51
|
A novel mechanism of pre-transplant insulin resistance contributing to post-transplant complications: Cyclosporin A-induced O-GlcNAcylation. Biochem Biophys Res Commun 2017; 492:172-177. [DOI: 10.1016/j.bbrc.2017.08.033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 08/09/2017] [Indexed: 01/30/2023]
|
52
|
Kim G, Cao L, Reece EA, Zhao Z. Impact of protein O-GlcNAcylation on neural tube malformation in diabetic embryopathy. Sci Rep 2017; 7:11107. [PMID: 28894244 PMCID: PMC5593976 DOI: 10.1038/s41598-017-11655-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 08/25/2017] [Indexed: 01/15/2023] Open
Abstract
Diabetes mellitus in early pregnancy can cause neural tube defects (NTDs) in embryos by perturbing protein activity, causing cellular stress, and increasing programmed cell death (apoptosis) in the tissues required for neurulation. Hyperglycemia augments a branch pathway in glycolysis, the hexosamine biosynthetic pathway (HBP), to increase uridine diphosphate-N-acetylglucosamine (UDP-GlcNAc). GlcNAc can be added to proteins by O-GlcNAc transferase (OGT) to regulate protein activity. In the embryos of diabetic mice, OGT is highly activated in association with increases in global protein O-GlcNAcylation. In neural stem cells in vitro, high glucose elevates O-GlcNAcylation and reactive oxygen species, but the elevations can be suppressed by an OGT inhibitor. Inhibition of OGT in diabetic pregnant mice in vivo decreases NTD rate in the embryos. This effect is associated with reduction in global O-GlcNAcylation, alleviation of intracellular stress, and decreases in apoptosis in the embryos. These suggest that OGT plays an important role in diabetic embryopathy via increasing protein O-GlcNAcylation, and that inhibiting OGT could be a candidate approach to prevent birth defects in diabetic pregnancies.
Collapse
Affiliation(s)
- Gyuyoup Kim
- Department of Obstetrics, Gynecology and Reproductive Sciences,University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Lixue Cao
- Department of Obstetrics, Gynecology and Reproductive Sciences,University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - E Albert Reece
- Department of Obstetrics, Gynecology and Reproductive Sciences,University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Zhiyong Zhao
- Department of Obstetrics, Gynecology and Reproductive Sciences,University of Maryland School of Medicine, Baltimore, Maryland, USA.
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
53
|
Badgett MJ, Boyes B, Orlando R. Predicting the Retention Behavior of Specific O-Linked Glycopeptides. J Biomol Tech 2017; 28:122-126. [PMID: 28785176 DOI: 10.7171/jbt.17-2803-003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
O-Linked glycosylation is a common post-translational modification that can alter the overall structure, polarity, and function of proteins. Reverse-phase (RP) chromatography is the most common chromatographic approach to analyze O-glycosylated peptides and their unmodified counterparts, even though this approach often does not provide adequate separation of these two species. Hydrophilic interaction liquid chromatography (HILIC) can be a solution to this problem, as the polar glycan interacts with the polar stationary phase and potentially offers the ability to resolve the peptide from its modified form(s). In this paper, HILIC is used to separate peptides with O-N-acetylgalactosamine (O-GalNAc), O-N-acetylglucosamine (O-GlcNAc), and O-fucose additions from their native forms, and coefficients representing the extent of hydrophilicity were derived using linear regression analysis as a means to predict the retention times of peptides with these modifications.
Collapse
Affiliation(s)
- Majors J Badgett
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia 30602, USA; and
| | - Barry Boyes
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia 30602, USA; and.,Advanced Materials Technology, Wilmington, Delaware 19810, USA
| | - Ron Orlando
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia 30602, USA; and
| |
Collapse
|
54
|
He Y, Ma X, Li D, Hao J. Thiamet G mediates neuroprotection in experimental stroke by modulating microglia/macrophage polarization and inhibiting NF-κB p65 signaling. J Cereb Blood Flow Metab 2017; 37:2938-2951. [PMID: 27864466 PMCID: PMC5536801 DOI: 10.1177/0271678x16679671] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 10/24/2016] [Accepted: 10/25/2016] [Indexed: 12/29/2022]
Abstract
Inflammatory responses are accountable for secondary injury induced by acute ischemic stroke (AIS). Previous studies indicated that O-GlcNAc modification (O-GlcNAcylation) is involved in the pathology of AIS, and increase of O-GlcNAcylation by glucosamine attenuated the brain damage after ischemia/reperfusion. Inhibition of β-N-acetylglucosaminidase (OGA) with thiamet G (TMG) is an alternative option for accumulating O-GlcNAcylated proteins. In this study, we investigate the neuroprotective effect of TMG in a mouse model of experimental stroke. Our results indicate that TMG administration either before or after middle cerebral artery occlusion (MCAO) surgery dramatically reduced infarct volume compared with that in untreated controls. TMG treatment ameliorated the neurological deficits and improved clinical outcomes in neurobehavioral tests by modulating the expression of pro-inflammatory and anti-inflammatory cytokines. Additionally, TMG administration reduced the number of Iba1+ cells in MCAO mice, decreased expression of the M1 markers, and increased expression of the M2 markers in vivo. In vitro, M1 polarization of BV2 cells was inhibited by TMG treatment. Moreover, TMG decreased the expression of iNOS and COX2 mainly by suppressing NF-κB p65 signaling. These results suggest that TMG exerts a neuroprotective effect and could be useful as an anti-inflammatory agent for ischemic stroke therapy.
Collapse
Affiliation(s)
| | | | - Daojing Li
- Department of Neurology and Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Junwei Hao
- Department of Neurology and Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
55
|
Bierhansl L, Conradi LC, Treps L, Dewerchin M, Carmeliet P. Central Role of Metabolism in Endothelial Cell Function and Vascular Disease. Physiology (Bethesda) 2017; 32:126-140. [PMID: 28202623 PMCID: PMC5337830 DOI: 10.1152/physiol.00031.2016] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The importance of endothelial cell (EC) metabolism and its regulatory role in the angiogenic behavior of ECs during vessel formation and in the function of different EC subtypes determined by different vascular beds has been recognized only in the last few years. Even more importantly, apart from a role of nitric oxide and reactive oxygen species in EC dysfunction, deregulations of EC metabolism in disease only recently received increasing attention. Although comprehensive metabolic characterization of ECs still needs further investigation, the concept of targeting EC metabolism to treat vascular disease is emerging. In this overview, we summarize EC-specific metabolic pathways, describe the current knowledge on their deregulation in vascular diseases, and give an outlook on how vascular endothelial metabolism can serve as a target to normalize deregulated endothelium.
Collapse
Affiliation(s)
- Laura Bierhansl
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, University of Leuven, Leuven, Belgium; and
- Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, University of Leuven, Leuven, Belgium
| | - Lena-Christin Conradi
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, University of Leuven, Leuven, Belgium; and
- Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, University of Leuven, Leuven, Belgium
| | - Lucas Treps
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, University of Leuven, Leuven, Belgium; and
- Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, University of Leuven, Leuven, Belgium
| | - Mieke Dewerchin
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, University of Leuven, Leuven, Belgium; and
- Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, University of Leuven, Leuven, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, University of Leuven, Leuven, Belgium; and
- Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, University of Leuven, Leuven, Belgium
| |
Collapse
|
56
|
Qin CX, Sleaby R, Davidoff AJ, Bell JR, De Blasio MJ, Delbridge LM, Chatham JC, Ritchie RH. Insights into the role of maladaptive hexosamine biosynthesis and O-GlcNAcylation in development of diabetic cardiac complications. Pharmacol Res 2016; 116:45-56. [PMID: 27988387 DOI: 10.1016/j.phrs.2016.12.016] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 10/28/2016] [Accepted: 12/13/2016] [Indexed: 12/21/2022]
Abstract
Diabetes mellitus significantly increases the risk of heart failure, independent of coronary artery disease. The mechanisms implicated in the development of diabetic heart disease, commonly termed diabetic cardiomyopathy, are complex, but much of the impact of diabetes on the heart can be attributed to impaired glucose handling. It has been shown that the maladaptive nutrient-sensing hexosamine biosynthesis pathway (HBP) contributes to diabetic complications in many non-cardiac tissues. Glucose metabolism by the HBP leads to enzymatically-regulated, O-linked attachment of a sugar moiety molecule, β-N-acetylglucosamine (O-GlcNAc), to proteins, affecting their biological activity (similar to phosphorylation). In normal physiology, transient activation of HBP/O-GlcNAc mechanisms is an adaptive, protective means to enhance cell survival; interventions that acutely suppress this pathway decrease tolerance to stress. Conversely, chronic dysregulation of HBP/O-GlcNAc mechanisms has been shown to be detrimental in certain pathological settings, including diabetes and cancer. Most of our understanding of the impact of sustained maladaptive HBP and O-GlcNAc protein modifications has been derived from adipose tissue, skeletal muscle and other non-cardiac tissues, as a contributing mechanism to insulin resistance and progression of diabetic complications. However, the long-term consequences of persistent activation of cardiac HBP and O-GlcNAc are not well-understood; therefore, the goal of this timely review is to highlight current understanding of the role of the HBP pathway in development of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Cheng Xue Qin
- Heart Failure Pharmacology, Baker IDI Heart & Diabetes Institute, Melbourne VIC 3004, Australia; Department of Pharmacology, University of Melbourne, VIC 3010, Australia
| | - Rochelle Sleaby
- Heart Failure Pharmacology, Baker IDI Heart & Diabetes Institute, Melbourne VIC 3004, Australia; Department of Physiology, University of Melbourne, VIC 3010, Australia
| | - Amy J Davidoff
- University of New England, Biddeford, ME, 04072, United States
| | - James R Bell
- Department of Physiology, University of Melbourne, VIC 3010, Australia
| | - Miles J De Blasio
- Heart Failure Pharmacology, Baker IDI Heart & Diabetes Institute, Melbourne VIC 3004, Australia; School of BioSciences, University of Melbourne, VIC 3010, Australia
| | | | - John C Chatham
- University of Alabama at Birmingham, Birmingham, AL, 35233, United States
| | - Rebecca H Ritchie
- Heart Failure Pharmacology, Baker IDI Heart & Diabetes Institute, Melbourne VIC 3004, Australia; Department of Pharmacology, University of Melbourne, VIC 3010, Australia; Department of Medicine, Monash University, Clayton 3800, VIC, Australia.
| |
Collapse
|
57
|
Cividini F, Scott BT, Dai A, Han W, Suarez J, Diaz-Juarez J, Diemer T, Casteel DE, Dillmann WH. O-GlcNAcylation of 8-Oxoguanine DNA Glycosylase (Ogg1) Impairs Oxidative Mitochondrial DNA Lesion Repair in Diabetic Hearts. J Biol Chem 2016; 291:26515-26528. [PMID: 27816939 DOI: 10.1074/jbc.m116.754481] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 11/03/2016] [Indexed: 11/06/2022] Open
Abstract
mtDNA damage in cardiac myocytes resulting from increased oxidative stress is emerging as an important factor in the pathogenesis of diabetic cardiomyopathy. A prevalent lesion that occurs in mtDNA damage is the formation of 8-hydroxy-2'-deoxyguanosine (8-OHdG), which can cause mutations when not repaired properly by 8-oxoguanine DNA glycosylase (Ogg1). Although the mtDNA repair machinery has been described in cardiac myocytes, the regulation of this repair has been incompletely investigated. Here we report that the hearts of type 1 diabetic mice, despite having increased Ogg1 protein levels, had significantly lower Ogg1 activity than the hearts of control, non-type 1 diabetic mice. In diabetic hearts, we further observed increased levels of 8-OHdG and an increased amount of mtDNA damage. Interestingly, Ogg1 was found to be highly O-GlcNAcylated in diabetic mice compared with controls. In vitro experiments demonstrated that O-GlcNAcylation inhibits Ogg1 activity, which could explain the mtDNA lesion accumulation observed in vivo Reducing Ogg1 O-GlcNAcylation in vivo by introducing a dominant negative O-GlcNAc transferase mutant (F460A) restored Ogg1 enzymatic activity and, consequently, reduced 8-OHdG and mtDNA damage despite the adverse hyperglycemic milieu. Taken together, our results implicate hyperglycemia-induced O-GlcNAcylation of Ogg1 in increased mtDNA damage and, therefore, provide a new plausible biochemical mechanism for diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Federico Cividini
- From the Department of Medicine, University of California, San Diego, La Jolla, California 92093-0671 and
| | - Brian T Scott
- From the Department of Medicine, University of California, San Diego, La Jolla, California 92093-0671 and
| | - Anzhi Dai
- From the Department of Medicine, University of California, San Diego, La Jolla, California 92093-0671 and
| | - Wenlong Han
- From the Department of Medicine, University of California, San Diego, La Jolla, California 92093-0671 and
| | - Jorge Suarez
- From the Department of Medicine, University of California, San Diego, La Jolla, California 92093-0671 and
| | - Julieta Diaz-Juarez
- the Department of Pharmacology, Instituto Nacional de Cardiología, Juan Badiano 41, Barrio Belisario Domínguez Secc XVI, 14080 Tlalpan, DF, Mexico
| | - Tanja Diemer
- From the Department of Medicine, University of California, San Diego, La Jolla, California 92093-0671 and
| | - Darren E Casteel
- From the Department of Medicine, University of California, San Diego, La Jolla, California 92093-0671 and
| | - Wolfgang H Dillmann
- From the Department of Medicine, University of California, San Diego, La Jolla, California 92093-0671 and
| |
Collapse
|
58
|
Zhao L, Feng Z, Yang X, Liu J. The regulatory roles of O-GlcNAcylation in mitochondrial homeostasis and metabolic syndrome. Free Radic Res 2016; 50:1080-1088. [PMID: 27646831 DOI: 10.1080/10715762.2016.1239017] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Nutrients excess is one of the leading causes of metabolic syndrome globally. Protein post-translational O-GlcNAc modification has been recognized as an essential nutrient sensor of the cell. Emerging studies suggest that O-GlcNAcylation lies at the core linking nutritional stress to insulin resistance. Mitochondria are the major site for ATP production in most eukaryotes. Mitochondrial dysfunction and oxidative stress have long been considered as an important mechanism underlying insulin resistance. The metabolic process is under the influence of environmental and nutritional factors, thus sensing and transducing nutritional signals sit at the pivot of metabolism control. For a long time little was known about O-GlcNAcylation within mitochondria since mitochondrial O-GlcNAcylation was regarded rare. Recent findings have demonstrated that O-GlcNAcylation is widely spread among mitochondrial proteins, and that mitochondrial function and oxidative stress both can be regulated by O-GlcNAcylation, particularly under diabetic circumstances.
