Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
2.
J Cardiovasc Pharmacol ; 62(6): 567-75, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23921313

ABSTRACT

Depending on their concentrations, both nitric oxide (NO) and reactive oxygen species (ROS) take part either in myocardial ischemia reperfusion injury or in protection by ischemic and pharmacological preconditioning (Ipre) and postconditioning (Ipost). At the beginning of reperfusion, a transient release of NO is promptly scavenged by ROS to form the highly toxic peroxynitrite, which is responsible for a further increase of ROS through endothelial nitric oxide synthase uncoupling. The protective role of NO has suggested the use of NO donors to mimic Ipre and Ipost. However, NO donors have not always given the expected protection, possibly because they are responsible for the production of different amounts of ROS that depend on the amount of released NO. This review is focused on the role of the balance of NO and ROS in myocardial injury and its prevention by Ipre and Ipost and after the use of NO donors given with or without antioxidant compounds to mimic Ipre and Ipost.


Subject(s)
Ischemic Postconditioning , Ischemic Preconditioning, Myocardial , Myocardial Reperfusion Injury/metabolism , Nitric Oxide/metabolism , Reactive Oxygen Species/metabolism , Animals , Antioxidants/adverse effects , Antioxidants/therapeutic use , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Coronary Vessels/physiopathology , Drug Interactions , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Heart/drug effects , Heart/physiopathology , Humans , Mitochondria, Heart/drug effects , Mitochondria, Heart/enzymology , Mitochondria, Heart/metabolism , Myocardial Reperfusion Injury/physiopathology , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/therapy , Myocardium/enzymology , Myocardium/metabolism , Nitric Oxide/agonists , Nitric Oxide/antagonists & inhibitors , Nitric Oxide Donors/adverse effects , Nitric Oxide Donors/therapeutic use , Reactive Oxygen Species/agonists , Reactive Oxygen Species/antagonists & inhibitors
3.
J Cardiovasc Pharmacol ; 59(3): 241-8, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22030894

ABSTRACT

Low concentrations of a hydrophilic nitric oxide donor (NOD) are reported to reduce myocardial reperfusion injury only when combined with a lipophilic antioxidant (AOX) to form a hybrid molecule (HYB). Here we tested whether liposoluble NOD requires to be combined with AOX to be protective. Isolated rat hearts underwent 30 minutes of ischemia and 120 minutes of reperfusion. To induce postconditioning, 1 µM solutions of the following liposoluble compounds were given during the first 20 minutes of reperfusion: NOD with weak (w-NOD) or strong NO-releasing potency (s-NOD); weak HYB built up with w-NOD and a per se ineffective AOX lead; strong HYB built up with s-NOD and the same AOX; mixtures of w-NOD plus AOX or s-NOD plus AOX. A significant reduction of infarct size with improved recovery of cardiac function was obtained only with weak HYB. We suggest that w-NOD requires the synergy with a per se ineffective AOX to protect. The synergy is possible only if the 2 moieties enter the cell simultaneously as a hybrid, but not as a mixture. It seems that strong HYB was ineffective because an excessive intracellular NO release produces a large amount of reactive species, as shown from the increased nitrotyrosine production.


Subject(s)
Antioxidants/pharmacology , Cardiotonic Agents/pharmacology , Myocardial Reperfusion Injury/prevention & control , Nitric Oxide Donors/pharmacology , Animals , Antioxidants/administration & dosage , Antioxidants/chemistry , Cardiotonic Agents/administration & dosage , Cardiotonic Agents/chemistry , Drug Synergism , Ischemic Postconditioning/methods , Male , Myocardial Infarction/physiopathology , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/physiopathology , Nitric Oxide/metabolism , Nitric Oxide Donors/administration & dosage , Nitric Oxide Donors/chemistry , Rats , Rats, Wistar , Tyrosine/analogs & derivatives , Tyrosine/metabolism
4.
Am J Physiol Heart Circ Physiol ; 300(6): H2308-15, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21378145

ABSTRACT

We studied whether apelin-13 is cardioprotective against ischemia/reperfusion injury if given as either a pre- or postconditioning mimetic and whether the improved postischemic mechanical recovery induced by apelin-13 depends only on the reduced infarct size or also on a recovery of function of the viable myocardium. We also studied whether nitric oxide (NO) is involved in apelin-induced protection and whether the reported ischemia-induced overexpression of the apelin receptor (APJ) plays a role in cardioprotection. Langendorff-perfused rat hearts underwent 30 min of global ischemia and 120 min of reperfusion. Left ventricular pressure was recorded. Infarct size and lactate dehydrogenase release were determined to evaluate the severity of myocardial injury. Apelin-13 was infused at 0.5 µM concentration for 20 min either before ischemia or in early reperfusion, without and with NO synthase inhibition by N(G)-nitro-l-arginine (l-NNA). In additional experiments, before ischemia also 1 µM apelin-13 was tested. APJ protein level was measured before and after ischemia. Whereas before ischemia apelin-13 (0.5 and 1.0 µM) was ineffective, after ischemia it reduced infarct size from 54 ± 2% to 26 ± 4% of risk area (P < 0.001) and limited the postischemic myocardial contracture (P < 0.001). l-NNA alone increased postischemic myocardial contracture. This increase was attenuated by apelin-13, which, however, was unable to reduce infarct size. Ischemia increased APJ protein level after 15-min perfusion, i.e., after most of reperfusion injury has occurred. Apelin-13 protects the heart only if given after ischemia. In this protection NO plays an important role. Apelin-13 efficiency as postconditioning mimetic cannot be explained by the increased APJ level.


