 expression after hepatic warm ischemia-reperfusion in the rat. Gut Liver 2011; 5:486-92. [PMID: 22195248 PMCID: PMC3240793 DOI: 10.5009/gnl.2011.5.4.486] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2010] [Revised: 04/04/2011] [Accepted: 05/06/2011] [Indexed: 11/22/2022] Open
Abstract
Background/Aims The role of uncoupling protein-2 (UCP2) in the liver is currently unclear. Emerging evidence suggests a relationship between UCP2 and oxidative stress. In the present study, we tested the hypothesis that UCP2 expression in the liver might change during warm ischemia-reperfusion (I/R) according to oxidative stress. Methods Wistar rats were subjected to 40 (short ischemia) or 90 (long ischemia) minutes of partial lobar ischemia followed by 4 hours of reperfusion. UCP2 expression in the ischemic and nonischemic lobes was assessed using reverse transcription-polymerase chain reaction and immunohistochemistry. Malondialdehyde concentrations in the liver tissue were also compared. Results Malondialdehyde concentrations in the ischemic lobes were significantly higher in the long ischemia group. In the ischemic lobes of the short ischemia group, UCP2 protein expression was induced in hepatocytes, which did not express the protein prior to treatment, and the expression levels were higher than in the long ischemia group. The intralobular distribution of UCP2 seemed to correlate inversely with that of the necrotic area. UCP2 expression was observed, even in nonischemic lobes with similar intralobular heterogeneity. Conclusions UCP2 was induced in hepatocytes after warm I/R. Although the primitive role of UCP2 expression may be cytoprotective in nature, its actual protective effect in hepatic I/R may be minimal
Collapse
Affiliation(s)
- Mizuki Ninomiya
- Department of Surgery and Science, Kyushu University, Fukuoka Graduate School of Medical Sciences, Fukuoka, Japan
| | | | | | | | | |
Collapse
|
134
|
Hirasaka K, Lago CU, Kenaston MA, Fathe K, Nowinski SM, Nikawa T, Mills EM. Identification of a redox-modulatory interaction between uncoupling protein 3 and thioredoxin 2 in the mitochondrial intermembrane space. Antioxid Redox Signal 2011; 15:2645-61. [PMID: 21619484 PMCID: PMC3183655 DOI: 10.1089/ars.2011.3888] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
UNLABELLED Uncoupling protein 3 (UCP3) is a member of the mitochondrial solute carrier superfamily that is enriched in skeletal muscle and controls mitochondrial reactive oxygen species (ROS) production, but the mechanisms underlying this function are unclear. AIMS The goal of this work focused on the identification of mechanisms underlying UCP3 functions. RESULTS Here we report that the N-terminal, intermembrane space (IMS)-localized hydrophilic domain of mouse UCP3 interacts with the N-terminal mitochondrial targeting signal of thioredoxin 2 (Trx2), a mitochondrial thiol reductase. Cellular immunoprecipitation and in vitro pull-down assays show that the UCP3-Trx2 complex forms directly, and that the Trx2 N-terminus is both necessary and sufficient to confer UCP3 binding. Mutation studies show that neither a catalytically inactivated Trx2 mutant, nor a mutant Trx2 bearing the N-terminal targeting sequence of cytochrome c oxidase (COXMTS-Trx2) bind UCP3. Biochemical analyses using permeabilized mitochondria, and live cell experiments using bimolecular fluorescence complementation show that the UCP3-Trx2 complex forms specifically in the IMS. Finally, studies in C2C12 myocytes stably overexpressing UCP3 (2.5-fold) and subjected to Trx2 knockdown show that Trx2 is required for the UCP3-dependent mitigation of complex III-driven mitochondrial ROS generation. UCP3 expression was increased in mice fed a high fat diet, leading to increased localization of Trx2 to the IMS. UCP3 overexpression also increased expression of the glucose transporter GLUT4 in a Trx2-dependent fashion. INNOVATION This is the first report of a mitochondrial protein-protein interaction with UCP3 and the first demonstration that UCP3 binds directly, and in cells and tissues with mitochondrial thioredoxin 2. CONCLUSION These studies identify a novel UCP3-Trx2 complex, a novel submitochondrial localization of Trx2, and a mechanism underlying UCP3-regulated mitochondrial ROS production.