Collapse
Affiliation(s)
- Lin Zhao
- a Center for Mitochondrial Biology and Medicine, the Key Laboratory of Biomedical Information Engineering of Ministry of Education , School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University , Xi'an , China
| | - Zhihui Feng
- a Center for Mitochondrial Biology and Medicine, the Key Laboratory of Biomedical Information Engineering of Ministry of Education , School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University , Xi'an , China
| | - Xiaoyong Yang
- b Section of Comparative Medicine and Department of Cellular and Molecular Physiology , Yale University School of Medicine , New Haven , CT , USA
| | - Jiankang Liu
- a Center for Mitochondrial Biology and Medicine, the Key Laboratory of Biomedical Information Engineering of Ministry of Education , School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University , Xi'an , China
| |
Collapse
|
59
|
Cencioni C, Atlante S, Savoia M, Martelli F, Farsetti A, Capogrossi MC, Zeiher AM, Gaetano C, Spallotta F. The double life of cardiac mesenchymal cells: Epimetabolic sensors and therapeutic assets for heart regeneration. Pharmacol Ther 2016; 171:43-55. [PMID: 27742569 DOI: 10.1016/j.pharmthera.2016.10.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Organ-specific mesenchymal cells naturally reside in the stroma, where they are exposed to some environmental variables affecting their biology and functions. Risk factors such as diabetes or aging influence their adaptive response. In these cases, permanent epigenetic modifications may be introduced in the cells with important consequences on their local homeostatic activity and therapeutic potential. Numerous results suggest that mesenchymal cells, virtually present in every organ, may contribute to tissue regeneration mostly by paracrine mechanisms. Intriguingly, the heart is emerging as a source of different cells, including pericytes, cardiac progenitors, and cardiac fibroblasts. According to phenotypic, functional, and molecular criteria, these should be classified as mesenchymal cells. Not surprisingly, in recent years, the attention on these cells as therapeutic tools has grown exponentially, although only very preliminary data have been obtained in clinical trials to date. In this review, we summarized the state of the art about the phenotypic features, functions, regenerative properties, and clinical applicability of mesenchymal cells, with a particular focus on those of cardiac origin.
Collapse
Affiliation(s)
- Chiara Cencioni
- Division of Cardiovascular Epigenetics, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany; Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| | - Sandra Atlante
- Division of Cardiovascular Epigenetics, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany; Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| | - Matteo Savoia
- Division of Cardiovascular Epigenetics, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany; Universitá Cattolica, Institute of Medical Pathology, 00138 Rome, Italy; Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS-Policlinico San Donato, San Donato Milanese, Milan 20097, Italy.
| | - Antonella Farsetti
- Consiglio Nazionale delle Ricerche, Istituto di Biologia Cellulare e Neurobiologia, Roma, Italy; Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| | - Maurizio C Capogrossi
- Laboratorio di Patologia Vascolare, Istituto Dermopatico dell'Immacolata, Roma, Italy.
| | - Andreas M Zeiher
- Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| | - Carlo Gaetano
- Division of Cardiovascular Epigenetics, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany; Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| | - Francesco Spallotta
- Division of Cardiovascular Epigenetics, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany; Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| |
Collapse
|
60
|
Mailleux F, Gélinas R, Beauloye C, Horman S, Bertrand L. O-GlcNAcylation, enemy or ally during cardiac hypertrophy development? Biochim Biophys Acta Mol Basis Dis 2016; 1862:2232-2243. [PMID: 27544701 DOI: 10.1016/j.bbadis.2016.08.012] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Revised: 08/12/2016] [Accepted: 08/13/2016] [Indexed: 12/11/2022]
Abstract
O-linked attachment of the monosaccharide β-N-acetyl-glucosamine (O-GlcNAcylation) is a post-translational modification occurring on serine and threonine residues, which is evolving as an important mechanism for the regulation of various cellular processes. The present review will, first, provide a general background on the molecular regulation of protein O-GlcNAcylation and will summarize the role of this post-translational modification in various acute cardiac pathologies including ischemia-reperfusion. Then, we will focus on research studies examining protein O-GlcNAcylation in the context of cardiac hypertrophy. A particular emphasis will be laid on the convergent but also divergent actions of O-GlcNAcylation according to the type of hypertrophy investigated, including physiological, pressure overload-induced and diabetes-linked cardiac hypertrophy. In an attempt to distinguish whether O-GlcNAcylation is detrimental or beneficial, this review will present the different O-GlcNAcylated targets involved in hypertrophy development. We will finally argue on potential interest to target O-GlcNAc processes to treat cardiac hypertrophy. This article is part of a Special Issue entitled: The role of post-translational protein modifications on heart and vascular metabolism edited by Jason R.B. Dyck & Jan F.C. Glatz.
Collapse
Affiliation(s)
- Florence Mailleux
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Brussels, Belgium
| | - Roselle Gélinas
- Montreal Heart Institute, Montreal, Canada; Department of Medicine, Université de Montréal, Montreal, Canada
| | - Christophe Beauloye
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Brussels, Belgium; Cliniques Universitaires Saint-Luc, Division of Cardiology, Brussels, Belgium
| | - Sandrine Horman
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Brussels, Belgium
| | - Luc Bertrand
- Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Brussels, Belgium.
| |
Collapse
|
61
|
Taniguchi N, Takahashi M, Kizuka Y, Kitazume S, Shuvaev VV, Ookawara T, Furuta A. Glycation vs. glycosylation: a tale of two different chemistries and biology in Alzheimer's disease. Glycoconj J 2016; 33:487-97. [PMID: 27325408 DOI: 10.1007/s10719-016-9690-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 05/24/2016] [Accepted: 05/25/2016] [Indexed: 01/21/2023]
Abstract
In our previous studies, we reported that the activity of an anti-oxidant enzyme, Cu,Zn-superoxide dismutase (Cu,Zn-SOD) became decreased as the result of glycation in vitro and in vivo. Glycated Cu,Zn-SOD produces hydroxyl radicals in the presence of transition metals due to the formation of a Schiff base adduct and a subsequent Amadori product. This results in the site-specific cleavage of the molecule, followed by random fragmentation. The glycation of other anti-oxidant enzymes such as glutathione peroxidase and thioredoxin reductase results in a loss or decrease in enzyme activity under pathological conditions, resulting in oxidative stress. The inactivation of anti-oxidant enzymes induces oxidative stress in aging, diabetes and neurodegenerative disorders. It is well known that the levels of Amadori products and N(e)-(carboxylmethyl)lysine (CML) and other carbonyl compounds are increased in diabetes, a situation that will be discussed by the other authors in this special issue. We and others, reported that the glycation products accumulate in the brains of patients with Alzheimer's disease (AD) patients as well as in cerebrospinal fluid (CSF), suggesting that glycation plays a pivotal role in the development of AD. We also showed that enzymatic glycosylation is implicated in the pathogenesis of AD and that oxidative stress is also important in this process. Specific types of glycosylation reactions were found to be up- or downregulated in AD patients, and key AD-related molecules including the amyloid-precursor protein (APP), tau, and APP-cleaving enzymes were shown to be functionally modified as the result of glycosylation. These results suggest that glycation as well as glycosylation are involved in oxidative stress that is associated with aging, diabetes and neurodegenerative diseases such as AD.
Collapse
Affiliation(s)
- Naoyuki Taniguchi
- Systems Glycobiology Research Group, RIKEN-Max Planck Joint Research Center for Systems Chemical Biology, Global Research Cluster, RIKEN, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan.
| | - Motoko Takahashi
- Department of Biochemistry, Sapporo Medical University School of Medicine, South-1 West-17, Chuo-ku, Sapporo, 060-8556, Japan
| | - Yasuhiko Kizuka
- Systems Glycobiology Research Group, RIKEN-Max Planck Joint Research Center for Systems Chemical Biology, Global Research Cluster, RIKEN, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| | - Shinobu Kitazume
- Systems Glycobiology Research Group, RIKEN-Max Planck Joint Research Center for Systems Chemical Biology, Global Research Cluster, RIKEN, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| | - Vladimir V Shuvaev
- Department of Systems Pharmacology and Translational Therapeutics, Center for Translational Targeted Therapeutics and Nanomedicine of the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Tomomi Ookawara
- Laboratory of Biochemistry, School of Pharmacy, Hyogo University of Health Sciences, 1-3-6 Minatojima, Chuo-ku, Kobe, Hyogo, 650-8530, Japan
| | - Akiko Furuta
- Department of Cellular and Molecular Neuropathology, Juntendo University School of Medicine, Hongo 2-1-1, Bunkyou-ku, Tokyo, 113-8421, Japan
| |
Collapse
|
62
|
Lambert M, Richard E, Duban-Deweer S, Krzewinski F, Deracinois B, Dupont E, Bastide B, Cieniewski-Bernard C. O-GlcNAcylation is a key modulator of skeletal muscle sarcomeric morphometry associated to modulation of protein-protein interactions. Biochim Biophys Acta Gen Subj 2016; 1860:2017-30. [PMID: 27301331 DOI: 10.1016/j.bbagen.2016.06.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 05/18/2016] [Accepted: 06/06/2016] [Indexed: 12/27/2022]
Abstract
BACKGROUND The sarcomere structure of skeletal muscle is determined through multiple protein-protein interactions within an intricate sarcomeric cytoskeleton network. The molecular mechanisms involved in the regulation of this sarcomeric organization, essential to muscle function, remain unclear. O-GlcNAcylation, a post-translational modification modifying several key structural proteins and previously described as a modulator of the contractile activity, was never considered to date in the sarcomeric organization. METHODS C2C12 skeletal myotubes were treated with Thiamet-G (OGA inhibitor) in order to increase the global O-GlcNAcylation level. RESULTS Our data clearly showed a modulation of the O-GlcNAc level more sensitive and dynamic in the myofilament-enriched fraction than total proteome. This fine O-GlcNAc level modulation was closely related to changes of the sarcomeric morphometry. Indeed, the dark-band and M-line widths increased, while the I-band width and the sarcomere length decreased according to the myofilament O-GlcNAc level. Some structural proteins of the sarcomere such as desmin, αB-crystallin, α-actinin, moesin and filamin-C have been identified within modulated protein complexes through O-GlcNAc level variations. Their interactions seemed to be changed, especially for desmin and αB-crystallin. CONCLUSIONS For the first time, our findings clearly demonstrate that O-GlcNAcylation, through dynamic regulations of the structural interactome, could be an important modulator of the sarcomeric structure and may provide new insights in the understanding of molecular mechanisms of neuromuscular diseases characterized by a disorganization of the sarcomeric structure. GENERAL SIGNIFICANCE In the present study, we demonstrated a role of O-GlcNAcylation in the sarcomeric structure modulation.
Collapse
Affiliation(s)
- Matthias Lambert
- Univ.Lille, EA7369-URePSSS, Unité de Recherche Pluridisciplinaire Sport, Santé, Société, Equipe « Activité Physique, Muscle, Santé », F-59000 Lille, France
| | - Elodie Richard
- BiCeL (BioImaging Center of Lille - Campus Lille 1), Univ.Lille, FR3688 CNRS FRABio, F-59000 Lille, France
| | - Sophie Duban-Deweer
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), EA2465, Université d'Artois, Faculté Jean Perrin, 62307 Lens, France
| | - Frederic Krzewinski
- PAGés (Plateforme d'Analyses des Glycoconjugués), Univ.Lille, CNRS, UMR 8576, UGSF, Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
| | - Barbara Deracinois
- Univ.Lille, EA7369-URePSSS, Unité de Recherche Pluridisciplinaire Sport, Santé, Société, Equipe « Activité Physique, Muscle, Santé », F-59000 Lille, France
| | - Erwan Dupont
- Univ.Lille, EA7369-URePSSS, Unité de Recherche Pluridisciplinaire Sport, Santé, Société, Equipe « Activité Physique, Muscle, Santé », F-59000 Lille, France
| | - Bruno Bastide
- Univ.Lille, EA7369-URePSSS, Unité de Recherche Pluridisciplinaire Sport, Santé, Société, Equipe « Activité Physique, Muscle, Santé », F-59000 Lille, France
| | - Caroline Cieniewski-Bernard
- Univ.Lille, EA7369-URePSSS, Unité de Recherche Pluridisciplinaire Sport, Santé, Société, Equipe « Activité Physique, Muscle, Santé », F-59000 Lille, France.
| |
Collapse
|
63
|
Cannon MV, Silljé HHW, Sijbesma JWA, Vreeswijk-Baudoin I, Ciapaite J, van der Sluis B, van Deursen J, Silva GJJ, de Windt LJ, Gustafsson JÅ, van der Harst P, van Gilst WH, de Boer RA. Cardiac LXRα protects against pathological cardiac hypertrophy and dysfunction by enhancing glucose uptake and utilization. EMBO Mol Med 2016; 7:1229-43. [PMID: 26160456 PMCID: PMC4568954 DOI: 10.15252/emmm.201404669] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Pathological cardiac hypertrophy is characterized by a shift in metabolic substrate utilization from fatty acids to glucose, but the molecular events underlying the metabolic remodeling remain poorly understood. Here, we investigated the role of liver X receptors (LXRs), which are key regulators of glucose and lipid metabolism, in cardiac hypertrophic pathogenesis. Using a transgenic approach in mice, we show that overexpression of LXRα acts to protect the heart against hypertrophy, fibrosis, and dysfunction. Gene expression profiling studies revealed that genes regulating metabolic pathways were differentially expressed in hearts with elevated LXRα. Functionally, LXRα overexpression in isolated cardiomyocytes and murine hearts markedly enhanced the capacity for myocardial glucose uptake following hypertrophic stress. Conversely, this adaptive response was diminished in LXRα-deficient mice. Transcriptional changes induced by LXRα overexpression promoted energy-independent utilization of glucose via the hexosamine biosynthesis pathway, resulting in O-GlcNAc modification of GATA4 and Mef2c and the induction of cytoprotective natriuretic peptide expression. Our results identify LXRα as a key cardiac transcriptional regulator that helps orchestrate an adaptive metabolic response to chronic cardiac stress, and suggest that modulating LXRα may provide a unique opportunity for intervening in myocyte metabolism.
