Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Am J Physiol Heart Circ Physiol ; 298(3): H1022-8, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20061542

ABSTRACT

The lipid mediator sphingosine 1-phosphate (S1P) confers survival benefits in cardiomyocytes and isolated hearts subjected to oxidative stress. High-density lipoprotein (HDL) is a major carrier of S1P in the serum, but whether HDL-associated S1P directly mediates survival in a preparation composed exclusively of cardiomyocytes has not been demonstrated. Accordingly, we tested the hypothesis that signal activation and survival during simulated ischemia-reperfusion injury in response to HDL require lipoprotein-associated S1P. As a model, we used adult mouse cardiomyocytes subjected to hypoxia-reoxygenation. Cells were treated or not with autologous mouse HDL, which significantly increased myocyte viability as measured by trypan blue exclusion. This survival effect was abrogated by the S1P(1) and SIP(3) receptor antagonist VPC 23019. The selective S1P(3) antagonist CAY10444, the G(i) antagonist pertussis toxin, the MEK (MAPK/ERK) kinase inhibitor PD-98059, and the phosphoinositide-3 kinase inhibitor wortmannin also inhibited the prosurvival effect of HDL. We observed that HDL activated both Akt (protein kinase B) and the MEK1/2-ERK1/2 pathway and also stimulated phosphorylation of glycogen synthase kinase-3beta. ERK1/2 activation was through an S1P(1) subtype receptor-G(i) protein-dependent pathway, whereas the activation of Akt was inhibited by CAY10444, indicating mediation by S1P(3) subtype receptors. We conclude that HDL, via its cargo of S1P, can directly protect cardiomyocytes against simulated oxidative injury in the absence of vascular effects and that prosurvival signal activation is dependent on both S1P(1) and S1P(3) subtype receptors.


Subject(s)
Lipoproteins, HDL/pharmacology , Lysophospholipids/physiology , Myocardial Reperfusion Injury/physiopathology , Myocytes, Cardiac/drug effects , Sphingosine/analogs & derivatives , Animals , Cell Survival/drug effects , Cells, Cultured , Disease Models, Animal , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , MAP Kinase Kinase 1/physiology , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 3/physiology , Myocytes, Cardiac/pathology , Oxidative Stress/physiology , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction/physiology , Sphingosine/physiology
2.
J Cardiovasc Pharmacol ; 53(6): 486-94, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19433984

ABSTRACT

We examined the ability of sphingosine-1-phosphate (S1P) to desensitize extracellular signal-related kinase (ERK), a mitogen-activated protein kinase linked to antiapoptotic responses in the heart. In isolated adult mouse cardiomyocytes, S1P (10 nM-5 microM) induced ERK phosphorylation in a time- and dose-dependent manner. S1P stimulation of ERK was completely inhibited by an S1P1/3 subtype receptor antagonist (VPC23019), by a Gi protein inhibitor (pertussis toxin) and by a mitogen-activated protein kinase/ERK kinase inhibitor (PD98059). A selective S1P3 receptor antagonist (CAY10444) had no effect on S1P-induced ERK activation. The selective S1P1 agonist SEW2871 also induced ERK phosphorylation. Activation of ERK by restimulation with 100 nM S1P was suppressed after 1 hour of preincubation with 100 nM S1P but recovered fully the next day, suggesting receptor recycling. Similar results were obtained in protein kinase C epsilon-null cardiomyocytes. Treatment with the nonselective S1P receptor agonist FTY720 for 1 hour also reduced phospho-ERK expression in response to subsequent S1P stimulation. In contrast to S1P, some desensitization to FTY720 persisted after overnight exposure. Cell death induced by hypoxia/reoxygenation was reduced by pretreatment with exogenous S1P. This enhanced survival was abrogated by pretreatment with PD98059, VPC23019, or pertussis toxin. Thus, exogenous S1P induces rapid and reversible S1P1-mediated ERK phosphorylation. S1P-induced adult mouse cardiomyocyte survival requires ERK activation mediated via an S1P1-Gi pathway.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Myocytes, Cardiac/metabolism , Receptors, Lysosphingolipid/physiology , Animals , Cell Death/drug effects , Cell Hypoxia , Cell Survival/drug effects , Cells, Cultured , Enzyme Activation , In Vitro Techniques , Lysophospholipids/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/drug effects , Phosphorylation , Protein Kinase C/genetics , Receptors, Lysosphingolipid/agonists , Receptors, Lysosphingolipid/antagonists & inhibitors , Signal Transduction , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Thiazolidines/pharmacology
3.
Cardiovasc Res ; 76(1): 41-50, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17610857