Collapse
Affiliation(s)
- Katsuya Hirasaka
- Division of Pharmacology/Toxicology, University of Texas at Austin, Austin, Texas 78714, USA
| | | | | | | | | | | | | |
Collapse
|
135
|
Haschemi A, Chin BY, Jeitler M, Esterbauer H, Wagner O, Bilban M, Otterbein LE. Carbon monoxide induced PPARγ SUMOylation and UCP2 block inflammatory gene expression in macrophages. PLoS One 2011; 6:e26376. [PMID: 22046279 PMCID: PMC3201958 DOI: 10.1371/journal.pone.0026376] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Accepted: 09/26/2011] [Indexed: 01/19/2023] Open
Abstract
Carbon monoxide (CO) dampens pro-inflammatory responses in a peroxisome proliferator-activated receptor-γ (PPARγ) and p38 mitogen-activated protein kinase (MAPK) dependent manner. Previously, we demonstrated that CO inhibits lipopolysaccharide (LPS)-induced expression of the proinflammatory early growth response-1 (Egr-1) transcription factor in macrophages via activation of PPARγ. Here, we further characterize the molecular mechanisms by which CO modulates the activity of PPARγ and Egr-1 repression. We demonstrate that CO enhances SUMOylation of PPARγ which we find was attributed to mitochondrial ROS generation. Ectopic expression of a SUMOylation-defective PPARγ-K365R mutant partially abolished CO-mediated suppression of LPS-induced Egr-1 promoter activity. Expression of a PPARγ-K77R mutant did not impair the effect of CO. In addition to PPARγ SUMOylation, CO-activated p38 MAPK was responsible for Egr-1 repression. Blocking both CO-induced PPARγ SUMOylation and p38 activation, completely reversed the effects of CO on inflammatory gene expression. In primary macrophages isolated form C57/BL6 male mice, we identify mitochondrial ROS formation by CO as the upstream trigger for the observed effects on Egr-1 in part through uncoupling protein 2 (UCP2). Macrophages derived from bone marrow isolated from Ucp2 gene Knock-Out C57/BL6 mice (Ucp2(-/-)), produced significantly less ROS with CO exposure versus wild-type macrophages. Moreover, absence of UCP2 resulted in a complete loss of CO mediated Egr-1 repression. Collectively, these results indentify p38 activation, PPARγ-SUMOylation and ROS formation via UCP2 as a cooperative system by which CO impacts the inflammatory response.
Collapse
Affiliation(s)
- Arvand Haschemi
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Beek Yoke Chin
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Markus Jeitler
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Harald Esterbauer
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Oswald Wagner
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Martin Bilban
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Leo E. Otterbein
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
136
|
Divakaruni AS, Brand MD. The regulation and physiology of mitochondrial proton leak. Physiology (Bethesda) 2011; 26:192-205. [PMID: 21670165 DOI: 10.1152/physiol.00046.2010] [Citation(s) in RCA: 289] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Mitochondria couple respiration to ATP synthesis through an electrochemical proton gradient. Proton leak across the inner membrane allows adjustment of the coupling efficiency. The aim of this review is threefold: 1) introduce the unfamiliar reader to proton leak and its physiological significance, 2) review the role and regulation of uncoupling proteins, and 3) outline the prospects of proton leak as an avenue to treat obesity, diabetes, and age-related disease.