Collapse
Affiliation(s)
- Megan V Cannon
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Herman H W Silljé
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jürgen W A Sijbesma
- Department of Nuclear Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Inge Vreeswijk-Baudoin
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jolita Ciapaite
- Department Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Bart van der Sluis
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jan van Deursen
- Department of Biochemistry & Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Gustavo J J Silva
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| | - Leon J de Windt
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| | - Jan-Åke Gustafsson
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA Department of Biosciences and Nutrition, Novum, Karolinska Institutet, Huddinge, Sweden
| | - Pim van der Harst
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Wiek H van Gilst
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Rudolf A de Boer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
64
|
Salma W, Franekova V, Lund T, Höper A, Ludvigsen S, Lund J, Aasum E, Ytrehus K, Belke DD, Larsen TS. Dietary Calanus oil antagonizes angiotensin II-induced hypertension and tissue wasting in diet-induced obese mice. Prostaglandins Leukot Essent Fatty Acids 2016; 108:13-21. [PMID: 27154360 DOI: 10.1016/j.plefa.2016.03.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Revised: 03/19/2016] [Accepted: 03/21/2016] [Indexed: 10/22/2022]
Abstract
BACKGROUND We have recently shown that Calanus oil, which is extracted from the marine copepod Calanus finmarchicus, reduces fat deposition, suppresses adipose tissue inflammation and improves insulin sensitivity in high fat-fed rodents. This study expands upon our previous observations by examining whether dietary supplementation with Calanus oil could antagonize angiotensin II (Ang II)-induced hypertension and ventricular remodeling in mice given a high fat diet (HFD). METHODS C57BL/6J mice were initially subjected to 8 weeks of HFD with or without 2% (w/w) Calanus oil. Thereafter, animals within each group were randomized for the administration of either Ang II (1µg/kg/min) or saline for another two weeks, while still on the same dietary regimen. RESULTS Ang II caused a marked decline in body and organ weights in mice receiving non-supplemented HFD, a response which was clearly attenuated in mice receiving Calanus oil supplementation. Furthermore, Ang II-induced elevation in blood pressure was also attenuated in the Calanus oil-supplemented group. As expected, infusion of Ang II produced hypertrophy and up-regulation of marker genes (mRNA level) of both hypertrophy and fibrosis in cardiac muscle, but this response was unaffected by dietary Calanus oil. Fibrosis and inflammation were up-regulated also in the aorta following Ang II infusion. However, the inflammatory response was blocked by Calanus oil supplementation. A final, and unexpected, finding was that dietary intake of Calanus oil caused a robust increase in the level of O-GlcNAcylation in cardiac tissue. CONCLUSION These results suggest that dietary intake of oil from the marine copepod Calanus finmarchicus could be a beneficial addition to conventional hypertension treatment. The compound attenuates inflammation and the severe metabolic stress caused by Ang II infusion. Although the present study suggests that the anti-hypertensive effect of the oil (or its n-3 PUFAs constituents) is related to its anti-inflammatory action in the vessel wall, other mechanisms such as interaction with intracellular calcium mechanisms or a direct antagonistic effect on Ang II receptors should be examined.
Collapse
Affiliation(s)
- Wahida Salma
- Cardiovascular Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, N-9037 Tromsø, Norway
| | - Veronika Franekova
- Cardiovascular Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, N-9037 Tromsø, Norway
| | - Trine Lund
- Cardiovascular Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, N-9037 Tromsø, Norway
| | - Anje Höper
- Cardiovascular Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, N-9037 Tromsø, Norway
| | - Stian Ludvigsen
- Cardiovascular Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, N-9037 Tromsø, Norway
| | - Jim Lund
- Cardiovascular Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, N-9037 Tromsø, Norway
| | - Ellen Aasum
- Cardiovascular Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, N-9037 Tromsø, Norway
| | - Kirsti Ytrehus
- Cardiovascular Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, N-9037 Tromsø, Norway
| | - Darrell D Belke
- Faculty of Kinesiology, University of Calgary, 3300 University Drive NW, Calgary, Alberta T2N 4N1, Canada
| | - Terje S Larsen
- Cardiovascular Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, N-9037 Tromsø, Norway.
| |
Collapse
|
65
|
Gong CX, Liu F, Iqbal K. O-GlcNAcylation: A regulator of tau pathology and neurodegeneration. Alzheimers Dement 2016; 12:1078-1089. [PMID: 27126545 DOI: 10.1016/j.jalz.2016.02.011] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 12/16/2015] [Accepted: 02/03/2016] [Indexed: 01/21/2023]
Abstract
O-GlcNAcylation is the posttranslational modification of intracellular proteins by O-linked β-N-acetylglucosamine (O-GlcNAc). The discovery of O-GlcNAc modification of tau and its impact on tau phosphorylation has attracted recent research interest in O-GlcNAc studies in the Alzheimer's disease (AD) field. Modification of proteins by O-GlcNAc occurs extensively in the brain. The expressions and activities of the enzymes catalyzing O-GlcNAc cycling are several-fold higher in the brain than in the peripheral tissues. The O-GlcNAcylation levels of brain proteins including tau are decreased in AD brain, probably due to decreased brain glucose metabolism. The reduction of brain O-GlcNAcylation appears to mediate the molecular mechanism by which decreased brain glucose metabolism contributes to neurodegeneration. Studies on mouse models of tauopathies suggest a neuroprotective role of pharmacological elevation of brain O-GlcNAc, which could potentially be a promising approach for treating AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Cheng-Xin Gong
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA.
| | - Fei Liu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Khalid Iqbal
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| |
Collapse
|
66
|
Qiao Z, Dang C, Zhou B, Li S, Zhang W, Jiang J, Zhang J, Ma Y, Kong R, Ma Z. Downregulation of O-linked N-acetylglucosamine transferase by RNA interference decreases MMP9 expression in human esophageal cancer cells. Oncol Lett 2016; 11:3317-3323. [PMID: 27123109 PMCID: PMC4840913 DOI: 10.3892/ol.2016.4428] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 01/21/2016] [Indexed: 02/07/2023] Open
Abstract
O-linked N-acetylglucosamine transferase (OGT) catalyzes O-linked glycosylation (O-GlcNAcylation). O-GlcNAcylation is a post-translational carbohydrate modification of diverse nuclear and cytosolic proteins by the addition of O-linked β-N-acetylglucosamine. It was recently demonstrated that OGT and the level of O-GlcNAcylation are upregulated in esophageal cancer; however, the physiological consequences of this upregulation remain unknown. The current study reports that OGT knockdown by short hairpin RNA (shRNA) did not affect cell viability; however, cell migration in esophageal cancer Eca-109 cells was significantly reduced. OGT-specific shRNA vectors efficiently decreased the protein and mRNA levels of OGT and the RL2 level (a marker of O-GlcNAcylation levels) in Eca-109 esophageal cancer cells. In addition, colony formation and cell proliferation assays demonstrated that OGT-specific shRNA decreased the proliferation of Eca-109 cells; however, there was no significant statistical difference between OGT-specific shRNA and control shRNA. Notably, transwell assays demonstrated that the migratory ability of Eca-109 cells was significantly suppressed following knockdown of the OGT gene. Correspondingly, western blot analyses demonstrated that OGT knockdown significantly downregulated the expression of matrix metalloproteinase 9 (MMP9) in Eca-109 cells. These results suggest that OGT may promote the migration, invasion and metastasis of esophageal cancer cells by enhancing the stability or expression of MMP9.
Collapse
Affiliation(s)
- Zhe Qiao
- Department of Thoracic Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Chengxue Dang
- Department of Surgical Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Bin Zhou
- Department of Thoracic Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Shaomin Li
- Department of Thoracic Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Wei Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Jiantao Jiang
- Department of Thoracic Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Jin Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Yuefeng Ma
- Department of Thoracic Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Ranran Kong
- Department of Thoracic Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Zhenchuan Ma
- Department of Thoracic Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| |
Collapse
|
67
|
Pascual F, Coleman RA. Fuel availability and fate in cardiac metabolism: A tale of two substrates. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:1425-33. [PMID: 26993579 DOI: 10.1016/j.bbalip.2016.03.014] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 03/10/2016] [Accepted: 03/11/2016] [Indexed: 12/12/2022]
Abstract
The heart's extraordinary metabolic flexibility allows it to adapt to normal changes in physiology in order to preserve its function. Alterations in the metabolic profile of the heart have also been attributed to pathological conditions such as ischemia and hypertrophy; however, research during the past decade has established that cardiac metabolic adaptations can precede the onset of pathologies. It is therefore critical to understand how changes in cardiac substrate availability and use trigger events that ultimately result in heart dysfunction. This review examines the mechanisms by which the heart obtains fuels from the circulation or from mobilization of intracellular stores. We next describe experimental models that exhibit either an increase in glucose use or a decrease in FA oxidation, and how these aberrant conditions affect cardiac metabolism and function. Finally, we highlight the importance of alternative, relatively under-investigated strategies for the treatment of heart failure. This article is part of a Special Issue entitled: Heart Lipid Metabolism edited by G.D. Lopaschuk.
Collapse
Affiliation(s)
- Florencia Pascual
- Department of Nutrition, University of North Carolina at Chapel Hill, 27599, USA.
| | - Rosalind A Coleman
- Department of Nutrition, University of North Carolina at Chapel Hill, 27599, USA.
| |
Collapse
|
68
|
Lawler PR, Akinkuolie AO, Chandler PD, Moorthy MV, Vandenburgh MJ, Schaumberg DA, Lee IM, Glynn RJ, Ridker PM, Buring JE, Mora S. Circulating N-Linked Glycoprotein Acetyls and Longitudinal Mortality Risk. Circ Res 2016; 118:1106-15. [PMID: 26951635 DOI: 10.1161/circresaha.115.308078] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 03/03/2016] [Indexed: 02/07/2023]
Abstract
RATIONALE Circulating glycoprotein N-acetyl glucosamine residues have recently been associated with incident cardiovascular disease and diabetes mellitus. OBJECTIVE Using a plasma glycan biosignature (GlycA) to identify circulating N-acetyl glycan groups, we examined the longitudinal association between GlycA and mortality among initially healthy individuals. METHODS AND RESULTS We quantified GlycA by 400 MHz (1)H nuclear magnetic resonance spectroscopy in 27,524 participants in the Women's Health Study (NCT00000479). The primary outcome was all-cause mortality. We replicated the findings in an independent cohort of 12,527 individuals in the Justification for the Use of statins in Prevention: an Intervention Trial Evaluating Rosuvastatin (JUPITER) trial (NCT00239681). We also undertook secondary examination of cardiovascular disease and cancer mortality in the Women's Health Study. In the Women's Health Study, during 524,515 person-years of follow-up (median, 20.5 years), there were 3523 deaths. Risk factor-adjusted multivariable Cox proportional hazard ratio (95% confidence interval) per SD increment in GlycA for all-cause mortality was significantly increased at 5 years (1.21 [1.06-1.40]) and during maximal follow-up (1.14 [1.09-1.16]). Similar risk for all-cause mortality was observed in the replication cohort (1.33 [1.21-1.45]). In the Women's Health Study, risk of cardiovascular disease mortality was increased at 5 years (1.43 [1.05-1.95]) and during maximal follow-up (1.15 [1.04-1.26]) and of cancer mortality at 5 years (1.23 [1.02-1.47]) and during maximal follow-up (1.08 [1.01-1.16]). Examination of correlations and mortality associations adjusted for high-sensitivity C-reactive protein, fibrinogen, and intercellular adhesion molecule-1, suggested that GlycA reflects summative risk related to multiple pathways of systemic inflammation. CONCLUSIONS Among initially healthy individuals, elevated baseline circulating glycoprotein N-acetyl methyl groups were associated with longitudinal risk of all-cause, cardiovascular, and cancer mortality.