ABSTRACT

OBJECTIVE: Sphingosine kinase (SphK) is a key enzyme in the synthesis of sphingosine 1-phosphate (S1P), a bioactive sphingolipid. SphK is involved in ischemic preconditioning (IPC). To date no studies in genetically altered animals have examined the role of SphK1 in myocardial ischemia/reperfusion (IR) injury and IPC. METHODS AND RESULTS: Wild-type and SphK1 null mouse hearts were subjected to IR (50 min global ischemia and 40 min reperfusion) in a Langendorff apparatus. IPC consisted of 2 min of global ischemia and 2 min of reperfusion for two cycles. At baseline, there were no differences in left ventricular developed pressure (LVDP), +/-dP/dtmax, and LV end-diastolic pressure (EDP) between SphK1 mutant and wild-type (WT) mouse hearts. In the mutants, total SphK enzyme activity was reduced by 44% and S1P levels were decreased by 41%. SphK1 null hearts subjected to IR exhibited more cardiac damage compared with WT: LVDP and +/-dP/dtmax decreased, LVEDP increased, and infarct size increased (n=6, P<0.05). Apoptosis was markedly enhanced in SphK1 mutant IR mouse hearts. IPC was cardioprotective in WT hearts, but this protection appeared to be ineffective in SphK1 null hearts. There was no change in infarct size in the IPC+IR group compared to the IR group in the null hearts (50.1+/-5.0% vs 45.0+/-3.8%, n=6, P=NS). IPC remained ineffective in the null hearts even when the index ischemia time was shortened by 10 min. CONCLUSIONS: Deletion of the SphK1 gene sensitizes the myocardium to IR injury and appears to impair the protective effect of IPC. These data provide the first genetic evidence that the SphK1-S1P pathway is a critical mediator of IPC and cell survival.


Subject(s)
Mutation , Myocardium/enzymology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Reperfusion Injury/enzymology , Animals , Apoptosis , Blotting, Western/methods , Creatine Kinase/analysis , Creatine Kinase/metabolism , Disease Susceptibility , In Situ Nick-End Labeling , Ischemic Preconditioning, Myocardial , Lysophospholipids/metabolism , Mice , Mice, Knockout , Myocardium/pathology , Perfusion , Phosphotransferases (Alcohol Group Acceptor)/analysis , Phosphotransferases (Alcohol Group Acceptor)/genetics , Reperfusion Injury/pathology , Sphingosine/analogs & derivatives , Sphingosine/metabolism
4.
Circ Res ; 92(2): 203-11, 2003 Feb 07.
Article in English | MEDLINE | ID: mdl-12574148

ABSTRACT

AlphaB-crystallin (alphaBC), a small heat shock protein expressed in high levels in the heart, is phosphorylated on Ser-19, 45, and 59 after stress. However, it is not known whether alphaBC phosphorylation directly affects cell survival. In the present study, constructs were prepared that encode forms of alphaBC harboring Ser to Ala (blocks phosphorylation) or Ser to Glu (mimics phosphorylation) mutations at positions 19, 45, and 59. The effects of each form on apoptosis of cultured cardiac myocytes after hyperosmotic or hypoxic stress were assessed. Compared with controls, cells that expressed alphaBC with Ser to Ala substitutions at all three positions, alphaBC(AAA), exhibited more stress-induced apoptosis. Cells expressing either alphaBC(AAE) or (EEE) exhibited 3-fold less apoptosis than cells expressing alphaBC(AAA), indicating that phosphorylation of Ser-59 confers protection. alphaBC is known to bind to procaspase-3 and to decrease caspase-3 activation. Compared with cells expressing alphaBC(AAA), the activation of caspase-3 was decreased by 3-fold in cells expressing alphaBC(AAE). These results demonstrate that mimicking the phosphorylation of alphaBC on Ser-59 is necessary and sufficient to confer caspase-3 inhibition and protection of cardiac myocytes against hyperosmotic or hypoxic stress. These findings provide direct evidence that alphaBC(S59P) contributes to the cardioprotection observed after physiologically relevant stresses, such as transient hypoxia. Identifying the targets of alphaBC(S59P) will reveal important details about the mechanism underlying the cytoprotective effects of this small heat shock protein.


Subject(s)
Apoptosis/physiology , Myocytes, Cardiac/metabolism , alpha-Crystallin B Chain/genetics , Amino Acid Substitution , Animals , Apoptosis/drug effects , Base Sequence , Caspase 3 , Caspases/metabolism , Cell Hypoxia/physiology , Cell Survival , Cells, Cultured , In Situ Nick-End Labeling , Molecular Mimicry/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Osmolar Concentration , Phosphorylation , Rats , Rats, Sprague-Dawley , Sorbitol/pharmacology , Structure-Activity Relationship , Transfection , alpha-Crystallin B Chain/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...