Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 7: 41166, 2017 01 27.
Article in English | MEDLINE | ID: mdl-28128227

ABSTRACT

Hyperglycemia (HG) stimulates the production of reactive oxygen species in the heart through activation of NADPH oxidase 2 (NOX2). This production is independent of glucose metabolism but requires sodium/glucose cotransporters (SGLT). Seven SGLT isoforms (SGLT1 to 6 and sodium-myoinositol cotransporter-1, SMIT1) are known, although their expression and function in the heart remain elusive. We investigated these 7 isoforms and found that only SGLT1 and SMIT1 were expressed in mouse, rat and human hearts. In cardiomyocytes, galactose (transported through SGLT1) did not activate NOX2. Accordingly, SGLT1 deficiency did not prevent HG-induced NOX2 activation, ruling it out in the cellular response to HG. In contrast, myo-inositol (transported through SMIT1) reproduced the toxic effects of HG. SMIT1 overexpression exacerbated glucotoxicity and sensitized cardiomyocytes to HG, whereas its deletion prevented HG-induced NOX2 activation. In conclusion, our results show that heart SMIT1 senses HG and triggers NOX2 activation. This could participate in the redox signaling in hyperglycemic heart and contribute to the pathophysiology of diabetic cardiomyopathy.


Subject(s)
Heat-Shock Proteins/metabolism , Hyperglycemia/metabolism , Myocardium/metabolism , NADPH Oxidase 2/metabolism , Reactive Oxygen Species/metabolism , Symporters/metabolism , Animals , Disease Models, Animal , Gene Knockout Techniques , Heat-Shock Proteins/genetics , Humans , Male , Mice , Rats , Sodium-Glucose Transporter 1 , Symporters/genetics
2.
Am J Physiol Heart Circ Physiol ; 307(8): H1120-33, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25128166

ABSTRACT

Exposure of cardiomyocytes to high glucose concentrations (HG) stimulates reactive oxygen species (ROS) production by NADPH oxidase (NOX2). NOX2 activation is triggered by enhanced glucose transport through a sodium-glucose cotransporter (SGLT) but not by a stimulation of glucose metabolism. The aim of this work was to identify potential therapeutic approaches to counteract this glucotoxicity. In cultured adult rat cardiomyocytes incubated with 21 mM glucose (HG), AMP-activated protein kinase (AMPK) activation by A769662 or phenformin nearly suppressed ROS production. Interestingly, glucagon-like peptide 1 (GLP-1), a new antidiabetic drug, concomitantly induced AMPK activation and prevented the HG-mediated ROS production (maximal effect at 100 nM). α2-AMPK, the major isoform expressed in cardiomyocytes (but not α1-AMPK), was activated in response to GLP-1. Anti-ROS properties of AMPK activators were not related to changes in glucose uptake or glycolysis. Using in situ proximity ligation assay, we demonstrated that AMPK activation prevented the HG-induced p47phox translocation to caveolae, whatever the AMPK activators used. NOX2 activation by either α-methyl-d-glucopyranoside, a glucose analog transported through SGLT, or angiotensin II was also counteracted by GLP-1. The crucial role of AMPK in limiting HG-mediated NOX2 activation was demonstrated by overexpressing a constitutively active form of α2-AMPK using adenoviral infection. This overexpression prevented NOX2 activation in response to HG, whereas GLP-1 lost its protective action in α2-AMPK-deficient mouse cardiomyocytes. Under HG, the GLP-1/AMPK pathway inhibited PKC-ß2 phosphorylation, a key element mediating p47phox translocation. In conclusion, GLP-1 induces α2-AMPK activation and blocks HG-induced p47phox translocation to the plasma membrane, thereby preventing glucotoxicity.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Glucagon-Like Peptide 1/pharmacology , Glucose/pharmacology , Hypoglycemic Agents/pharmacology , Myocytes, Cardiac/metabolism , NADPH Oxidases/metabolism , AMP-Activated Protein Kinases/genetics , Animals , Biphenyl Compounds , Cells, Cultured , Male , Membrane Glycoproteins/metabolism , Methylglucosides/pharmacology , Myocytes, Cardiac/drug effects , NADPH Oxidase 2 , NADPH Oxidases/genetics , Phenformin/pharmacology , Protein Kinase C/metabolism , Protein Transport , Pyrones/pharmacology , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Thiophenes/pharmacology
3.
J Mol Cell Cardiol ; 74: 32-43, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24805196