Collapse
Affiliation(s)
- Patrick R Lawler
- From the Center for Lipid Metabolomics (P.R.L., A.O.A., S.M.), the Cardiovascular Division (P.R.L., P.MR., S.M.) and Division of Preventive Medicine (P.R.L., A.O.A., P.D.C., M.V.M., M.J.V., I.-M.L., R.J.G., P.MR., J.E.B., S.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Harvard Chan School of Public Health, Boston, MA (P.R.L., I-M.L., R.J.G., P.MR., J.E.B.), and Department of Ophthalmology and Visual Sciences, Center for Translational Medicine, John A. Moran Eye Center, University of Utah School of Medicine, Salt Lake City (D.A.S.)
| | - Akintunde O Akinkuolie
- From the Center for Lipid Metabolomics (P.R.L., A.O.A., S.M.), the Cardiovascular Division (P.R.L., P.MR., S.M.) and Division of Preventive Medicine (P.R.L., A.O.A., P.D.C., M.V.M., M.J.V., I.-M.L., R.J.G., P.MR., J.E.B., S.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Harvard Chan School of Public Health, Boston, MA (P.R.L., I-M.L., R.J.G., P.MR., J.E.B.), and Department of Ophthalmology and Visual Sciences, Center for Translational Medicine, John A. Moran Eye Center, University of Utah School of Medicine, Salt Lake City (D.A.S.)
| | - Paulette D Chandler
- From the Center for Lipid Metabolomics (P.R.L., A.O.A., S.M.), the Cardiovascular Division (P.R.L., P.MR., S.M.) and Division of Preventive Medicine (P.R.L., A.O.A., P.D.C., M.V.M., M.J.V., I.-M.L., R.J.G., P.MR., J.E.B., S.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Harvard Chan School of Public Health, Boston, MA (P.R.L., I-M.L., R.J.G., P.MR., J.E.B.), and Department of Ophthalmology and Visual Sciences, Center for Translational Medicine, John A. Moran Eye Center, University of Utah School of Medicine, Salt Lake City (D.A.S.)
| | - M Vinayaga Moorthy
- From the Center for Lipid Metabolomics (P.R.L., A.O.A., S.M.), the Cardiovascular Division (P.R.L., P.MR., S.M.) and Division of Preventive Medicine (P.R.L., A.O.A., P.D.C., M.V.M., M.J.V., I.-M.L., R.J.G., P.MR., J.E.B., S.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Harvard Chan School of Public Health, Boston, MA (P.R.L., I-M.L., R.J.G., P.MR., J.E.B.), and Department of Ophthalmology and Visual Sciences, Center for Translational Medicine, John A. Moran Eye Center, University of Utah School of Medicine, Salt Lake City (D.A.S.)
| | - Martin J Vandenburgh
- From the Center for Lipid Metabolomics (P.R.L., A.O.A., S.M.), the Cardiovascular Division (P.R.L., P.MR., S.M.) and Division of Preventive Medicine (P.R.L., A.O.A., P.D.C., M.V.M., M.J.V., I.-M.L., R.J.G., P.MR., J.E.B., S.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Harvard Chan School of Public Health, Boston, MA (P.R.L., I-M.L., R.J.G., P.MR., J.E.B.), and Department of Ophthalmology and Visual Sciences, Center for Translational Medicine, John A. Moran Eye Center, University of Utah School of Medicine, Salt Lake City (D.A.S.)
| | - Debra A Schaumberg
- From the Center for Lipid Metabolomics (P.R.L., A.O.A., S.M.), the Cardiovascular Division (P.R.L., P.MR., S.M.) and Division of Preventive Medicine (P.R.L., A.O.A., P.D.C., M.V.M., M.J.V., I.-M.L., R.J.G., P.MR., J.E.B., S.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Harvard Chan School of Public Health, Boston, MA (P.R.L., I-M.L., R.J.G., P.MR., J.E.B.), and Department of Ophthalmology and Visual Sciences, Center for Translational Medicine, John A. Moran Eye Center, University of Utah School of Medicine, Salt Lake City (D.A.S.)
| | - I-Min Lee
- From the Center for Lipid Metabolomics (P.R.L., A.O.A., S.M.), the Cardiovascular Division (P.R.L., P.MR., S.M.) and Division of Preventive Medicine (P.R.L., A.O.A., P.D.C., M.V.M., M.J.V., I.-M.L., R.J.G., P.MR., J.E.B., S.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Harvard Chan School of Public Health, Boston, MA (P.R.L., I-M.L., R.J.G., P.MR., J.E.B.), and Department of Ophthalmology and Visual Sciences, Center for Translational Medicine, John A. Moran Eye Center, University of Utah School of Medicine, Salt Lake City (D.A.S.)
| | - Robert J Glynn
- From the Center for Lipid Metabolomics (P.R.L., A.O.A., S.M.), the Cardiovascular Division (P.R.L., P.MR., S.M.) and Division of Preventive Medicine (P.R.L., A.O.A., P.D.C., M.V.M., M.J.V., I.-M.L., R.J.G., P.MR., J.E.B., S.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Harvard Chan School of Public Health, Boston, MA (P.R.L., I-M.L., R.J.G., P.MR., J.E.B.), and Department of Ophthalmology and Visual Sciences, Center for Translational Medicine, John A. Moran Eye Center, University of Utah School of Medicine, Salt Lake City (D.A.S.)
| | - Paul M Ridker
- From the Center for Lipid Metabolomics (P.R.L., A.O.A., S.M.), the Cardiovascular Division (P.R.L., P.MR., S.M.) and Division of Preventive Medicine (P.R.L., A.O.A., P.D.C., M.V.M., M.J.V., I.-M.L., R.J.G., P.MR., J.E.B., S.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Harvard Chan School of Public Health, Boston, MA (P.R.L., I-M.L., R.J.G., P.MR., J.E.B.), and Department of Ophthalmology and Visual Sciences, Center for Translational Medicine, John A. Moran Eye Center, University of Utah School of Medicine, Salt Lake City (D.A.S.)
| | - Julie E Buring
- From the Center for Lipid Metabolomics (P.R.L., A.O.A., S.M.), the Cardiovascular Division (P.R.L., P.MR., S.M.) and Division of Preventive Medicine (P.R.L., A.O.A., P.D.C., M.V.M., M.J.V., I.-M.L., R.J.G., P.MR., J.E.B., S.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Harvard Chan School of Public Health, Boston, MA (P.R.L., I-M.L., R.J.G., P.MR., J.E.B.), and Department of Ophthalmology and Visual Sciences, Center for Translational Medicine, John A. Moran Eye Center, University of Utah School of Medicine, Salt Lake City (D.A.S.)
| | - Samia Mora
- From the Center for Lipid Metabolomics (P.R.L., A.O.A., S.M.), the Cardiovascular Division (P.R.L., P.MR., S.M.) and Division of Preventive Medicine (P.R.L., A.O.A., P.D.C., M.V.M., M.J.V., I.-M.L., R.J.G., P.MR., J.E.B., S.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Harvard Chan School of Public Health, Boston, MA (P.R.L., I-M.L., R.J.G., P.MR., J.E.B.), and Department of Ophthalmology and Visual Sciences, Center for Translational Medicine, John A. Moran Eye Center, University of Utah School of Medicine, Salt Lake City (D.A.S.).
| |
Collapse
|
69
|
He K, Song S, Zou Z, Feng M, Wang D, Wang Y, Li X, Ye X. The Hypoglycemic and Synergistic Effect of Loganin, Morroniside, and Ursolic Acid Isolated from the Fruits of Cornus officinalis. Phytother Res 2016; 30:283-91. [PMID: 26619955 DOI: 10.1002/ptr.5529] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 10/17/2015] [Accepted: 11/06/2015] [Indexed: 01/05/2023]
Abstract
Hypoglycemic activity-guided separation of ethanol extracts from the fruits of Cornus officinalis Sieb. et Zucc (CO) led to the isolation of loganin, morroniside, and ursolic acid. The antidiabetic capacity of CO extracts and related compounds was further investigated in diabetes mellitus mice. The results suggested that both CO extracts and pure compounds could ameliorate diabetes-associated damages and complications. Oral administration of loganin and morroniside decreased fasting blood glucose levels in diabetes mellitus mice. Ursolic acid exhibited the highest reactive oxygen species scavenging activity and α-glucosidase inhibitory activity. Notably, we noticed an interesting synergistic effect between loganin and ursolic acid. Given these favorable hypoglycemic properties, C. officinalis, a food and medicinal plant in China, may be used as a valuable food supplement for the treatment of diabetes mellitus.
Collapse
Affiliation(s)
- Kai He
- School of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China
- Department of Clinical Laboratory, Hunan University of Medicine, Hunan, 418000, China
- Engineer Research Center of Chongqing Pharmaceutical Process and Quality Control, Chongqing, 400715, China
| | - Shanghua Song
- School of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China
- Engineer Research Center of Chongqing Pharmaceutical Process and Quality Control, Chongqing, 400715, China
| | - Zongyao Zou
- School of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China
- Engineer Research Center of Chongqing Pharmaceutical Process and Quality Control, Chongqing, 400715, China
| | - Min Feng
- School of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China
- College of Environmental and Biological Engineering, Chongqing Technology and Business University, Chongqing, 400067, China
| | - Dezhen Wang
- School of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China
- Engineer Research Center of Chongqing Pharmaceutical Process and Quality Control, Chongqing, 400715, China
| | - Yanzhi Wang
- School of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China
- Engineer Research Center of Chongqing Pharmaceutical Process and Quality Control, Chongqing, 400715, China
| | - Xuegang Li
- School of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China
- Engineer Research Center of Chongqing Pharmaceutical Process and Quality Control, Chongqing, 400715, China
| | - Xiaoli Ye
- School of Life Sciences, Southwest University, Chongqing, 400715, China
| |
Collapse
|
70
|
PPARs: Protectors or Opponents of Myocardial Function? PPAR Res 2015; 2015:835985. [PMID: 26713088 PMCID: PMC4680114 DOI: 10.1155/2015/835985] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 11/05/2015] [Accepted: 11/08/2015] [Indexed: 12/15/2022] Open
Abstract
Over 5 million people in the United States suffer from the complications of heart failure (HF), which is a rapidly expanding health complication. Disorders that contribute to HF include ischemic cardiac disease, cardiomyopathies, and hypertension. Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear receptor family. There are three PPAR isoforms: PPARα, PPARγ, and PPARδ. They can be activated by endogenous ligands, such as fatty acids, as well as by pharmacologic agents. Activators of PPARs are used for treating several metabolic complications, such as diabetes and hyperlipidemia that are directly or indirectly associated with HF. However, some of these drugs have adverse effects that compromise cardiac function. This review article aims to summarize the current basic and clinical research findings of the beneficial or detrimental effects of PPAR biology on myocardial function.
Collapse
|
71
|
Makino A, Dai A, Han Y, Youssef KD, Wang W, Donthamsetty R, Scott BT, Wang H, Dillmann WH. O-GlcNAcase overexpression reverses coronary endothelial cell dysfunction in type 1 diabetic mice. Am J Physiol Cell Physiol 2015; 309:C593-9. [PMID: 26269457 PMCID: PMC4628934 DOI: 10.1152/ajpcell.00069.2015] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 07/29/2015] [Indexed: 02/06/2023]
Abstract
Cardiovascular disease is the primary cause of morbidity and mortality in diabetes, and endothelial dysfunction is commonly seen in these patients. Increased O-linked N-acetylglucosamine (O-GlcNAc) protein modification is one of the central pathogenic features of diabetes. Modification of proteins by O-GlcNAc (O-GlcNAcylation) is regulated by two key enzymes: β-N-acetylglucosaminidase [O-GlcNAcase (OGA)], which catalyzes the reduction of protein O-GlcNAcylation, and O-GlcNAc transferase (OGT), which induces O-GlcNAcylation. However, it is not known whether reducing O-GlcNAcylation can improve endothelial dysfunction in diabetes. To examine the effect of endothelium-specific OGA overexpression on protein O-GlcNAcylation and coronary endothelial function in diabetic mice, we generated tetracycline-inducible, endothelium-specific OGA transgenic mice, and induced OGA by doxycycline administration in streptozotocin-induced type 1 diabetic mice. OGA protein expression was significantly decreased in mouse coronary endothelial cells (MCECs) isolated from diabetic mice compared with control MCECs, whereas OGT protein level was markedly increased. The level of protein O-GlcNAcylation was increased in diabetic compared with control mice, and OGA overexpression significantly decreased the level of protein O-GlcNAcylation in MCECs from diabetic mice. Capillary density in the left ventricle and endothelium-dependent relaxation in coronary arteries were significantly decreased in diabetes, while OGA overexpression increased capillary density to the control level and restored endothelium-dependent relaxation without changing endothelium-independent relaxation. We found that connexin 40 could be the potential target of O-GlcNAcylation that regulates the endothelial functions in diabetes. These data suggest that OGA overexpression in endothelial cells improves endothelial function and may have a beneficial effect on coronary vascular complications in diabetes.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/biosynthesis
- Antigens, Neoplasm/genetics
- Cells, Cultured
- Connexins/metabolism
- Coronary Artery Disease/enzymology
- Coronary Artery Disease/genetics
- Coronary Artery Disease/physiopathology
- Coronary Vessels/drug effects
- Coronary Vessels/enzymology
- Coronary Vessels/physiopathology
- Diabetes Mellitus, Experimental/enzymology
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/physiopathology
- Diabetes Mellitus, Type 1/enzymology
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/physiopathology
- Diabetic Angiopathies/enzymology
- Diabetic Angiopathies/genetics
- Diabetic Angiopathies/physiopathology
- Endothelial Cells/drug effects
- Endothelial Cells/enzymology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/enzymology
- Endothelium, Vascular/physiopathology
- Enzyme Induction
- Enzyme Inhibitors/pharmacology
- Glycosylation
- Histone Acetyltransferases/antagonists & inhibitors
- Histone Acetyltransferases/biosynthesis
- Histone Acetyltransferases/genetics
- Humans
- Hyaluronoglucosaminidase/antagonists & inhibitors
- Hyaluronoglucosaminidase/biosynthesis
- Hyaluronoglucosaminidase/genetics
- Male
- Mice, Transgenic
- N-Acetylglucosaminyltransferases/metabolism
- Neovascularization, Physiologic
- Protein Processing, Post-Translational
- Signal Transduction
- Vasodilation
- beta-N-Acetylhexosaminidases/antagonists & inhibitors
- beta-N-Acetylhexosaminidases/biosynthesis
- beta-N-Acetylhexosaminidases/genetics
- Gap Junction alpha-5 Protein
Collapse
Affiliation(s)
- Ayako Makino
- Department of Physiology, University of Arizona, Tucson, Arizona; Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and Department of Medicine, University of California, San Diego, La Jolla, California
| | - Anzhi Dai
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and Department of Medicine, University of California, San Diego, La Jolla, California
| | - Ying Han
- Department of Physiology, University of Arizona, Tucson, Arizona; Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | - Katia D Youssef
- Department of Physiology, University of Arizona, Tucson, Arizona
| | - Weihua Wang
- Department of Physiology, University of Arizona, Tucson, Arizona
| | - Reshma Donthamsetty
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | - Brian T Scott
- Department of Medicine, University of California, San Diego, La Jolla, California
| | - Hong Wang
- Department of Medicine, University of California, San Diego, La Jolla, California
| | - Wolfgang H Dillmann
- Department of Medicine, University of California, San Diego, La Jolla, California
| |
Collapse
|
72
|