Collapse
Affiliation(s)
- Ajit S Divakaruni
- Medical Research Council Mitochondrial Biology Unit, Wellcome Trust/MRC Building, Cambridge, United Kingdom
| | | |
Collapse
|
137
|
Wang Q, Zhang M, Liang B, Shirwany N, Zhu Y, Zou MH. Activation of AMP-activated protein kinase is required for berberine-induced reduction of atherosclerosis in mice: the role of uncoupling protein 2. PLoS One 2011; 6:e25436. [PMID: 21980456 PMCID: PMC3181327 DOI: 10.1371/journal.pone.0025436] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Accepted: 09/05/2011] [Indexed: 12/15/2022] Open
Abstract
AIMS Berberine, a botanical alkaloid purified from Coptidis rhizoma, is reported to activate the AMP-activated protein kinase (AMPK). Whether AMPK is required for the protective effects of berberine in cardiovascular diseases remains unknown. This study was designed to determine whether AMPK is required for berberine-induced reduction of oxidative stress and atherosclerosis in vivo. METHODS ApoE (ApoE⁻/⁻) mice and ApoE⁻/⁻/AMPK alpha 2⁻/⁻ mice that were fed Western diets were treated with berberine for 8 weeks. Atherosclerotic aortic lesions, expression of uncoupling protein 2 (UCP2), and markers of oxidative stress were evaluated in isolated aortas. RESULTS In ApoE⁻/⁻ mice, chronic administration of berberine significantly reduced aortic lesions, markedly reduced oxidative stress and expression of adhesion molecules in aorta, and significantly increased UCP2 levels. In contrast, in ApoE⁻/⁻/AMPK alpha 2⁻/⁻ mice, berberine had little effect on those endpoints. In cultured human umbilical vein endothelial cells (HUVECs), berberine significantly increased UCP2 mRNA and protein expression in an AMPK-dependent manner. Transfection of HUVECs with nuclear respiratory factor 1 (NRF1)-specific siRNA attenuated berberine-induced expression of UCP2, whereas transfection with control siRNA did not. Finally, berberine promoted mitochondrial biogenesis that contributed to up-regulation of UCP2 expression. CONCLUSION We conclude that berberine reduces oxidative stress and vascular inflammation, and suppresses atherogenesis via a mechanism that includes stimulation of AMPK-dependent UCP2 expression.
Collapse
Affiliation(s)
- Qilong Wang
- Section of Molecular Medicine, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Miao Zhang
- Section of Molecular Medicine, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Bin Liang
- Section of Molecular Medicine, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Najeeb Shirwany
- Section of Molecular Medicine, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Yi Zhu
- Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China
| | - Ming-Hui Zou
- Section of Molecular Medicine, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
- * E-mail:
| |
Collapse
|
138
|
Mailloux RJ, Harper ME. Uncoupling proteins and the control of mitochondrial reactive oxygen species production. Free Radic Biol Med 2011; 51:1106-15. [PMID: 21762777 DOI: 10.1016/j.freeradbiomed.2011.06.022] [Citation(s) in RCA: 406] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Revised: 06/14/2011] [Accepted: 06/16/2011] [Indexed: 12/16/2022]
Abstract
Reactive oxygen species (ROS), natural by-products of aerobic respiration, are important cell signaling molecules, which left unchecked can severely impair cellular functions and induce cell death. Hence, cells have developed a series of systems to keep ROS in the nontoxic range. Uncoupling proteins (UCPs) 1-3 are mitochondrial anion carrier proteins that are purported to play important roles in minimizing ROS emission from the electron transport chain. The function of UCP1 in this regard is highly contentious. However, UCPs 2 and 3 are generally thought to be activated by ROS or ROS by-products to induce proton leak, thus providing a negative feedback loop for mitochondrial ROS production. In our laboratory, we have not only confirmed that ROS activate UCP2 and UCP3, but also demonstrated that UCP2 and UCP3 are controlled by covalent modification by glutathione. Furthermore, the reversible glutathionylation is required to activate/inhibit UCP2 and UCP3, but not UCP1. Hence, our findings are consistent with the notion that UCPs 2 and 3 are acutely activated by ROS, which then directly modulate the glutathionylation status of the UCP to decrease ROS emission and participate in cell signaling mechanisms.
Collapse
Affiliation(s)
- Ryan J Mailloux
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa Faculty of Medicine, Ottawa, ON, Canada K1G8M5
| | | |
Collapse
|
139
|
Silveira LR, Pinheiro CHDJ, Zoppi CC, Hirabara SM, Vitzel KF, Bassit RA, Barbosa MR, Sampaio IH, Melo IHP, Fiamoncini J, Carneiro EM, Curi R. [Regulation of glucose and fatty acid metabolism in skeletal muscle during contraction]. ACTA ACUST UNITED AC 2011; 55:303-13. [PMID: 21881812 DOI: 10.1590/s0004-27302011000500002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2011] [Accepted: 05/12/2011] [Indexed: 03/10/2023]
Abstract
The glucose-fatty acid cycle explains the preference for fatty acid during moderate and long duration physical exercise. In contrast, there is a high glucose availability and oxidation rate in response to intense physical exercise. The reactive oxygen species (ROS) production during physical exercise suggests that the redox balance is important to regulate of lipids/carbohydrate metabolism. ROS reduces the activity of the Krebs cycle, and increases the activity of mitochondrial uncoupling proteins. The opposite effects happen during moderate physical activity. Thus, some issues is highlighted in the present review: Why does skeletal muscle prefer lipids in the basal and during moderate physical activity? Why does glucose-fatty acid fail to carry out their effects during intense physical exercise? How skeletal muscles regulate the lipids and carbohydrate metabolism during the contraction-relaxation cycle?