ABSTRACT

Cardiac fibroblasts (CF) are crucial in left ventricular (LV) healing and remodeling after myocardial infarction (MI). They are typically activated into myofibroblasts that express alpha-smooth muscle actin (α-SMA) microfilaments and contribute to the formation of contractile and mature collagen scars that minimize the adverse dilatation of infarcted areas. CF predominantly express the α1 catalytic subunit of AMP-activated protein kinase (AMPKα1), while AMPKα2 is the major catalytic isoform in cardiomyocytes. AMPKα2 is known to protect the heart by preserving the energy charge of cardiac myocytes during injury, but whether AMPKα1 interferes with maladaptative heart responses remains unexplored. In this study, we investigated the role of AMPKα1 in modulating LV dilatation and CF fibrosis during post-MI remodeling. AMPKα1 knockout (KO) and wild type (WT) mice were subjected to permanent ligation of the left anterior descending coronary artery. The absence of AMPKα1 was associated with increased CF proliferation in infarcted areas, while expression of the myodifferentiation marker α-SMA was decreased. Faulty maturation of myofibroblasts might derive from severe down-regulation of the non-canonical transforming growth factor-beta1/p38 mitogen-activated protein kinase (TGF-ß1/p38 MAPK) pathway in KO infarcts. In addition, lysyl oxidase (LOX) protein expression was dramatically reduced in the scar of KO hearts. Although infarct size was similar in AMPK-KO and WT hearts subjected to MI, these changes resulted in compromised scar contractility, defective scar collagen maturation, and exacerbated adverse remodeling, as indicated by increased LV diastolic dimension 30days after MI. Our data genetically demonstrate the centrality of AMPKα1 in post-MI scar formation and highlight the specificity of this catalytic isoform in cardiac fibroblast/myofibroblast biology.


Subject(s)
AMP-Activated Protein Kinases/genetics , Cicatrix/genetics , Myocardial Contraction , Myocardial Infarction/genetics , Myofibroblasts/enzymology , Ventricular Remodeling , AMP-Activated Protein Kinases/deficiency , AMP-Activated Protein Kinases/metabolism , Actins/genetics , Actins/metabolism , Animals , Cell Proliferation , Cicatrix/enzymology , Cicatrix/pathology , Cicatrix/physiopathology , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Female , Gene Expression Regulation , Mice , Mice, Knockout , Myocardial Infarction/enzymology , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/enzymology , Myocardium/pathology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Myofibroblasts/pathology , Protein-Lysine 6-Oxidase/genetics , Protein-Lysine 6-Oxidase/metabolism , Signal Transduction , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Am J Physiol Heart Circ Physiol ; 306(12): H1619-30, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24748590

ABSTRACT

AMP-activated protein kinase (AMPK), a key cellular sensor of energy, regulates metabolic homeostasis and plays a protective role in the ischemic or diabetic heart. Stimulation of cardiac glucose uptake contributes to this AMPK-mediated protection. The small-molecule AMPK activator A-769662, which binds and directly activates AMPK, has recently been characterized. A-769662-dependent AMPK activation protects the heart against an ischemia-reperfusion episode but is unable to stimulate skeletal muscle glucose uptake. Here, we tried to reconcile these conflicting findings by investigating the impact of A-769662 on cardiac AMPK signaling and glucose uptake. We showed that A-769662 promoted AMPK activation, resulting in the phosphorylation of several downstream targets, but was incapable of stimulating glucose uptake in cultured cardiomyocytes and the perfused heart. The lack of glucose uptake stimulation can be explained by A-769662's narrow specificity, since it selectively activates cardiac AMPK heterotrimeric complexes containing α2/ß1-subunits, the others being presumably required for this metabolic outcome. However, when combined with classical AMPK activators, such as metformin, phenformin, oligomycin, or hypoxia, which impact AMPK heterotrimers more broadly via elevation of cellular AMP levels, A-769662 induced more profound AMPK phosphorylation and subsequent glucose uptake stimulation. The synergistic effect of A-769662 under such ischemia-mimetic conditions protected cardiomyocytes against ROS production and cell death. In conclusion, despite the fact that A-769662 activates AMPK, it alone does not significantly stimulate glucose uptake. However, strikingly, its ability of potentiating the action on other AMPK activators makes it a potentially useful participant in the protective role of AMPK in the heart.