Ramirez-Correa GA, Ma J, Slawson C, Zeidan Q, Lugo-Fagundo NS, Xu M, Shen X, Gao WD, Caceres V, Chakir K, DeVine L, Cole RN, Marchionni L, Paolocci N, Hart GW, Murphy AM. Removal of Abnormal Myofilament O-GlcNAcylation Restores Ca2+ Sensitivity in Diabetic Cardiac Muscle. Diabetes 2015; 64:3573-87. [PMID: 26109417 PMCID: PMC4587639 DOI: 10.2337/db14-1107] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 05/14/2015] [Indexed: 11/13/2022]
Abstract
Contractile dysfunction and increased deposition of O-linked β-N-acetyl-d-glucosamine (O-GlcNAc) in cardiac proteins are a hallmark of the diabetic heart. However, whether and how this posttranslational alteration contributes to lower cardiac function remains unclear. Using a refined β-elimination/Michael addition with tandem mass tags (TMT)-labeling proteomic technique, we show that CpOGA, a bacterial analog of O-GlcNAcase (OGA) that cleaves O-GlcNAc in vivo, removes site-specific O-GlcNAcylation from myofilaments, restoring Ca(2+) sensitivity in streptozotocin (STZ) diabetic cardiac muscles. We report that in control rat hearts, O-GlcNAc and O-GlcNAc transferase (OGT) are mainly localized at the Z-line, whereas OGA is at the A-band. Conversely, in diabetic hearts O-GlcNAc levels are increased and OGT and OGA delocalized. Consistent changes were found in human diabetic hearts. STZ diabetic hearts display increased physical interactions of OGA with α-actin, tropomyosin, and myosin light chain 1, along with reduced OGT and increased OGA activities. Our study is the first to reveal that specific removal of O-GlcNAcylation restores myofilament response to Ca(2+) in diabetic hearts and that altered O-GlcNAcylation is due to the subcellular redistribution of OGT and OGA rather than to changes in their overall activities. Thus, preventing sarcomeric OGT and OGA displacement represents a new possible strategy for treating diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Genaro A Ramirez-Correa
- Division of Cardiology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Junfeng Ma
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Chad Slawson
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS
| | - Quira Zeidan
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Nahyr S Lugo-Fagundo
- Division of Cardiology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Mingguo Xu
- Division of Cardiology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Xiaoxu Shen
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Wei Dong Gao
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Viviane Caceres
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Khalid Chakir
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Lauren DeVine
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Robert N Cole
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Luigi Marchionni
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Nazareno Paolocci
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Gerald W Hart
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Anne M Murphy
- Division of Cardiology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
73
|
Liu GD, Xu C, Feng L, Wang F. The augmentation of O-GlcNAcylation reduces glyoxal-induced cell injury by attenuating oxidative stress in human retinal microvascular endothelial cells. Int J Mol Med 2015; 36:1019-27. [PMID: 26311324 PMCID: PMC4564096 DOI: 10.3892/ijmm.2015.2319] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Accepted: 08/11/2015] [Indexed: 12/13/2022] Open
Abstract
It has recently been reported that O-linked β-N-acetyl glucosamine (O-GlcNAc) modification (a simple intracellular serine (Ser)/threonine (Thr)-linked monosaccharide) in human retinal microvascular endothelial cells (HRECs) is related to diabetic retinopathy (DR). During O-GlcNAcylation, O-GlcNAc is added to Ser and Thr residues. As the generation of reactive oxygen species (ROS) is one of the characteristics of advanced glycation end product (AGE) injury, and the most important key pathogenic factor of DR, in the present study, we aimed to investigate the association between O-GlcNAcylation and ROS generation in order to ascertain whether O-GlcNAcylation mitigates cellular injury through the generation of ROS. For this purpose, HRECs were divided into 4 groups as follows: HRECs treated with normal glucose (5 mM), HRECs treated with glyoxal (500 µM), glyoxal-treated HRECs also treated with 200 µM PUGNAc, and glyoxal-treated HRECs infected with O-GlcNAc transferase (OGT) siRNA. We detected increased O-GlcNAc levels and increased ROS production in the glyoxal-treated HRECs. The cellular redox status was determined by cellular ROS staining and by measuring the expression levels of the antioxidant genes, superoxide dismutase (SOD) and glutathione peroxidase (GPX). While the augmentation of O-GlcNAcylation following treatment with PUGNAc significantly attenuated the production of ROS (p<0.01) and increased the expression levels of SOD and GPX, the reduction of O-GlcNAcylation following infection with OGT siRNA, exacerbated the production of ROS (p<0.01) and decreased the expression of antioxidant genes. The effects of O-GlcNAcylation on the viability of HRECs were significant (p<0.01), particularly in the hydrogen peroxide (H2O2)-treated HRECs. Treatment with PUGNAc reduced glyoxal-induced cell apoptosis and transfection with OGT siRNA increased HREC apoptosis; these results were confirmed by flow cytometry and by the assessment of mitochondrial membrane potential. The augmentation of O-GlcNAcylation exerted cytoprotective effects on the HRECs by reducing the generation of ROS, increasing the expression of antioxidant genes, preventing the dissipation of mitochondrial membrane potential and preventing HREC apoptosis. Therefore, it can be concluded that O-GlcNAcylation plays a role in the early developmental process of DR.
Collapse
Affiliation(s)
- Guo Dong Liu
- Department of Ophthalmology, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Chong Xu
- Department of Ophthalmology, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Le Feng
- Department of Ophthalmology, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Fang Wang
- Department of Ophthalmology, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai 200072, P.R. China
| |
Collapse
|
74
|
High glucose induces mitochondrial dysfunction independently of protein O-GlcNAcylation. Biochem J 2015; 467:115-26. [PMID: 25627821 DOI: 10.1042/bj20141018] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Diabetes is characterized by hyperglycaemia and perturbations in intermediary metabolism. In particular, diabetes can augment flux through accessory pathways of glucose metabolism, such as the hexosamine biosynthetic pathway (HBP), which produces the sugar donor for the β-O-linked-N-acetylglucosamine (O-GlcNAc) post-translational modification of proteins. Diabetes also promotes mitochondrial dysfunction. Nevertheless, the relationships among diabetes, hyperglycaemia, mitochondrial dysfunction and O-GlcNAc modifications remain unclear. In the present study, we tested whether high-glucose-induced increases in O-GlcNAc modifications directly regulate mitochondrial function in isolated cardiomyocytes. Augmentation of O-GlcNAcylation with high glucose (33 mM) was associated with diminished basal and maximal cardiomyocyte respiration, a decreased mitochondrial reserve capacity and lower Complex II-dependent respiration (P<0.05); however, pharmacological or genetic modulation of O-GlcNAc modifications under normal or high glucose conditions showed few significant effects on mitochondrial respiration, suggesting that O-GlcNAc does not play a major role in regulating cardiomyocyte mitochondrial function. Furthermore, an osmotic control recapitulated high-glucose-induced changes to mitochondrial metabolism (P<0.05) without increasing O-GlcNAcylation. Thus, increased O-GlcNAcylation is neither sufficient nor necessary for high-glucose-induced suppression of mitochondrial metabolism in isolated cardiomyocytes.
Collapse
|
75
|
Kim EJ, Bond MR, Love DC, Hanover JA. Chemical tools to explore nutrient-driven O-GlcNAc cycling. Crit Rev Biochem Mol Biol 2015; 49:327-42. [PMID: 25039763 DOI: 10.3109/10409238.2014.931338] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Posttranslational modifications (PTM) including glycosylation, phosphorylation, acetylation, methylation and ubiquitination dynamically alter the proteome. The evolutionarily conserved enzymes O-linked N-acetylglucosamine (O-GlcNAc) transferase (OGT) and O-GlcNAcase are responsible for the addition and removal, respectively, of the nutrient-sensitive PTM of protein serine and threonine residues with O-GlcNAc. Indeed, the O-GlcNAc modification acts at every step in the "central dogma" of molecular biology and alters signaling pathways leading to amplified or blunted biological responses. The cellular roles of OGT and the dynamic PTM O-GlcNAc have been clarified with recently developed chemical tools including high-throughput assays, structural and mechanistic studies and potent enzyme inhibitors. These evolving chemical tools complement genetic and biochemical approaches for exposing the underlying biological information conferred by O-GlcNAc cycling.
Collapse
Affiliation(s)
- Eun J Kim
- Department of Science Education-Chemistry Major, Daegu University , Daegu , S. Korea and
| | | | | | | |
Collapse
|
76
|
Shin ES, Sorenson CM, Sheibani N. Diabetes and retinal vascular dysfunction. J Ophthalmic Vis Res 2015; 9:362-73. [PMID: 25667739 PMCID: PMC4307665 DOI: 10.4103/2008-322x.143378] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Accepted: 01/19/2014] [Indexed: 02/06/2023] Open
Abstract
Diabetes predominantly affects the microvascular circulation of the retina resulting in a range of structural changes unique to this tissue. These changes ultimately lead to altered permeability, hyperproliferation of endothelial cells and edema, and abnormal vascularization of the retina with resulting loss of vision. Enhanced production of inflammatory mediators and oxidative stress are primary insults with significant contribution to the pathogenesis of diabetic retinopathy (DR). We have determined the identity of the retinal vascular cells affected by hyperglycemia, and have delineated the cell autonomous impact of high glucose on function of these cells. We discuss some of the high glucose specific changes in retinal vascular cells and their contribution to retinal vascular dysfunction. This knowledge provides novel insight into the molecular and cellular defects contributing to the development and progression of diabetic retinopathy, and will aid in the development of innovative, as well as target specific therapeutic approaches for prevention and treatment of DR.
Collapse
Affiliation(s)
- Eui Seok Shin
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Christine M Sorenson
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA ; McPherson Eye Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Nader Sheibani
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA ; McPherson Eye Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
77
|
Glembotski CC. Finding the missing link between the unfolded protein response and O-GlcNAcylation in the heart. Circ Res 2014; 115:546-8. [PMID: 25170091 DOI: 10.1161/circresaha.114.304855] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
78
|
Marsh SA, Collins HE, Chatham JC. Protein O-GlcNAcylation and cardiovascular (patho)physiology. J Biol Chem 2014; 289:34449-56. [PMID: 25336635 DOI: 10.1074/jbc.r114.585984] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Our understanding of the role of protein O-GlcNAcylation in the regulation of the cardiovascular system has increased rapidly in recent years. Studies have linked increased O-GlcNAc levels to glucose toxicity and diabetic complications; conversely, acute activation of O-GlcNAcylation has been shown to be cardioprotective. However, it is also increasingly evident that O-GlcNAc turnover plays a central role in the delicate regulation of the cardiovascular system. Therefore, the goals of this minireview are to summarize our current understanding of how changes in O-GlcNAcylation influence cardiovascular pathophysiology and to highlight the evidence that O-GlcNAc cycling is critical for normal function of the cardiovascular system.
Collapse
Affiliation(s)
- Susan A Marsh
- From the Section of Experimental and Systems Pharmacology, College of Pharmacy, Washington State University, Spokane, Washington 99210-1495 and
| | - Helen E Collins
- the Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama 35294-0019
| | - John C Chatham
- the Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama 35294-0019
| |
Collapse
|
79
|
Rong J, Han J, Dong L, Tan Y, Yang H, Feng L, Wang QW, Meng R, Zhao J, Wang SQ, Chen X. Glycan Imaging in Intact Rat Hearts and Glycoproteomic Analysis Reveal the Upregulation of Sialylation during Cardiac Hypertrophy. J Am Chem Soc 2014; 136:17468-76. [DOI: 10.1021/ja508484c] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Jie Rong
- School
of Chemical Biology and Biotechnology, Shenzhen Graduate School of Peking University, Shenzhen 518055, China
| | | | | | | | | | - Lianshun Feng
- School
of Chemical Biology and Biotechnology, Shenzhen Graduate School of Peking University, Shenzhen 518055, China
| | | | | | - Jing Zhao
- School
of Chemical Biology and Biotechnology, Shenzhen Graduate School of Peking University, Shenzhen 518055, China
- State
Key
Laboratory of Pharmaceutical Biotechnology, School of Life Sciences,
Institute of Chemistry and Biomedical Sciences, Nanjing University, Nanjing 210093, China
| | | | | |
Collapse
|
80
|
Ding N, Ping L, Shi Y, Feng L, Zheng X, Song Y, Zhu J. Thiamet-G-mediated inhibition of O-GlcNAcase sensitizes human leukemia cells to microtubule-stabilizing agent paclitaxel. Biochem Biophys Res Commun 2014; 453:392-7. [PMID: 25268318 DOI: 10.1016/j.bbrc.2014.09.097] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 09/22/2014] [Indexed: 12/18/2022]
Abstract
Although the microtubule-stabilizing agent paclitaxel has been widely used for treatment of several cancer types, particularly for the malignancies of epithelia origin, it only shows limited efficacy on hematological malignancies. Emerging roles of O-GlcNAcylation modification of proteins in various cancer types have implicated the key enzymes catalyzing this reversible modification as targets for cancer therapy. Here, we show that the highly selective O-GlcNAcase (OGA) inhibitor thiamet-G significantly sensitized human leukemia cell lines to paclitaxel, with an approximate 10-fold leftward shift of IC50. Knockdown of OGA by siRNAs or inhibition of OGA by thiamet-G did not influence the cell viability. Furthermore, we demonstrated that thiamet-G binds to OGA in competition with 4-methylumbelliferyl N-acetyl-β-d-glucosaminide dehydrate, an analogue of O-GlcNAc UDP, thereby suppressing the activity of OGA. Importantly, inhibition of OGA by thiamet-G decreased the phosphorylation of microtubule-associated protein Tau and caused alterations of microtubule network in cells. It is noteworthy that paclitaxel combined with thiamet-G resulted in more profound perturbations on microtubule stability than did either one alone, which may implicate the underlying mechanism of thiamet-G-mediated sensitization of leukemia cells to paclitaxel. These findings thus suggest that a regimen of paclitaxel combined with OGA inhibitor might be more effective for the treatment of human leukemia.