Collapse
Affiliation(s)
- Leonardo R Silveira
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, SP, Brasil.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
140
|
Hexokinase II acts through UCP3 to suppress mitochondrial reactive oxygen species production and maintain aerobic respiration. Biochem J 2011; 437:301-11. [PMID: 21554247 DOI: 10.1042/bj20110571] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
UCP3 (uncoupling protein-3) mitigates mitochondrial ROS (reactive oxygen species) production, but the mechanisms are poorly understood. Previous studies have also examined UCP3 effects, including decreased ROS production, during metabolic states when fatty acid oxidation is high (e.g. a fasting state). However, the role of UCP3 when carbohydrate oxidation is high (e.g. fed state) has remained largely unexplored. In the present study, we show that mitochondrial-bound HK (hexokinase) II curtails oxidative stress and enhances aerobic metabolism of glucose in the fed state in a UCP3-dependent manner. Genetic knockout or inhibition of UCP3 significantly decreased mitochondrial-bound HKII. Furthermore, UCP3 was required for the HKII-mediated decrease in mitochondrial ROS emission. Intriguingly, the UCP3-mediated modulation of mitochondria-associated HKII was only observed in cells cultured under high-glucose conditions. UCP3 was required to maintain high rates of aerobic metabolism in high-glucose-treated cells and in muscle of fed mice. Deficiency in UCP3 resulted in a metabolic shift that favoured anaerobic glycolytic metabolism, increased glucose uptake and increased sensitivity to oxidative challenge. PET (positron emission tomography) of [18F]fluoro-deoxyglucose uptake confirmed these findings in UCP3-knockout and wild-type mice. Collectively, our findings link the anti-oxidative and metabolic functions of UCP3 through a surprising molecular connection with mitochondrial-bound HKII.
Collapse
|
141
|
Chung ER, Shin SC, Heo JP. Association between SNP Marker of Uncoupling Protein 3 Gene and Meat Yield and Marbling Score Traits in Korean Cattle. Korean J Food Sci Anim Resour 2011. [DOI: 10.5851/kosfa.2011.31.4.530] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
142
|
Santandreu FM, Valle A, Oliver J, Roca P. Resveratrol potentiates the cytotoxic oxidative stress induced by chemotherapy in human colon cancer cells. Cell Physiol Biochem 2011; 28:219-28. [PMID: 21865729 DOI: 10.1159/000331733] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/08/2011] [Indexed: 12/20/2022] Open
Abstract
The treatment of advanced colorectal cancer with 5-fluorouracil has two major problems: development of tumor resistance and toxicity toward normal tissues. The aim of this study was to investigate the possible advantages of combining 5-fluorouracil (5-FU) with resveratrol (trans-3, 4', 5-trihydroxystilbene) for treating HT-29 and SW-620 colorectal carcinoma cell lines. Since combined treatment using 5-FU with resveratrol resulted in a significant decrease in long-term cell survival, we investigated the possible basis of this synergistic interaction at a molecular level, focusing on oxidative stress as a possible mediator of cell death. Resveratrol established interactions with the mitochondria of cancer cells and induced an imbalance in cellular antioxidant activities, leading to a significant increase in the levels of both intracellular reactive oxygen species (ROS) and lipid peroxides. Combined treatment with resveratrol sensitized colon cancer cells to 5-fluorouracil, inducing a further increase in oxidative stress, which was linked to the inhibition of AKT and STAT3 proteins, which are known to have oncogenic potential in colorectal carcinomas.
