Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Cardiovasc Med (Hagerstown) ; 14(10): 698-704, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23695182

ABSTRACT

The history began in the 19th century with Ascanio Sobrero (1812-1888), the discoverer of glycerol trinitrate (nitroglycerine, NTG), and with Angelo Angeli (1864-1931), the discoverer of sodium trioxodinitrate (Angeli's salt). It is likely that Angeli and Sobrero never met, but their two histories will join each other more than a century later. In fact, it has been discovered that both NTG and Angeli's salt are able to induce a preconditioning effect. As NTG has a long history as an antianginal drug its newly discovered property as a preconditioning agent has also been tested in humans. Angeli's salt properties as a preconditioning and inotropic agent have only been tested in animals so far.


Subject(s)
Cardiovascular Agents/therapeutic use , Myocardial Reperfusion Injury/prevention & control , Nitric Oxide Donors/therapeutic use , Nitrites/therapeutic use , Nitroglycerin/therapeutic use , Vasodilator Agents/therapeutic use , Animals , Cardiovascular Agents/history , Cardiovascular Agents/metabolism , History, 19th Century , History, 20th Century , Humans , Myocardial Reperfusion Injury/history , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/physiopathology , Nitric Oxide/metabolism , Nitric Oxide Donors/history , Nitric Oxide Donors/metabolism , Nitrites/history , Nitrites/metabolism , Nitroglycerin/history , Nitroglycerin/metabolism , Signal Transduction/drug effects , Vasodilator Agents/history , Vasodilator Agents/metabolism
2.
Basic Res Cardiol ; 101(2): 180-9, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16450075

ABSTRACT

Post-conditioning (Post-C) induced cardioprotection involves activation of guanylyl-cyclase. In the ischemic preconditioning scenario, the downstream targets of cGMP include mitochondrial ATP-sensitive K(+) (mK(ATP)) channels and protein kinase C (PKC), which involve reactive oxygen species (ROS) production. This study tests the hypothesis that mK(ATP), PKC and ROS are also involved in the Post-C protection. Isolated rat hearts underwent 30 min global ischemia (I) and 120 min reperfusion (R) with or without Post-C (i.e., 5 cycles of 10 s R/I immediately after the 30 min ischemia). In 6 groups (3 with and 3 without Post-C) either mK(ATP) channel blocker, 5- hydroxydecanoate (5-HD), or PKC inhibitor, chelerythrine (CHE) or ROS scavenger, N-acetyl-cysteine (NAC), were given during the entire reperfusion (120 min). In other 6 groups (3 with and 3 without Post-C), 5-HD, CHE or NAC were infused for 117 min only starting after 3 min of reperfusion not to interfere with the early effects of Post-C and/or reperfusion. In an additional group NAC was given during Post-C maneuvers (i.e., 3 min only). Myocardial damage was evaluated using nitro-blue tetrazolium staining and lactate dehydrogenase (LDH) release. Post-C attenuated myocardial infarct size (21 +/- 3% vs. 64 +/- 5% in control; p < 0.01). Such an effect was abolished by 5-HD or CHE given during either the 120 or 117 min of reperfusion as well as by NAC given during the 120 min or the initial 3 min of reperfusion. However, delayed NAC (i.e., 117 min infusion) did not alter the protective effect of Post- C (infarct size 32 +/- 5%; p < 0.01 vs. control, NS vs. Post-C). CHE, 5-HD or NAC given in the absence of Post-C did not alter the effects of I/R. Similar results were obtained in terms of LDH release. Our data show that Post-C induced protection involves an early redox-sensitive mechanism as well as a persistent activation of mK(ATP) and PKC, suggesting that the mK(ATP)/ROS/PKC pathway is involved in post-conditioning.