Collapse
Affiliation(s)
- Ning Ding
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute, No. 52 Fucheng Road, Haidian District, Beijing 100142, China
| | - Lingyan Ping
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute, No. 52 Fucheng Road, Haidian District, Beijing 100142, China
| | - Yunfei Shi
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital & Institute, No. 52 Fucheng Road, Haidian District, Beijing 100142, China
| | - Lixia Feng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute, No. 52 Fucheng Road, Haidian District, Beijing 100142, China
| | - Xiaohui Zheng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute, No. 52 Fucheng Road, Haidian District, Beijing 100142, China
| | - Yuqin Song
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute, No. 52 Fucheng Road, Haidian District, Beijing 100142, China.
| | - Jun Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute, No. 52 Fucheng Road, Haidian District, Beijing 100142, China.
| |
Collapse
|
81
|
Verdegem D, Moens S, Stapor P, Carmeliet P. Endothelial cell metabolism: parallels and divergences with cancer cell metabolism. Cancer Metab 2014; 2:19. [PMID: 25250177 PMCID: PMC4171726 DOI: 10.1186/2049-3002-2-19] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 08/14/2014] [Indexed: 02/08/2023] Open
Abstract
The stromal vasculature in tumors is a vital conduit of nutrients and oxygen for cancer cells. To date, the vast majority of studies have focused on unraveling the genetic basis of vessel sprouting (also termed angiogenesis). In contrast to the widely studied changes in cancer cell metabolism, insight in the metabolic regulation of angiogenesis is only just emerging. These studies show that metabolic pathways in endothelial cells (ECs) importantly regulate angiogenesis in conjunction with genetic signals. In this review, we will highlight these emerging insights in EC metabolism and discuss them in perspective of cancer cell metabolism. While it is generally assumed that cancer cells have unique metabolic adaptations, not shared by healthy non-transformed cells, we will discuss parallels and highlight differences between endothelial and cancer cell metabolism and consider possible novel therapeutic opportunities arising from targeting both cancer and endothelial cells.
Collapse
Affiliation(s)
- Dries Verdegem
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, Department of Oncology, University of Leuven, Leuven 3000, Belgium ; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, K.U.Leuven, Campus Gasthuisberg, Herestraat 49, box 912, Leuven 3000, Belgium
| | - Stijn Moens
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, Department of Oncology, University of Leuven, Leuven 3000, Belgium ; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, K.U.Leuven, Campus Gasthuisberg, Herestraat 49, box 912, Leuven 3000, Belgium
| | - Peter Stapor
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, Department of Oncology, University of Leuven, Leuven 3000, Belgium ; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, K.U.Leuven, Campus Gasthuisberg, Herestraat 49, box 912, Leuven 3000, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, Department of Oncology, University of Leuven, Leuven 3000, Belgium ; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, K.U.Leuven, Campus Gasthuisberg, Herestraat 49, box 912, Leuven 3000, Belgium
| |
Collapse
|
82
|
Muthusamy S, DeMartino AM, Watson LJ, Brittian KR, Zafir A, Dassanayaka S, Hong KU, Jones SP. MicroRNA-539 is up-regulated in failing heart, and suppresses O-GlcNAcase expression. J Biol Chem 2014; 289:29665-76. [PMID: 25183011 DOI: 10.1074/jbc.m114.578682] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Derangements in metabolism and related signaling pathways characterize the failing heart. One such signal, O-linked β-N-acetylglucosamine (O-GlcNAc), is an essential post-translational modification regulated by two enzymes, O-GlcNAc transferase and O-GlcNAcase (OGA), which modulate the function of many nuclear and cytoplasmic proteins. We recently reported reduced OGA expression in the failing heart, which is consistent with the pro-adaptive role of increased O-GlcNAcylation during heart failure; however, molecular mechanisms regulating these enzymes during heart failure remain unknown. Using miRNA microarray analysis, we observed acute and chronic changes in expression of several miRNAs. Here, we focused on miR-539 because it was predicted to target OGA mRNA. Indeed, co-transfection of the OGA-3'UTR containing reporter plasmid and miR-539 overexpression plasmid significantly reduced reporter activity. Overexpression of miR-539 in neonatal rat cardiomyocytes significantly suppressed OGA expression and consequently increased O-GlcNAcylation; conversely, the miR-539 inhibitor rescued OGA protein expression and restored O-GlcNAcylation. In conclusion, this work identifies the first target of miR-539 in the heart and the first miRNA that regulates OGA. Manipulation of miR-539 may represent a novel therapeutic target in the treatment of heart failure and other metabolic diseases.
Collapse
Affiliation(s)
- Senthilkumar Muthusamy
- From the Institute of Molecular Cardiology, and, Diabetes and Obesity Center, University of Louisville, Louisville, Kentucky 40202
| | - Angelica M DeMartino
- From the Institute of Molecular Cardiology, and, Diabetes and Obesity Center, University of Louisville, Louisville, Kentucky 40202
| | - Lewis J Watson
- From the Institute of Molecular Cardiology, and, Diabetes and Obesity Center, University of Louisville, Louisville, Kentucky 40202
| | - Kenneth R Brittian
- From the Institute of Molecular Cardiology, and, Diabetes and Obesity Center, University of Louisville, Louisville, Kentucky 40202
| | - Ayesha Zafir
- From the Institute of Molecular Cardiology, and, Diabetes and Obesity Center, University of Louisville, Louisville, Kentucky 40202
| | - Sujith Dassanayaka
- From the Institute of Molecular Cardiology, and, Diabetes and Obesity Center, University of Louisville, Louisville, Kentucky 40202
| | - Kyung U Hong
- From the Institute of Molecular Cardiology, and, Diabetes and Obesity Center, University of Louisville, Louisville, Kentucky 40202
| | - Steven P Jones
- From the Institute of Molecular Cardiology, and, Diabetes and Obesity Center, University of Louisville, Louisville, Kentucky 40202
| |
Collapse
|
83
|
Cubedo J, Padró T, Badimon L. Glycoproteome of human apolipoprotein A-I: N- and O-glycosylated forms are increased in patients with acute myocardial infarction. Transl Res 2014; 164:209-22. [PMID: 24709669 DOI: 10.1016/j.trsl.2014.03.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 03/12/2014] [Accepted: 03/13/2014] [Indexed: 02/07/2023]
Abstract
High-density lipoprotein (HDL) functionality, which is closely associated with its composition and transport capabilities, determines its role in atheroprotection. During acute phase processes, HDL seems to lose its anti-inflammatory and cytoprotective properties. In this study, we hypothesized that after an acute myocardial infarction apolipoprotein (Apo) A-I, the main protein component of HDL, might undergo changes in its molecular processing. Therefore, we have characterized the Apo A-I proteome during the evolution of new-onset acute myocardial infarction (AMI). To this end, serum Apo A-I was characterized by 2-dimensional electrophoresis/mass-spectrometry in controls and AMI patients at admission (within the first 6 hours after pain onset) and 8 hours, 16 hours, 24 hours, and 3 days afterward. The Apo A-I glycoproteome was analyzed by lectin-based glycoprotein isolation methods and deglycosylation assays, and Apo A-I serum levels were evaluated by enzyme-linked immunosorbent assay (ELISA). The Apo A-I proteomic signature (5 spots: 28 kDa/pI:5-5.75) was significantly altered in AMI patients 3 days after the event with respect to controls. Increased levels of N- and O-glycosylated Apo A-I forms were found post-AMI. Apo A-I serum levels measured by ELISA were significantly changed and related to left ventricular ejection fraction, troponin-T, and C-reactive protein. The Apo A-I molecule measured by ELISA corresponded to the main glycosylated spots and was specifically O-GlcNAcylated in AMI patients. Therefore, our results demonstrate that Apo A-I is both N- and O-glycosylated and that there is an increase in Apo A-I glycosylation after AMI. Furthermore, the specific increase in the O-GlcNAcylated forms could have a relevant prognostic value and a protective role in the evolution of AMI.
Collapse
Affiliation(s)
- Judit Cubedo
- Cardiovascular Research Center (CSIC-ICCC), Barcelona, Spain; Sant Pau Biomedical Research Institute Sant Pau, Barcelona, Spain
| | - Teresa Padró
- Cardiovascular Research Center (CSIC-ICCC), Barcelona, Spain; Sant Pau Biomedical Research Institute Sant Pau, Barcelona, Spain
| | - Lina Badimon
- Cardiovascular Research Center (CSIC-ICCC), Barcelona, Spain; Sant Pau Biomedical Research Institute Sant Pau, Barcelona, Spain; Cardiovascular Research Chair UAB, Barcelona, Spain.
| |
Collapse
|
84
|
Myslicki JP, Belke DD, Shearer J. Role of O-GlcNAcylation in nutritional sensing, insulin resistance and in mediating the benefits of exercise. Appl Physiol Nutr Metab 2014; 39:1205-13. [PMID: 25203141 DOI: 10.1139/apnm-2014-0122] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The purpose of this review is to highlight the role of O-linked β-N-acetylglucosamine (O-GlcNAc) protein modification in metabolic disease states and to summarize current knowledge of how exercise affects this important post-translational signalling pathway. O-GlcNAc modification is an intracellular tool capable of integrating energy supply with demand. The accumulation of excess energy associated with obesity and insulin resistance is mediated, in part, by the hexosamine biosynthetic pathway (HBP), which results in the O-GlcNAcylation of a myriad of proteins, thereby affecting their respective function, stability, and localization. Insulin resistance is related to the excessive O-GlcNAcylation of key metabolic proteins causing a chronic blunting of insulin signalling pathways and precipitating the accompanying pathologies, such as heart and kidney disease. Lifestyle modifications such as diet and exercise also modify the pathway. Exercise is a front-line and cost-effective therapeutic approach for insulin resistance, and recent work shows that the intervention can alter O-GlcNAc gene expression, signalling, and protein modification. However, there is currently no consensus on the effect of frequency, intensity, type, and duration of exercise on O-GlcNAc modification, the HBP, and its related enzymes. On one end of the spectrum, mild, prolonged swim training reduces O-GlcNAcylation, while on the other end, higher intensity treadmill running increases cardiac protein O-GlcNAc modification. Clearly, a balance between acute and chronic stress of exercise is needed to reap the benefits of the intervention on O-GlcNAc signalling.
Collapse
Affiliation(s)
- Jason P Myslicki
- a Faculty of Kinesiology, University of Calgary, Calgary, AB T2N 1N4, Canada
| | | | | |
Collapse
|
85
|
Abstract
The discovery of adult cardiac stem cells (CSCs) and their potential to restore functional cardiac tissue has fueled unprecedented interest in recent years. Indeed, stem-cell–based therapies have the potential to transform the treatment and prognosis of heart failure, for they have the potential to eliminate the underlying cause of the disease by reconstituting the damaged heart with functional cardiac cells. Over the last decade, several independent laboratories have demonstrated the utility of c-kit+/Lin- resident CSCs in alleviating left ventricular dysfunction and remodeling in animal models of acute and chronic myocardial infarction. Recently, the first clinical trial of autologous CSCs for treatment of heart failure resulting from ischemic heart disease (Stem Cell Infusion in Patients with Ischemic cardiOmyopathy [SCIPIO]) has been conducted, and the interim results are quite promising. In this phase I trial, no adverse effects attributable to the CSC treatment have been noted, and CSC-treated patients showed a significant improvement in ejection fraction at 1 year (+13.7 absolute units versus baseline), accompanied by a 30.2 % reduction in infarct size. Moreover, the CSC-induced enhancement in cardiac structure and function was associated with a significant improvement in the New York Heart Association (NYHA) functional class and in the quality of life, as measured by the Minnesota Living with Heart failure Questionnaire. These results are exciting and warrant larger, phase II studies. However, CSC therapy for cardiac repair is still in its infancy, and many hurdles need to be overcome to further enhance the therapeutic efficacy of CSCs.
Collapse
Affiliation(s)
- Kyung U Hong
- Institute of Molecular Cardiology, Division of Cardiovascular Medicine, Department of Medicine, University of Louisville, Louisville, KY, 40202, USA,
| | | |
Collapse
|
86
|
Aging leads to elevation of O-GlcNAcylation and disruption of mitochondrial homeostasis in retina. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:425705. [PMID: 24987494 PMCID: PMC4060167 DOI: 10.1155/2014/425705] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 05/09/2014] [Indexed: 12/21/2022]
Abstract
Retina is particularly susceptible to aging as oxidative damage accumulates within retina, leading to age-related retinal dysfunction or even visual loss. However, the underlying mechanisms still remain obscure and effective therapeutic strategy is urgently in need. Here, we quested for the answer particularly focusing on mitochondrial homeostasis and O-GlcNAcylation in rat retina. By comparing expression of electron transfer chain complexes and key factors in mitochondrial biogenesis and dynamics in retinas of aged and young Sprague-Dawley rats, we found that mitochondrial Complex I, II, IV and V were increased in aged retina with decreased mtTFA and Mfn2. Also, we noticed that p38 and JNK of MAPK signaling were substantially more activated in aged retina, suggesting stress induction. In addition, we found that pan-O-GlcNAcylation was remarkably stronger with lower OGA expression in aged retina. To further elucidate the roles of Mfn2 and O-GlcNAcylation, we employed ARPE-19 cells and found that ATP production, oxygen consumption, and mitochondrial membrane potential were reduced and ROS level was increased by Mfn2 knockdown, while treating with PUGNAc or UDP-GlcNAc heightened oxygen consumption and reduced ROS. Our results suggest disrupted mitochondrial homeostasis may increase oxidative stress; yet enhanced O-GlcNAcylation might defend against oxidative stress and promote mitochondrial respiration in aged retina.