Collapse
Affiliation(s)
- Francisca M Santandreu
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d'Investigació en Ciències de la Salut (IUNICS). Universitat de les Illes Balears, Palma de Mallorca, Illes Balears, Spain
| | | | | | | |
Collapse
|
143
|
Wong KE, Kong J, Zhang W, Szeto FL, Ye H, Deb DK, Brady MJ, Li YC. Targeted expression of human vitamin D receptor in adipocytes decreases energy expenditure and induces obesity in mice. J Biol Chem 2011; 286:33804-10. [PMID: 21840998 DOI: 10.1074/jbc.m111.257568] [Citation(s) in RCA: 134] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Our previous studies demonstrated a high fat diet-resistant lean phenotype of vitamin D receptor (VDR)-null mutant mice mainly due to increased energy expenditure, suggesting an involvement of the VDR in energy metabolism. Here, we took a transgenic approach to further define the role of VDR in adipocyte biology. We used the aP2 gene promoter to target the expression of the human (h) VDR in adipocytes in mice. In contrast to the VDR-null mice, the aP2-hVDR Tg mice developed obesity compared with the wild-type counterparts without changes in food intake. The increase in fat mass was mainly due to markedly reduced energy expenditure, which was correlated with decreased locomotive activity and reduced fatty acid β-oxidation and lipolysis in the adipose tissue in the transgenic mice. Consistently, the expression of genes involved in the regulation of fatty acid transport, thermogenesis, and lipolysis were suppressed in the transgenic mice. Taken together, these data confirm an important role of the VDR in the regulation of energy metabolism.
Collapse
Affiliation(s)
- Kari E Wong
- Committee on Molecular Metabolism and Nutrition, Division of Biological Sciences,The University of Chicago, Chicago, Illinois 60637, USA
| | | | | | | | | | | | | | | |
Collapse
|
144
|
Absence of mitochondrial uncoupling protein 3: effect on thymus and spleen in the fed and fasted mice. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2011; 1807:1064-74. [PMID: 21689632 DOI: 10.1016/j.bbabio.2011.06.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2011] [Revised: 06/04/2011] [Accepted: 06/06/2011] [Indexed: 11/21/2022]
Abstract
Mitochondrial uncoupling protein 3 (UCP3) is constitutively expressed in mitochondria from thymus and spleen of mice, and confocal microscopy has been used to visualize UCP3 in situ in mouse thymocytes. UCP3 is present in mitochondria of thymus and spleen up to at least 16 weeks after birth, but levels decrease by a half in thymus and a fifth in spleen after three weeks, probably reflecting the suckling to weaning transition. UCP3 protein levels increase approximately 3-fold in thymus on starvation, but expression levels in spleen were unaffected by starvation. Lack of UCP3 had little effect on thymus mass or thymocyte number. However, lack of UCP3 affected spleen mass and splenocyte number (in the fasted state) and results in reduced CD4+ single positive cell numbers and reduced double negative cells in the thymus, but as a 2-fold increase in the proportion of CD4(+), CD8(+) and DP cells in spleen. Starvation attenuates these proportionate differences in the spleen. A lack of UCP3 had no apparent effect on basal oxygen consumption of thymocytes or splenocytes or on oxygen consumption due to mitochondrial proton leak. Splenocytes from UCP3 knock-out mice are also more resistant to apoptosis than those from wild-type mice. Overall we can conclude that UCP3 affects thymocyte and spleen cell profiles in the fed and fasted states.
Collapse
|
145
|
|
146
|
Miyashita K, Nishikawa S, Beppu F, Tsukui T, Abe M, Hosokawa M. The allenic carotenoid fucoxanthin, a novel marine nutraceutical from brown seaweeds. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2011; 91:1166-74. [PMID: 21433011 DOI: 10.1002/jsfa.4353] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2010] [Revised: 01/17/2011] [Accepted: 01/18/2011] [Indexed: 05/24/2023]
Abstract
Obesity and type 2 diabetes are pathologies with rapidly growing prevalence throughout the world. A few molecular targets offer the most hope for anti-obesity and anti-diabetic therapeutics. One of the keys to success will be the induction of uncoupling protein 1 (UCP1) in abdominal white adipose tissue (WAT) and the regulation of cytokine secretions from both abdominal adipose cells and macrophage cells infiltrated into adipose tissue. Anti-obesity and anti-diabetic effects of fucoxanthin, a characteristic carotenoid found in brown seaweeds, have been reported. Nutrigenomic studies reveal that fucoxanthin induces UCP1 in abdominal WAT mitochondria, leading to the oxidation of fatty acids and heat production in WAT. Fucoxanthin improves insulin resistance and decreases blood glucose levels through the regulation of cytokine secretions from WAT. The key structure of carotenoids for the expression of anti-obesity effect is suggested to be the carotenoid end of the polyene chromophore, which contains an allenic bond and two hydroxyl groups.