Subject(s)
AMP-Activated Protein Kinases/drug effects , AMP-Activated Protein Kinases/metabolism , Glucose/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Pyrones/pharmacology , Thiophenes/pharmacology , Adenosine Monophosphate/metabolism , Animals , Biphenyl Compounds , Cells, Cultured , Insulin/pharmacology , Male , Models, Animal , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/drug effects , Phenformin/pharmacology , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
5.
Cardiovasc Res ; 101(1): 20-9, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24104879

ABSTRACT

AIMS: Mesenchymal stem cells (MSCs) are widely used for cell therapy, particularly for the treatment of ischaemic heart disease. Mechanisms underlying control of their metabolism and proliferation capacity, critical elements for their survival and differentiation, have not been fully characterized. AMP-activated protein kinase (AMPK) is a key regulator known to metabolically protect cardiomyocytes against ischaemic injuries and, more generally, to inhibit cell proliferation. We hypothesized that AMPK plays a role in control of MSC metabolism and proliferation. METHODS AND RESULTS: MSCs isolated from murine bone marrow exclusively expressed the AMPKα1 catalytic subunit. In contrast to cardiomyocytes, a chronic exposure of MSCs to hypoxia failed to induce cell death despite the absence of AMPK activation. This hypoxic tolerance was the consequence of a preference of MSC towards glycolytic metabolism independently of oxygen availability and AMPK signalling. On the other hand, A-769662, a well-characterized AMPK activator, was able to induce a robust and sustained AMPK activation. We showed that A-769662-induced AMPK activation inhibited MSC proliferation. Proliferation was not arrested in MSCs derived from AMPKα1-knockout mice, providing genetic evidence that AMPK is essential for this process. Among AMPK downstream targets proposed to regulate cell proliferation, we showed that neither the p70 ribosomal S6 protein kinase/eukaryotic elongation factor 2-dependent protein synthesis pathway nor p21 was involved, whereas p27 expression was increased by A-769662. Silencing p27 expression partially prevented the A-769662-dependent inhibition of MSC proliferation. CONCLUSION: MSCs resist hypoxia independently of AMPK whereas chronic AMPK activation inhibits MSC proliferation, p27 being involved in this regulation.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Hypoxia/enzymology , Mesenchymal Stem Cells/enzymology , Myocytes, Cardiac/enzymology , Animals , Biphenyl Compounds , Cell Proliferation , Cell Survival , Cell- and Tissue-Based Therapy , Cells, Cultured , Elongation Factor 2 Kinase/metabolism , Enzyme Activation , Heart Diseases/therapy , Hypoxia/physiopathology , Isoenzymes/metabolism , Mice , Mitochondrial Turnover , Pyrones , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Thiophenes , p21-Activated Kinases/metabolism
6.
Cardiovasc Res ; 92(2): 237-46, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21859816

ABSTRACT

AIMS: Exposure to high glucose (HG) stimulates reactive oxygen species (ROS) production by NADPH oxidase in cardiomyocytes, but the underlying mechanism remains elusive. In this study, we have dissected the link between glucose transport and metabolism and NADPH oxidase activation under hyperglycaemic conditions. METHODS AND RESULTS: Primary cultures of adult rat cardiomyocytes were exposed to HG concentration (HG, 21 mM) and compared with the normal glucose level (LG, 5 mM). HG exposure activated Rac1GTP and induced p47phox translocation to the plasma membrane, resulting in NADPH oxidase (NOX2) activation, increased ROS production, insulin resistance, and eventually cell death. Comparison of the level of O-linked N-acetylglucosamine (O-GlcNAc) residues in LG- and HG-treated cells did not reveal any significant difference. Inhibition of the pentose phosphate pathway (PPP) by 6-aminonicotinamide counteracted ROS production in response to HG but did not prevent Rac-1 upregulation and p47phox translocation leading to NOX2 activation. Modulation of glucose uptake barely affected oxidative stress and toxicity induced by HG. More interestingly, non-metabolizable glucose analogues (i.e. 3-O-methyl-D-glucopyranoside and α-methyl-D-glucopyranoside) reproduced the toxic effect of HG. Inhibition of the sodium/glucose cotransporter SGLT1 by phlorizin counteracted HG-induced NOX2 activation and ROS production. CONCLUSION: Increased glucose metabolism by itself does not trigger NADPH oxidase activation, although PPP is required to provide NOX2 with NADPH and to produce ROS. NOX2 activation results from glucose transport through SGLT1, suggesting that an extracellular metabolic signal transduces into an intracellular ionic signal.


Subject(s)
Glucose/metabolism , Hyperglycemia/enzymology , Membrane Glycoproteins/metabolism , Myocytes, Cardiac/enzymology , NADPH Oxidases/metabolism , Sodium-Glucose Transporter 1/metabolism , 6-Aminonicotinamide/pharmacology , Acetylglucosamine/metabolism , Animals , Cell Death , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Activation , Glucose/analogs & derivatives , Glycosylation , Hyperglycemia/pathology , Hyperglycemia/physiopathology , Insulin Resistance , Male , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , NADPH Oxidase 2 , Pentose Phosphate Pathway/drug effects , Phlorhizin/pharmacology , Protein Processing, Post-Translational , Protein Transport , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Sodium-Glucose Transporter 1/antagonists & inhibitors , rac1 GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...