Subject(s)
Myocardial Ischemia/physiopathology , Myocardial Reperfusion Injury/prevention & control , Potassium Channels/metabolism , Protein Kinase C/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/physiology , Adenosine Triphosphate/metabolism , Animals , Enzyme Activation/drug effects , Enzyme Activation/physiology , Free Radical Scavengers/pharmacology , Heart/drug effects , Heart/physiopathology , Male , Mitochondria/metabolism , Myocardium/pathology , Organ Culture Techniques , Oxidation-Reduction , Potassium Channel Blockers/pharmacology , Protein Kinase Inhibitors/pharmacology , Rats , Signal Transduction/drug effects
3.
Basic Res Cardiol ; 101(2): 168-79, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16132172

ABSTRACT

We aimed to assess the role of the nitric oxide (NO)-cGMP pathway in cardioprotection by brief intermittent ischemias at the onset of reperfusion (i.e., post-conditioning (Post-con)). We also evaluated the role of coronary flow and pressure in Post-con. Rat isolated hearts perfused at constant- flow or -pressure underwent 30 min global ischemia and 120 min reperfusion. Post-con obtained with brief ischemias of different duration (modified, MPost-con) was compared with Post-con obtained with ischemias of identical duration (classical, C-Post-con) and with ischemic preconditioning (IP). Infarct size was evaluated using nitro-blue tetrazolium staining and lactate dehydrogenase (LDH) release. In the groups, NO synthase (NOS) or guanylyl-cyclase (GC) was inhibited with LNAME and ODQ, respectively. In the subgroups, the enzyme immunoassay technique was used to quantify cGMP release. In the constant-flow model, M-Post-con and C-Post-con were equally effective, but more effective than IP in reducing infarct size. The cardioprotection by M-Post-con was only blunted by the NOS-inhibitor, but was abolished by the GC-antagonist. Post-ischemic cGMP release was enhanced by MPost-con. In the constant-pressure model IP, M-Post-con and C-Post-con were equally effective in reducing infarct size. Post-con protocols were more effective in the constant-flow than in the constant-pressure model. In all groups, LDH release during reperfusion was proportional to infarct size. In conclusion, Post-con depends upon GC activation, which can be achieved by NOS-dependent and NOS-independent pathways. The benefits of M- and CPost-con are similar. However, protection by Post-con is greater in the constant-flow than in the constant-pressure model.


Subject(s)
Coronary Circulation , Cyclic GMP/metabolism , Myocardial Infarction/pathology , Myocardial Ischemia/physiopathology , Myocardial Reperfusion Injury/prevention & control , Nitric Oxide/metabolism , Animals , Coronary Circulation/drug effects , Coronary Circulation/physiology , Enzyme Inhibitors/pharmacology , Guanylate Cyclase/drug effects , Guanylate Cyclase/metabolism , Heart/drug effects , Heart/physiopathology , Ischemic Preconditioning, Myocardial , Male , Nitric Oxide Synthase/drug effects , Nitric Oxide Synthase/metabolism , Organ Culture Techniques , Rats , Rats, Wistar , Time Factors
4.
Life Sci ; 77(16): 2004-17, 2005 Sep 02.
Article in English | MEDLINE | ID: mdl-15919095

ABSTRACT

Ischemic preconditioning (IP) triggers cardioprotection via a signaling pathway that converges on mitochondria. The effects of the inhibition of carnitine palmitoyltransferase I (CPT-I), a key enzyme for transport of long chain fatty acids (LCFA) into the mitochondria, on ischemia/reperfusion (I/R) injury are unknown. Here we investigated, in isolated perfused rat hearts, whether sub-chronic CPT-I inhibition (5 days i.p. injection of 25 mg/kg/day of Etomoxir) affects I/R-induced damages and whether cardioprotection by IP can be induced after this inhibition. Effects of global ischemia (30 min) and reperfusion (120 min) were examined in hearts harvested from Control (untreated), Vehicle- or Etomoxir-treated animals. In subsets of hearts from the three treated groups, IP was induced by three cycles of 3 min ischemia followed by 10 min reperfusion prior to I/R. The extent of I/R injury under each condition was assessed by changes in infarct size as well as in myocardial contractility. Postischemic contractility, as indexed by developed pressure and dP/dt(max), was similarly affected by I/R, and was similarly improved with IP in Control, Vehicle or Etomoxir treated animals. Infarct size was also similar in the three subsets without IP, and was significantly reduced by IP regardless of CPT-I inhibition. We conclude that CPT-I inhibition does not affect I/R damages. Our data also show that IP affords myocardial protection in CPT-I inhibited hearts to a degree similar to untreated animals, suggesting that a long-term treatment with the metabolic anti-ischemic agent Etomoxir does not impede the possibility to afford cardioprotection by ischemic preconditioning.