Collapse
Affiliation(s)
- Kazuo Miyashita
- Faculty of Fisheries Sciences, Hokkaido University, 3-1-1 Minato, Hakodate, Hokkaido 041-8611, Japan.
| | | | | | | | | | | |
Collapse
|
147
|
Uncoupled respiration, ROS production, acute lipotoxicity and oxidative damage in isolated skeletal muscle mitochondria from UCP3-ablated mice. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2011; 1807:1095-105. [PMID: 21565164 DOI: 10.1016/j.bbabio.2011.04.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Revised: 04/06/2011] [Accepted: 04/07/2011] [Indexed: 12/22/2022]
Abstract
The function of uncoupling protein 3 (UCP3) is still not established. Mitochondrial uncoupling, control of ROS production, protection against lipotoxicity and protection against oxidative stress are functions classically discussed. To establish a role for UCP3 in these functions, we have here used UCP3 (-/-) mice, backcrossed for 10 generations on a C57Bl/6 background. In isolated skeletal muscle mitochondria, we examined uncoupled respiration, both unstimulated and in the presence of fatty acids. We did not observe any difference between mitochondria from wildtype and UCP3 (-/-) mice. We measured H(2)O(2) production rate and respiration rate under reactive oxygen species-generating conditions (succinate without rotenone) but found no effect of UCP3. We tested two models of acute lipotoxicity-fatty acid-induced oxidative inhibition and fatty acid-induced swelling-but did not observe any protective effect of UCP3. We examined oxidative stress by quantifying 4-hydroxynonenal protein adducts and protein carbonyls in the mitochondria-but did not observe any protective effect of UCP3. We conclude that under the experimental conditions tested here, we find no evidence for the function of UCP3 being basal or induced uncoupling, regulation of ROS production, protection against acute lipotoxicity or protection against oxidative damage.
Collapse
|
148
|
Mailloux RJ, Seifert EL, Bouillaud F, Aguer C, Collins S, Harper ME. Glutathionylation acts as a control switch for uncoupling proteins UCP2 and UCP3. J Biol Chem 2011; 286:21865-75. [PMID: 21515686 DOI: 10.1074/jbc.m111.240242] [Citation(s) in RCA: 146] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The mitochondrial uncoupling proteins 2 and 3 (UCP2 and -3) are known to curtail oxidative stress and participate in a wide array of cellular functions, including insulin secretion and the regulation of satiety. However, the molecular control mechanism(s) governing these proteins remains elusive. Here we reveal that UCP2 and UCP3 contain reactive cysteine residues that can be conjugated to glutathione. We further demonstrate that this modification controls UCP2 and UCP3 function. Both reactive oxygen species and glutathionylation were found to activate and deactivate UCP3-dependent increases in non-phosphorylating respiration. We identified both Cys(25) and Cys(259) as the major glutathionylation sites on UCP3. Additional experiments in thymocytes from wild-type and UCP2 null mice demonstrated that glutathionylation similarly diminishes non-phosphorylating respiration. Our results illustrate that UCP2- and UCP3-mediated state 4 respiration is controlled by reversible glutathionylation. Altogether, these findings advance our understanding of the roles UCP2 and UCP3 play in modulating metabolic efficiency, cell signaling, and oxidative stress processes.