Collapse
|
87
|
Gurel Z, Zaro BW, Pratt MR, Sheibani N. Identification of O-GlcNAc modification targets in mouse retinal pericytes: implication of p53 in pathogenesis of diabetic retinopathy. PLoS One 2014; 9:e95561. [PMID: 24788674 PMCID: PMC4006792 DOI: 10.1371/journal.pone.0095561] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 03/28/2014] [Indexed: 12/31/2022] Open
Abstract
Hyperglycemia is the primary cause of the majority of diabetes complications, including diabetic retinopathy (DR). Hyperglycemic conditions have a detrimental effect on many tissues and cell types, especially the retinal vascular cells including early loss of pericytes (PC). However, the mechanisms behind this selective sensitivity of retinal PC to hyperglycemia are undefined. The O-linked β-N-acetylglucosamine (O-GlcNAc) modification is elevated under hyperglycemic condition, and thus, may present an important molecular modification impacting the hyperglycemia-driven complications of diabetes. We have recently demonstrated that the level of O-GlcNAc modification in response to high glucose is variable in various retinal vascular cells. Retinal PC responded with the highest increase in O-GlcNAc modification compared to retinal endothelial cells and astrocytes. Here we show that these differences translated into functional changes, with an increase in apoptosis of retinal PC, not just under high glucose but also under treatment with O-GlcNAc modification inducers, PUGNAc and Thiamet-G. To gain insight into the molecular mechanisms involved, we have used click-It chemistry and LC-MS analysis and identified 431 target proteins of O-GlcNAc modification in retinal PC using an alkynyl-modified GlcNAc analog (GlcNAlk). Among the O-GlcNAc target proteins identified here 115 of them were not previously reported to be target of O-GlcNAc modification. We have identified at least 34 of these proteins with important roles in various aspects of cell death processes. Our results indicated that increased O-GlcNAc modification of p53 was associated with an increase in its protein levels in retinal PC. Together our results suggest that post-translational O-GlcNAc modification of p53 and its increased levels may contribute to selective early loss of PC during diabetes. Thus, modulation of O-GlcNAc modification may provide a novel treatment strategy to prevent the initiation and progression of DR.
Collapse
Affiliation(s)
- Zafer Gurel
- Department of Ophthalmology and Visual Sciences, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin, United States of America; McPherson Eye Research Institute, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Balyn W Zaro
- Departments of Chemistry and Molecular and Computational Biology, University of Southern California, Los Angeles, California, United States of America
| | - Matthew R Pratt
- Departments of Chemistry and Molecular and Computational Biology, University of Southern California, Los Angeles, California, United States of America
| | - Nader Sheibani
- Department of Ophthalmology and Visual Sciences, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin, United States of America; McPherson Eye Research Institute, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin, United States of America
| |
Collapse
|
88
|
Harwood KR, Hanover JA. Nutrient-driven O-GlcNAc cycling - think globally but act locally. J Cell Sci 2014; 127:1857-67. [PMID: 24762810 DOI: 10.1242/jcs.113233] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Proper cellular functioning requires that cellular machinery behave in a spatiotemporally regulated manner in response to global changes in nutrient availability. Mounting evidence suggests that one way this is achieved is through the establishment of physically defined gradients of O-GlcNAcylation (O-linked addition of N-acetylglucosamine to serine and threonine residues) and O-GlcNAc turnover. Because O-GlcNAcylation levels are dependent on the nutrient-responsive hexosamine signaling pathway, this modification is uniquely poised to inform upon the nutritive state of an organism. The enzymes responsible for O-GlcNAc addition and removal are encoded by a single pair of genes: both the O-GlcNAc transferase (OGT) and the O-GlcNAcase (OGA, also known as MGEA5) genes are alternatively spliced, producing protein variants that are targeted to discrete cellular locations where they must selectively recognize hundreds of protein substrates. Recent reports suggest that in addition to their catalytic functions, OGT and OGA use their multifunctional domains to anchor O-GlcNAc cycling to discrete intracellular sites, thus allowing them to establish gradients of deacetylase, kinase and phosphatase signaling activities. The localized signaling gradients established by targeted O-GlcNAc cycling influence many important cellular processes, including lipid droplet remodeling, mitochondrial functioning, epigenetic control of gene expression and proteostasis. As such, the tethering of the enzymes of O-GlcNAc cycling appears to play a role in ensuring proper spatiotemporal responses to global alterations in nutrient supply.
Collapse
Affiliation(s)
- Katryn R Harwood
- Laboratory of Cell and Molecular Biology, NIDDK, National Institutes of Health, Bethesda MD 20892-0851, USA
| | | |
Collapse
|
89
|
Abstract
The metabolic adaptations that support oncogenic growth can also render cancer cells dependent on certain nutrients. Along with the Warburg effect, increased utilization of glutamine is one of the metabolic hallmarks of the transformed state. Glutamine catabolism is positively regulated by multiple oncogenic signals, including those transmitted by the Rho family of GTPases and by c-Myc. The recent identification of mechanistically distinct inhibitors of glutaminase, which can selectively block cellular transformation, has revived interest in the possibility of targeting glutamine metabolism in cancer therapy. Here, we outline the regulation and roles of glutamine metabolism within cancer cells and discuss possible strategies for, and the consequences of, impacting these processes therapeutically.
Collapse
|
90
|
Wu YL, Lin AH, Chen CH, Huang WC, Wang HY, Liu MH, Lee TS, Ru Kou Y. Glucosamine attenuates cigarette smoke-induced lung inflammation by inhibiting ROS-sensitive inflammatory signaling. Free Radic Biol Med 2014; 69:208-18. [PMID: 24486342 DOI: 10.1016/j.freeradbiomed.2014.01.026] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 12/18/2013] [Accepted: 01/21/2014] [Indexed: 12/18/2022]
Abstract
Cigarette smoking causes persistent lung inflammation that is mainly regulated by redox-sensitive pathways. We have reported that cigarette smoke (CS) activates a NADPH oxidase-dependent reactive oxygen species (ROS)-sensitive AMP-activated protein kinase (AMPK) signaling pathway leading to induction of lung inflammation. Glucosamine, a dietary supplement used to treat osteoarthritis, has antioxidant and anti-inflammatory properties. However, whether glucosamine has similar beneficial effects against CS-induced lung inflammation remains unclear. Using a murine model we show that chronic CS exposure for 4 weeks increased lung levels of 4-hydroxynonenal (an oxidative stress biomarker), phospho-AMPK, and macrophage inflammatory protein 2 and induced lung inflammation; all of these CS-induced events were suppressed by chronic treatment with glucosamine. Using human bronchial epithelial cells, we demonstrate that cigarette smoke extract (CSE) sequentially activated NADPH oxidase; increased intracellular levels of ROS; activated AMPK, mitogen-activated protein kinases (MAPKs), nuclear factor-κB (NF-κB), and signal transducer and activator of transcription proteins 3 (STAT3); and induced interleukin-8 (IL-8). Additionally, using a ROS scavenger, a siRNA that targets AMPK, and various pharmacological inhibitors, we identified the signaling cascade that leads to induction of IL-8 by CSE. All these CSE-induced events were inhibited by glucosamine pretreatment. Our findings suggest a novel role for glucosamine in alleviating the oxidative stress and lung inflammation induced by chronic CS exposure in vivo and in suppressing the CSE-induced IL-8 in vitro by inhibiting both the ROS-sensitive NADPH oxidase/AMPK/MAPK signaling pathway and the downstream transcriptional factors NF-κB and STAT3.
Collapse
Affiliation(s)
- Yuh-Lin Wu
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan
| | - An-Hsuan Lin
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan
| | - Chao-Hung Chen
- Division of Thoracic Surgery, Mackay Memorial Hospital, Taipei, Taiwan; Department of Cosmetic Applications and Management, Mackay Medicine, Nursing and Management College, Taipei, Taiwan
| | - Wen-Chien Huang
- Division of Thoracic Surgery, Mackay Memorial Hospital, Taipei, Taiwan; Institute of Traditional Medicine, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan
| | - Hsin-Yi Wang
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan
| | - Meng-Han Liu
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan
| | - Tzong-Shyuan Lee
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan.
| | - Yu Ru Kou
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan.
| |
Collapse
|
91
|
Glucosamine for osteoarthritis: biological effects, clinical efficacy, and safety on glucose metabolism. ARTHRITIS 2014; 2014:432463. [PMID: 24678419 PMCID: PMC3941227 DOI: 10.1155/2014/432463] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Accepted: 12/20/2013] [Indexed: 01/01/2023]
Abstract
Osteoarthritis is a chronic degenerative disorder that currently represents one of the main causes of disability within the elderly population and an important presenting complaint overall. The pathophysiologic basis of osteoarthritis entails a complex group of interactions among biochemical and mechanical factors that have been better characterized in light of a recent spike in research on the subject. This has led to an ongoing search for ideal therapeutic management schemes for these patients, where glucosamine is one of the most frequently used alternatives worldwide due to their chondroprotective properties and their long-term effects. Its use in the treatment of osteoarthritis is well established; yet despite being considered effective by many research groups, controversy surrounds their true effectiveness. This situation stems from several methodological aspects which hinder appropriate data analysis and comparison in this context, particularly regarding objectives and target variables. Similar difficulties surround the assessment of the potential ability of glucosamine formulations to alter glucose metabolism. Nevertheless, evidence supporting diabetogenesis by glucosamine remains scarce in humans, and to date, this association should be considered only a theoretical possibility.
Collapse
|
92
|
Huynh K, Bernardo BC, McMullen JR, Ritchie RH. Diabetic cardiomyopathy: mechanisms and new treatment strategies targeting antioxidant signaling pathways. Pharmacol Ther 2014; 142:375-415. [PMID: 24462787 DOI: 10.1016/j.pharmthera.2014.01.003] [Citation(s) in RCA: 425] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Accepted: 01/08/2014] [Indexed: 12/14/2022]
Abstract
Cardiovascular disease is the primary cause of morbidity and mortality among the diabetic population. Both experimental and clinical evidence suggest that diabetic subjects are predisposed to a distinct cardiomyopathy, independent of concomitant macro- and microvascular disorders. 'Diabetic cardiomyopathy' is characterized by early impairments in diastolic function, accompanied by the development of cardiomyocyte hypertrophy, myocardial fibrosis and cardiomyocyte apoptosis. The pathophysiology underlying diabetes-induced cardiac damage is complex and multifactorial, with elevated oxidative stress as a key contributor. We now review the current evidence of molecular disturbances present in the diabetic heart, and their role in the development of diabetes-induced impairments in myocardial function and structure. Our focus incorporates both the contribution of increased reactive oxygen species production and reduced antioxidant defenses to diabetic cardiomyopathy, together with modulation of protein signaling pathways and the emerging role of protein O-GlcNAcylation and miRNA dysregulation in the progression of diabetic heart disease. Lastly, we discuss both conventional and novel therapeutic approaches for the treatment of left ventricular dysfunction in diabetic patients, from inhibition of the renin-angiotensin-aldosterone-system, through recent evidence favoring supplementation of endogenous antioxidants for the treatment of diabetic cardiomyopathy. Novel therapeutic strategies, such as gene therapy targeting the phosphoinositide 3-kinase PI3K(p110α) signaling pathway, and miRNA dysregulation, are also reviewed. Targeting redox stress and protective protein signaling pathways may represent a future strategy for combating the ever-increasing incidence of heart failure in the diabetic population.
Collapse
Affiliation(s)
- Karina Huynh
- Baker IDI Heart & Diabetes Institute, Melbourne, Australia; Department of Medicine, Monash University, Clayton, Victoria, Australia
| | | | - Julie R McMullen
- Baker IDI Heart & Diabetes Institute, Melbourne, Australia; Department of Medicine, Monash University, Clayton, Victoria, Australia; Department of Physiology, Monash University, Clayton, Victoria, Australia.
| | - Rebecca H Ritchie
- Baker IDI Heart & Diabetes Institute, Melbourne, Australia; Department of Medicine, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
93
|
Xu C, Liu G, Liu X, Wang F. O-GlcNAcylation under hypoxic conditions and its effects on the blood-retinal barrier in diabetic retinopathy. Int J Mol Med 2013; 33:624-32. [PMID: 24366041 DOI: 10.3892/ijmm.2013.1597] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2013] [Accepted: 12/12/2013] [Indexed: 11/06/2022] Open
Abstract
An increase in O-linked N-acetylglucosamine (O-GlcNAc) protein modifications has been observerd in db/db mouse retinas. O-GlcNAc-modified proteins in the db/db mouse retina have been shown to be localized in the ganglion cell layer, the inner nuclear layer, the retina pigment epithelium (RPE) layer and the inner plexiform layer, in which hypoxia-inducible factor 1α (HIF1α) has also been shown to be localized. In the current study, we examined whether hypoxia increases O-GlcNAcylation in retinal vascular cells under high glucose conditions and whether HIF1α activation is consistent with the response to and activation of O-GlcNAcylation in retinal lesions in diabetic retinopathy. In addition, the effects of O-GlcNAcylation on the blood-retinal barrier were verified in vitro by the inhibition of O-GlcNAcylation. A time-dependent increase in the O-GlcNAcylation in bovine retinal vascular endothelial cells (BRVECs) was observed following incubation of the cells with high glucose medium (glucose 4.5 g/l) under hypoxic (1-3% O2) conditions. Hypoxia-induced BRVEC O-GlcNAcylation was not observed when the BRVECs were transfected with siRNA targeting O-GlcNAc transferase (OGT) or treated with alloxan (an OGT inhibitor) prior to exposure to high glucose. The increase in BRVEC O-GlcNAcylation induced by high glucose, as well as by thiamet G [an O-GlcNAcase (OGA) inhibitor] led to a reduction in occludin expression levels in vitro, which was prevented by treatment with OGT siRNA and alloxan. In conclusion, the current study demonstrates the relationship between O-GlcNAc glycosylation and hypoxia during diabetic retinopathy and that hyperglycemia induced O2 consumption activates HIF1α and O-GlcNAc modification protein in the same retinal layer. The reduced protein BRVEC O-GlcNAcylation levels exert protective effects on the blood-retinal barrier and thus represent a potential therapeutic target for the treatment of diabetic retinopathy.