Subject(s)
Carnitine O-Palmitoyltransferase/antagonists & inhibitors , Epoxy Compounds/pharmacology , Ischemic Preconditioning, Myocardial , Myocardial Contraction/physiology , Myocardium/metabolism , Reperfusion Injury/metabolism , Analysis of Variance , Animals , Male , Myocardial Contraction/drug effects , Myocardial Infarction/metabolism , Rats , Rats, Wistar , Ventricular Pressure
5.
Am J Physiol Heart Circ Physiol ; 288(5): H2512-20, 2005 May.
Article in English | MEDLINE | ID: mdl-15637120

ABSTRACT

Ischemic preconditioning (IP) is a cardioprotective mechanism against myocellular death and cardiac dysfunction resulting from reperfusion of the ischemic heart. At present, the precise list of mediators involved in IP and the pathways of their mechanisms of action are not completely known. The aim of the present study was to investigate the role of platelet-activating factor (PAF), a phospholipid mediator that is known to be released by the ischemic-reperfused heart, as a possible endogenous agent involved in IP. Experiments were performed on Langendorff-perfused rat hearts undergoing 30 min of ischemia followed by 2 h of reperfusion. Treatment with a low concentration of PAF (2 x 10(-11) M) before ischemia reduced the extension of infarct size and improved the recovery of left ventricular developed pressure during reperfusion. The cardioprotective effect of PAF was comparable to that observed in hearts in which IP was induced by three brief (3 min) periods of ischemia separated by 5-min reperfusion intervals. The PAF receptor antagonist WEB-2170 (1 x 10(-9) M) abrogated the cardioprotective effect induced by both PAF and IP. The protein kinase C (PKC) inhibitor chelerythrine (5 x 10(-6) M) or the phosphoinositide 3-kinase (PI3K) inhibitor LY-294002 (5 x 10(-5) M) also reduced the cardioprotective effect of PAF. Western blot analysis revealed that following IP treatment or PAF infusion, the phosphorylation of PKC-epsilon and Akt (the downstream target of PI3K) was higher than that in control hearts. The present data indicate that exogenous applications of low quantities of PAF induce a cardioprotective effect through PI3K and PKC activation, similar to that afforded by IP. Moreover, the study suggests that endogenous release of PAF, induced by brief periods of ischemia and reperfusion, may participate to the triggering of the IP of the heart.


Subject(s)
Cardiotonic Agents/pharmacology , Ischemic Preconditioning, Myocardial , Myocardial Reperfusion Injury/drug therapy , Phosphatidylinositol 3-Kinases/metabolism , Platelet Activating Factor/pharmacology , Protein Kinase C/metabolism , Alkaloids , Animals , Azepines/pharmacology , Benzophenanthridines , Chromones/pharmacology , Coronary Circulation , Enzyme Inhibitors/pharmacology , In Vitro Techniques , Male , Morpholines/pharmacology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardium/enzymology , Myocardium/pathology , Necrosis , Perfusion , Phenanthridines/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Rats , Rats, Wistar , Triazoles/pharmacology , Ventricular Function, Left/drug effects
6.
Am J Physiol Heart Circ Physiol ; 287(5): H2192-200, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15217795