Collapse
Affiliation(s)
- Ryan J Mailloux
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada, K1H 8M5
| | | | | | | | | | | |
Collapse
|
149
|
Joubert R, Métayer-Coustard S, Crochet S, Cailleau-Audouin E, Dupont J, Duclos MJ, Tesseraud S, Collin A. Regulation of the expression of the avian uncoupling protein 3 by isoproterenol and fatty acids in chick myoblasts: possible involvement of AMPK and PPARalpha? Am J Physiol Regul Integr Comp Physiol 2011; 301:R201-8. [PMID: 21508290 DOI: 10.1152/ajpregu.00087.2010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The avian uncoupling protein 3 (UCP3), mainly expressed in muscle tissue, could be involved in fatty acid (FA) metabolism, limitation of reactive oxygen species production, and/or nonshivering thermogenesis. We recently demonstrated that UCP3 mRNA expression was increased by isoproterenol (Iso), a β-agonist, in chicken Pectoralis major. This upregulation was associated with changes in FA metabolism and variations in the activation of AMP-activated protein kinase (AMPK) and in the expression of the transcription factors peroxisome proliferator-activated receptor (PPAR)α, PPARβ/δ, and PPARγ coactivator-1α (PGC-1α). The aim of the present study was to elucidate the mechanisms involving AMPK and PPARα in UCP3 regulation in primary cultures of chick myoblasts. Avian UCP3 mRNA expression, associated with p38 mitogen-activated protein kinase (p38 MAPK) activation, was increased by Iso and/or FAs. The PKA pathway mediated the effects of Iso on UCP3 expression. FA stimulation also led to AMPK activation. Furthermore, the direct involvement of AMPK on UCP3 regulation was shown by using 5-aminoimidazole-4-carboxyamide ribonucleoside and Compound C. The use of the p38 MAPK inhibitor SB202190, which was associated with AMPK activation, also dramatically enhanced UCP3 mRNA expression. Finally the PPARα agonist WY-14643 strongly increased UCP3 mRNA expression. This study highlights the control of UCP3 expression by the β-adrenergic system and FA in chick myoblasts and demonstrates that its expression is directly regulated by AMPK and by PPARα. Overexpression of avian UCP3 might modulate energy utilization or limit oxidative stress when mitochondrial metabolism of FA is triggered by catecholamines.
Collapse
Affiliation(s)
- Romain Joubert
- INRA, UR83 Recherches Avicoles, F-37380 Nouzilly, France
| | | | | | | | | | | | | | | |
Collapse
|
150
|
Feed restriction up-regulates uncoupling protein 3 (UCP3) gene expression in heart and red muscle tissues of gilthead sea bream (Sparus aurata L.) New insights in substrate oxidation and energy expenditure. Comp Biochem Physiol A Mol Integr Physiol 2011; 159:296-302. [PMID: 21463702 DOI: 10.1016/j.cbpa.2011.03.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Revised: 03/24/2011] [Accepted: 03/25/2011] [Indexed: 01/26/2023]
Abstract
The physiological regulation of the mitochondrial uncoupling protein 3 (UCP3) remains practically unexplored in fish and the aim of this study was to examine the effects of ration size on the regulation of UCP3 in heart, red skeletal muscle and white skeletal muscle of gilthead sea bream (Sparus aurata L.). Juvenile fish were fed at three different levels for 11 weeks: i) full ration until visual satiety (R(100) group), ii) 70% of satiation (R(70) group) and iii) 70% of satiation with two finishing weeks at the maintenance ration (20% of the satiation level) (R(70-20) group). The thirty percent feed restriction improved fish performance, increasing feed conversion efficiency and circulating levels of insulin-like growth factor-I (IGF-I). Fish of the R(70-20) group showed reduced growth and low circulating levels of IGF-I in combination with increased circulating concentrations of growth hormone and free fatty acids. Feed restriction did not alter UCP3 transcript levels in white skeletal muscle, but improved this tissue's oxidative capacity as assessed by changes in glycolytic and oxidative mitochondrial enzyme activities. In contrast, in cardiac and red skeletal muscle tissues, this dietary treatment primarily increased UCP3 mRNA expression. The respiratory control ratio of freshly isolated heart mitochondria was slightly lower in R(70-20) fish than in R(100) fish, which suggests that there was an increase in mitochondrial uncoupling concomitant with the enhanced UCP3 mRNA expression. Altogether, these findings highlight the different adaptive mechanism of glycolytic and highly oxidative muscle tissues for their rapid adjustment to varying feed intake.
Collapse
|