Collapse
Affiliation(s)
- Chong Xu
- Department of Ophthalmology, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Guodong Liu
- Department of Ophthalmology, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Xiaoqiao Liu
- Department of Ophthalmology, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Fang Wang
- Department of Ophthalmology, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai 200072, P.R. China
| |
Collapse
|
94
|
Dassanayaka S, Jones SP. O-GlcNAc and the cardiovascular system. Pharmacol Ther 2013; 142:62-71. [PMID: 24287310 DOI: 10.1016/j.pharmthera.2013.11.005] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 11/01/2013] [Indexed: 12/28/2022]
Abstract
The cardiovascular system is capable of robust changes in response to physiologic and pathologic stimuli through intricate signaling mechanisms. The area of metabolism has witnessed a veritable renaissance in the cardiovascular system. In particular, the post-translational β-O-linkage of N-acetylglucosamine (O-GlcNAc) to cellular proteins represents one such signaling pathway that has been implicated in the pathophysiology of cardiovascular disease. This highly dynamic protein modification may induce functional changes in proteins and regulate key cellular processes including translation, transcription, and cell death. In addition, its potential interplay with phosphorylation provides an additional layer of complexity to post-translational regulation. The hexosamine biosynthetic pathway generally requires glucose to form the nucleotide sugar, UDP-GlcNAc. Accordingly, O-GlcNAcylation may be altered in response to nutrient availability and cellular stress. Recent literature supports O-GlcNAcylation as an autoprotective response in models of acute stress (hypoxia, ischemia, oxidative stress). Models of sustained stress, such as pressure overload hypertrophy, and infarct-induced heart failure, may also require protein O-GlcNAcylation as a partial compensatory mechanism. Yet, in models of Type II diabetes, O-GlcNAcylation has been implicated in the subsequent development of vascular, and even cardiac, dysfunction. This review will address this apparent paradox and discuss the potential mechanisms of O-GlcNAc-mediated cardioprotection and cardiovascular dysfunction. This discussion will also address potential targets for pharmacologic interventions and the unique considerations related to such targets.
Collapse
Affiliation(s)
- Sujith Dassanayaka
- Institute of Molecular Cardiology, Diabetes and Obesity Center, Division of Cardiovascular Medicine, Department of Medicine, University of Louisville, Louisville, KY, USA; Department of Physiology and Biophysics, University of Louisville, Louisville, KY, USA
| | - Steven P Jones
- Institute of Molecular Cardiology, Diabetes and Obesity Center, Division of Cardiovascular Medicine, Department of Medicine, University of Louisville, Louisville, KY, USA; Department of Physiology and Biophysics, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
95
|
Watson LJ, Long BW, DeMartino AM, Brittian KR, Readnower RD, Brainard RE, Cummins TD, Annamalai L, Hill BG, Jones SP. Cardiomyocyte Ogt is essential for postnatal viability. Am J Physiol Heart Circ Physiol 2013; 306:H142-53. [PMID: 24186210 DOI: 10.1152/ajpheart.00438.2013] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The singly coded gene O-linked-β-N-acetylglucosamine (O-GlcNAc) transferase (Ogt) resides on the X chromosome and is necessary for embryonic stem cell viability during embryogenesis. In mature cells, this enzyme catalyzes the posttranslational modification known as O-GlcNAc to various cellular proteins. Several groups, including our own, have shown that acute increases in protein O-GlcNAcylation are cardioprotective both in vitro and in vivo. Yet, little is known about how OGT affects cardiac function because total body knockout (KO) animals are not viable. Presently, we sought to establish the potential involvement of cardiomyocyte Ogt in cardiac maturation. Initially, we characterized a constitutive cardiomyocyte-specific (cm)OGT KO (c-cmOGT KO) mouse and found that only 12% of the c-cmOGT KO mice survived to weaning age (4 wk old); the surviving animals were smaller than their wild-type littermates, had dilated hearts, and showed overt signs of heart failure. Dysfunctional c-cmOGT KO hearts were more fibrotic, apoptotic, and hypertrophic. Several glycolytic genes were also upregulated; however, there were no gross changes in mitochondrial O2 consumption. Histopathology of the KO hearts indicated the potential involvement of endoplasmic reticulum stress, directing us to evaluate expression of 78-kDa glucose-regulated protein and protein disulfide isomerase, which were elevated. Additional groups of mice were subjected to inducible deletion of cmOGT, which did not produce overt dysfunction within the first couple of weeks of deletion. Yet, long-term loss (via inducible deletion) of cmOGT produced gradual and progressive cardiomyopathy. Thus, cardiomyocyte Ogt is necessary for maturation of the mammalian heart, and inducible deletion of cmOGT in the adult mouse produces progressive ventricular dysfunction.
Collapse
Affiliation(s)
- Lewis J Watson
- Department of Physiology and Biophysics, University of Louisville, Louisville, Kentucky; and
| | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Abstract
The heart has a high rate of ATP production and turnover that is required to maintain its continuous mechanical work. Perturbations in ATP-generating processes may therefore affect contractile function directly. Characterizing cardiac metabolism in heart failure (HF) revealed several metabolic alterations called metabolic remodeling, ranging from changes in substrate use to mitochondrial dysfunction, ultimately resulting in ATP deficiency and impaired contractility. However, ATP depletion is not the only relevant consequence of metabolic remodeling during HF. By providing cellular building blocks and signaling molecules, metabolic pathways control essential processes such as cell growth and regeneration. Thus, alterations in cardiac metabolism may also affect the progression to HF by mechanisms beyond ATP supply. Our aim is therefore to highlight that metabolic remodeling in HF not only results in impaired cardiac energetics but also induces other processes implicated in the development of HF such as structural remodeling and oxidative stress. Accordingly, modulating cardiac metabolism in HF may have significant therapeutic relevance that goes beyond the energetic aspect.
Collapse
Affiliation(s)
- Torsten Doenst
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich-Schiller-University Jena, Germany.
| | | | | |
Collapse
|
97
|
Zafir A, Readnower R, Long BW, McCracken J, Aird A, Alvarez A, Cummins TD, Li Q, Hill BG, Bhatnagar A, Prabhu SD, Bolli R, Jones SP. Protein O-GlcNAcylation is a novel cytoprotective signal in cardiac stem cells. Stem Cells 2013; 31:765-75. [PMID: 23335157 DOI: 10.1002/stem.1325] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 12/14/2012] [Indexed: 01/02/2023]
Abstract
Clinical trials demonstrate the regenerative potential of cardiac stem cell (CSC) therapy in the postinfarcted heart. Despite these encouraging preliminary clinical findings, the basic biology of these cells remains largely unexplored. The principal requirement for cell transplantation is to effectively prime them for survival within the unfavorable environment of the infarcted myocardium. In the adult mammalian heart, the β-O-linkage of N-acetylglucosamine (i.e., O-GlcNAc) to proteins is a unique post-translational modification that confers cardioprotection from various otherwise lethal stressors. It is not known whether this signaling system exists in CSCs. In this study, we demonstrate that protein O-GlcNAcylation is an inducible stress response in adult murine Sca-1(+) /lin(-) CSCs and exerts an essential prosurvival role. Posthypoxic CSCs responded by time-dependently increasing protein O-GlcNAcylation upon reoxygenation. We used pharmacological interventions for loss- and gain-of-function, that is, enzymatic inhibition of O-GlcNAc transferase (OGT) (adds the O-GlcNAc modification to proteins) by TT04, or inhibition of OGA (removes O-GlcNAc) by thiamet-G (ThG). Reduction in the O-GlcNAc signal (via TT04, or OGT gene deletion using Cre-mediated recombination) significantly sensitized CSCs to posthypoxic injury, whereas augmenting O-GlcNAc levels (via ThG) enhanced cell survival. Diminished O-GlcNAc levels render CSCs more susceptible to the onset of posthypoxic apoptotic processes via elevated poly(ADP-ribose) polymerase cleavage due to enhanced caspase-3/7 activation, whereas promoting O-GlcNAcylation can serve as a pre-emptive antiapoptotic signal regulating the survival of CSCs. Thus, we report the primary demonstration of protein O-GlcNAcylation as an important prosurvival signal in CSCs, which could enhance CSC survival prior to in vivo autologous transfer.
Collapse
Affiliation(s)
- Ayesha Zafir
- Department of Medicine, Division of Cardiovascular Medicine, Institute of Molecular Cardiology, Diabetes and Obesity Center, University of Louisville, Louisville, Kentucky 40202, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Assrir N, Richez C, Durand P, Guittet E, Badet B, Lescop E, Badet-Denisot MA. Mapping the UDP-N-acetylglucosamine regulatory site of human glucosamine-6P synthase by saturation-transfer difference NMR and site-directed mutagenesis. Biochimie 2013; 97:39-48. [PMID: 24075873 DOI: 10.1016/j.biochi.2013.09.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 09/13/2013] [Indexed: 10/26/2022]
Abstract
The enzyme glucosamine-6P Synthase (Gfat, L-glutamine:D-fructose-6P amidotransferase) is involved in the hexosamine biosynthetic pathway and catalyzes the formation of glucosamine-6P from the substrates d-fructose-6-phosphate and l-glutamine. In eukaryotic cells, Gfat is inhibited by UDPGlcNAc, the end product of the biochemical pathway. In this work we present the dissection of the binding and inhibition properties of this feedback inhibitor and of its fragments by a combination of STD-NMR experiments and inhibition measurements on the wild type human enzyme (hGfat) as well as on site-directed mutants. We demonstrate that the UDPGlcNAc binding site is located in the isomerase domain of hGfat. Two amino acid residues (G445 and G461) located at the bottom of the binding site are identified to play a key role in the specificity of UDPGlcNAc inhibition of hGfat activity vs its bacterial Escherichia coli counterpart. We also show that UDPGlcNAc subcomponents have distinct features: the nucleotidic moiety is entirely responsible for binding whereas the N-acetyl group is mandatory for inhibition but not for binding, and the sugar moiety acts as a linker between the nucleotidic and N-acetyl groups. Combining these structural recognition determinants therefore appears as a promising strategy to selectively inhibit hGfat, which may for example help reduce complications in diabetes.
Collapse
Affiliation(s)
- Nadine Assrir
- Institut de Chimie des Substances Naturelles, Centre de Recherche de Gif, UPR2301, CNRS, 1 avenue de la Terrasse, 91198 Gif-sur-Yvette, France
| | - Celine Richez
- Institut de Chimie des Substances Naturelles, Centre de Recherche de Gif, UPR2301, CNRS, 1 avenue de la Terrasse, 91198 Gif-sur-Yvette, France
| | - Philippe Durand
- Institut de Chimie des Substances Naturelles, Centre de Recherche de Gif, UPR2301, CNRS, 1 avenue de la Terrasse, 91198 Gif-sur-Yvette, France
| | - Eric Guittet
- Institut de Chimie des Substances Naturelles, Centre de Recherche de Gif, UPR2301, CNRS, 1 avenue de la Terrasse, 91198 Gif-sur-Yvette, France
| | - Bernard Badet
- Institut de Chimie des Substances Naturelles, Centre de Recherche de Gif, UPR2301, CNRS, 1 avenue de la Terrasse, 91198 Gif-sur-Yvette, France
| | - Ewen Lescop
- Institut de Chimie des Substances Naturelles, Centre de Recherche de Gif, UPR2301, CNRS, 1 avenue de la Terrasse, 91198 Gif-sur-Yvette, France.
| | - Marie-Ange Badet-Denisot
- Institut de Chimie des Substances Naturelles, Centre de Recherche de Gif, UPR2301, CNRS, 1 avenue de la Terrasse, 91198 Gif-sur-Yvette, France.
| |
Collapse
|
99
|
Krenz M, Baines C, Kalogeris T, Korthuis R. Cell Survival Programs and Ischemia/Reperfusion: Hormesis, Preconditioning, and Cardioprotection. ACTA ACUST UNITED AC 2013. [DOI: 10.4199/c00090ed1v01y201309isp044] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
100
|
Groves JA, Lee A, Yildirir G, Zachara NE. Dynamic O-GlcNAcylation and its roles in the cellular stress response and homeostasis. Cell Stress Chaperones 2013; 18:535-58. [PMID: 23620203 PMCID: PMC3745259 DOI: 10.1007/s12192-013-0426-y] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 03/29/2013] [Accepted: 04/01/2013] [Indexed: 12/15/2022] Open
Abstract
O-linked N-acetyl-β-D-glucosamine (O-GlcNAc) is a ubiquitous and dynamic post-translational modification known to modify over 3,000 nuclear, cytoplasmic, and mitochondrial eukaryotic proteins. Addition of O-GlcNAc to proteins is catalyzed by the O-GlcNAc transferase and is removed by a neutral-N-acetyl-β-glucosaminidase (O-GlcNAcase). O-GlcNAc is thought to regulate proteins in a manner analogous to protein phosphorylation, and the cycling of this carbohydrate modification regulates many cellular functions such as the cellular stress response. Diverse forms of cellular stress and tissue injury result in enhanced O-GlcNAc modification, or O-GlcNAcylation, of numerous intracellular proteins. Stress-induced O-GlcNAcylation appears to promote cell/tissue survival by regulating a multitude of biological processes including: the phosphoinositide 3-kinase/Akt pathway, heat shock protein expression, calcium homeostasis, levels of reactive oxygen species, ER stress, protein stability, mitochondrial dynamics, and inflammation. Here, we will discuss the regulation of these processes by O-GlcNAc and the impact of such regulation on survival in models of ischemia reperfusion injury and trauma hemorrhage. We will also discuss the misregulation of O-GlcNAc in diseases commonly associated with the stress response, namely Alzheimer's and Parkinson's diseases. Finally, we will highlight recent advancements in the tools and technologies used to study the O-GlcNAc modification.
Collapse
Affiliation(s)
- Jennifer A. Groves
- The Department of Biological Chemistry, The Johns Hopkins University School of Medicine, 725 N. Wolfe St, Baltimore, MD 21205-2185 USA
| | - Albert Lee
- The Department of Biological Chemistry, The Johns Hopkins University School of Medicine, 725 N. Wolfe St, Baltimore, MD 21205-2185 USA
| | - Gokben Yildirir
- The Department of Biological Chemistry, The Johns Hopkins University School of Medicine, 725 N. Wolfe St, Baltimore, MD 21205-2185 USA
| | - Natasha E. Zachara
- The Department of Biological Chemistry, The Johns Hopkins University School of Medicine, 725 N. Wolfe St, Baltimore, MD 21205-2185 USA
| |
Collapse
|