ABSTRACT

The amplitude of coronary reactive hyperemia (CRH), elicited by 15 s of ischemia, is reduced in hearts subjected to 5 min of ischemic preconditioning (IP). F0F1 ATP synthase activity and ATP concentration are also altered by IP. We hypothesized that F0F1 ATP synthase is differently modulated by the inhibitor protein IF(1) during CRH elicited before (CRHnp) and after (CRHprec) IP. Hemodynamic parameters were recorded in 10 anesthetized goats. Myocardial biopsies were obtained before IP (Cnp), during CRHnp, 4 and 6 min after the onset of CRHnp, after IP (Cprec), during CRHprec, and 4 min after CRHprec. F0F1 ATP synthase activity, ATP concentration, and ATP-to-ADP ratio (ATP/ADP) were determined. Compared with CRHnp, IP blunted CRHprec. F0F1 ATP synthase activity transiently increased during CRHnp, decreased 4 min after CRHnp, and returned to control 2 min later; it was lower after IP (Cprec) and did not change during and after CRHprec. All these changes in activity were modulated by IF1. During CRHnp, ATP concentration and ATP/ADP were reduced compared with Cnp and began to rise 6 min thereafter. During Cprec, both parameters were transiently reduced but increased during and after CRHprec. Hence, during CRHnp, F0F1 ATP synthase activity transiently increases and then decreases significantly. The short-lasting inhibition of the enzyme may explain why a few seconds of occlusion do not induce IP. After IP, F0F1 ATP synthase activity is blunted, and it is not affected by a subsequent 15 s of occlusion, which induces a blunted CRHprec. These results suggest that postischemic long-lasting inhibition of F0F1 ATP synthase activity may be a feature of the preconditioned heart. The increase in ATP concentration after preconditioning is in agreement with previous reports of reduced ATP hydrolysis by cytoplasmic ATPases.


Subject(s)
Coronary Circulation , Hyperemia/enzymology , Ischemic Preconditioning, Myocardial , Myocardium/enzymology , Proton-Translocating ATPases/metabolism , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , Energy Metabolism , Goats , Hemodynamics , Mitochondria, Heart/metabolism , Myocardium/metabolism , Osmolar Concentration
7.
Ital Heart J ; 4(6): 383-94, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12898803

ABSTRACT

In the coronary circulation, when reperfusion follows ischemia, endothelial dysfunction occurs. This is characterized by a reduced endothelial release of nitric oxide and by an increased release of reactive oxygen species and endothelin. The reduced availability of nitric oxide leads to the adhesion of neutrophils to the vascular endothelium, platelet aggregation and, with the contribution of endothelin, vasoconstriction, which are responsible for the "no-reflow" phenomenon. Neutrophil adhesion is followed by the release of the superoxide anion from neutrophils and endothelial cells. Preconditioning limits the endothelial damage by ischemia-reperfusion. A relevant role is attributed to the increased endothelial release of nitric oxide, while that of adenosine is controversial. Another effect of preconditioning on the coronary vasculature is the acceleration of vasodilation in reactive hyperemia after a brief coronary occlusion. The acceleration is prevented if myocardial protection is achieved by means of the activation of the mitochondrial adenosine triphosphate sensitive potassium channels by diazoxide and persists when ischemic preconditioning is induced after blockade of the same channels by 5-hydroxydecanoate.


Subject(s)
Coronary Vessels/physiopathology , Endothelium, Vascular/physiopathology , Ischemic Preconditioning, Myocardial , Myocardial Ischemia/prevention & control , Myocardial Ischemia/physiopathology , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/physiopathology , Adenosine/metabolism , Adenosine/physiology , Coronary Circulation/physiology , Coronary Vessels/metabolism , Endothelium, Vascular/metabolism , Humans , Nitric Oxide/metabolism , Nitric Oxide/physiology , Reactive Oxygen Species/metabolism
8.
Free Radic Biol Med ; 34(1): 33-43, 2003 Jan 01.
Article in English | MEDLINE | ID: mdl-12498977

ABSTRACT

Nitric oxide (NO) donors mimic the early phase of ischemic preconditioning (IPC). The effects of nitroxyl (HNO/NO(-)), the one-electron reduction product of NO, on ischemia/reperfusion (I/R) injury are unknown. Here we investigated whether HNO/NO(-), produced by decomposition of Angeli's salt (AS; Na(2)N(2)O(3)), has a cardioprotective effect in isolated perfused rat hearts. Effects were examined after intracoronary perfusion (19 min) of either AS (1 microM), the NO donor diethylamine/NO (DEA/NO, 0.5 microM), vehicle (100 nM NaOH) or buffer, followed by global ischemia (30 min) and reperfusion (30 min or 120 min in a subset of hearts). IPC was induced by three cycles of 3 min ischemia followed by 10 min reperfusion prior to I/R. The extent of I/R injury under each intervention was assessed by changes in myocardial contractility as well as lactate dehydrogenase (LDH) release and infarct size. Postischemic contractility, as indexed by developed pressure and dP/dt(max), was similarly improved with IPC and pre-exposure to AS, as opposed to control or DEA/NO-treated hearts. Infarct size and LDH release were also significantly reduced in IPC and AS groups, whereas DEA/NO was less effective in limiting necrosis. Co-infusion in the triggering phase of AS and the nitroxyl scavenger, N-acetyl-L-cysteine (4 mM) completely reversed the beneficial effects of AS, both at 30 and 120 min reperfusion. Our data show that HNO/NO(-) affords myocardial protection to a degree similar to IPC and greater than NO, suggesting that reactive nitrogen oxide species are not only necessary but also sufficient to trigger myocardial protection against reperfusion through species-dependent, pro-oxidative, and/or nitrosative stress-related mechanisms.


Subject(s)
Ischemic Preconditioning, Myocardial , Nitrogen Oxides/metabolism , Sulfhydryl Compounds/metabolism , Animals , Male , Myocardial Reperfusion Injury/metabolism , Rats , Rats, Wistar
9.
Life Sci ; 71(19): 2299-309, 2002 Sep 27.
Article in English | MEDLINE | ID: mdl-12215377

ABSTRACT

Ischemic preconditioning increases the velocity of vasodilatation and reduces the total hyperemic flow (THF) of a subsequent coronary reactive hyperemia (CRH). The increase in the velocity of vasodilatation has been shown to depend on an up-regulation of the endothelial release of nitric oxide, while the reduction of THF is attributed to an adenosine A(1) receptor-mediated mechanism. We investigated whether the changes in CRH induced by preconditioning ischemia (PI) can still be obtained after blockade of mitochondrial ATP-sensitive K(+) channels by sodium 5-hydroxydecanoate (5-HD), and whether the blockade per se affects the pattern of CRH. In anesthetized goats, flow was recorded from the left circumflex coronary artery (LCCA). CRH was obtained with the occlusion of LCCA for 15 s. PI was obtained by 2 cycles of 2.5 min of LCCA occlusion with a 5 min interval of reperfusion between the two occlusions. CRH was studied before and after i.v. administration of 5-HD (20 mg/kg), as well as in the presence of 5-HD after PI. Following 5-HD, the pattern of CRH remained unchanged. After 5-HD and PI, velocity of vasodilatation and total hyperemic flow of CRH showed the same changes as in previous studies after PI alone. It was concluded that the blockade of mitochondrial ATP-sensitive K(+) channels, which is reported to prevent myocardial protection, does not affect CRH and does not prevent PI from increasing the velocity of vasodilatation and reducing THF. These results demonstrate that the changes induced in CRH by preconditioning are independent of the opening of the mitochondrial ATP-sensitive K(+) channels.


Subject(s)
Adenosine Triphosphate/metabolism , Coronary Circulation/physiology , Hyperemia/physiopathology , Ischemic Preconditioning, Myocardial , Mitochondria/metabolism , Potassium Channels/metabolism , Anesthesia , Animals , Anti-Arrhythmia Agents/pharmacology , Coronary Circulation/drug effects , Decanoic Acids/pharmacology , Goats , Hemodynamics/drug effects , Hydroxy Acids/pharmacology , Mitochondria/drug effects , Potassium Channels/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...