Subject(s)
Intercellular Signaling Peptides and Proteins/therapeutic use , Myocardial Infarction/drug therapy , Myocardial Infarction/pathology , Myocardial Ischemia/physiopathology , Recovery of Function/drug effects , Animals , Apelin Receptors , Dose-Response Relationship, Drug , Heart/drug effects , Heart/physiology , Intercellular Signaling Peptides and Proteins/pharmacology , Male , Models, Animal , Myocardial Infarction/physiopathology , Nitric Oxide/physiology , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/physiology , Recovery of Function/physiology , Time Factors , Treatment Outcome
5.
Regul Pept ; 168(1-3): 10-20, 2011 Jun 07.
Article in English | MEDLINE | ID: mdl-21362443

ABSTRACT

This study is the first to report on vascular effect of the chromogranin A derived Vasostatin-I (CgA(1-76)) in vivo. Cardiovascular parameters were recorded in 29 rabbits with sympathetically decentralized right carotid vascular bed. The recombinant human STA CgA(1-78) (VS-1) was infused at 480 µg/kg over 25 min. Group I was kept awake while groups II-V were anesthetized with Ketamine-xylazine. VS-1 was given alone in groups I-II while in presence of either phentolamine, phentolamine plus propranolol or hexamethonium in groups III-V. Serum VS-1 peaked at 2 µg/ml (200 nM) before onset of vascular effects and declined rapidly to ~200 ng/ml within 30 min. In all groups but III and IV VS-1 induced a brief vasoconstriction, being larger in intact than in sympathetically decentralized beds. The VS-1 induced vasoconstriction was not altered by hexamethonium but was abolished by phentolamine. In presence of the α-adrenergic blocker a long lasting vasodilatation, unaffected by propranolol, was apparent on both innervated and decentralized sides. In conclusion, VS-1 induced an α-adrenoceptor-mediated vasoconstriction presumably brought about by noradrenaline release from sympathetic nerves when infused at a dose giving an initial serum concentration of ~200 nM. This initial vasoconstriction masked a persistent adrenoceptor-independent vasodilatation, consistent with previous reports from in vitro models.


Subject(s)
Cardiovascular Physiological Phenomena/drug effects , Chromogranin A/pharmacology , Peptide Fragments/pharmacology , Vasoconstriction/drug effects , Adrenergic alpha-Antagonists/pharmacology , Animals , Chromogranin A/blood , Humans , Male , Peptide Fragments/blood , Propranolol/pharmacology , Rabbits , Recombinant Proteins/blood , Recombinant Proteins/pharmacology , Structure-Activity Relationship
6.
Antioxid Redox Signal ; 14(5): 909-22, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-20615122

ABSTRACT

In the heart, a great part of ischaemia and reperfusion injuries occurs mainly during the first minutes of reperfusion. The opening of the mitochondrial permeability transition pores is the end point of the cascade to myocardial damage. Also, oxidative stress contributes to cell death. Postconditioning is a protective maneuver that can be selectively timed at the beginning of reperfusion. It is hypothesized that it acts via the reperfusion injury salvage kinase pathway, which includes nitric oxide-dependent and nitric oxide-independent cascades. Apelin is an endogenous peptide that can protect the heart from reperfusion injury if given at the beginning of reperfusion but not before ischaemia. It is hypothesized that it may trigger the reperfusion injury salvage kinase pathway via a specific apelin receptor. Apelin can also limit the oxidative stress by the activation of superoxide dismutase. Apelin and apelin receptor expression increase early after ischaemia and at the beginning of an ischaemic heart failure. These observations suggest that the endogenous release of the peptide can limit the severity of an infarction and ameliorate myocardial contractility compromised by the appearance of the failure. Due to its protective activities, apelin could be a therapeutic tool if administered with the same catheter used for angioplasty or after the maneuvers aimed at bypassing a coronary occlusion.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Receptors, G-Protein-Coupled/metabolism , Animals , Cattle , Free Radicals/metabolism , Humans , Ischemia/metabolism , Ischemia/prevention & control , Ischemic Postconditioning/methods , Mice , Myocardial Contraction/drug effects , Oxidative Stress/drug effects , Phosphotransferases/metabolism , Rats , Signal Transduction
7.
J Cell Mol Med ; 12(2): 507-21, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18419594

ABSTRACT

Little is known on the early homing features of transplanted mesenchymal stem cells (MSCs). We used the isolated rat heart model to study the homing of MSCs injected in the ventricular wall of a beating heart. In this model all types of cells and matrix elements with their interactions are represented, while external interferences by endothelial/neutrophil interaction and neurohormonal factors are excluded. We studied the morphology and marker expression of MSCs implanted in normal hearts and in the border-zone of infarcted myocardium. Early morphological adaptation of MSC homing differs between normal and infarcted hearts over the first 6 hrs after transplantation. In normal hearts, MSCs migrate very early through the interstitial milieu and begin to show morphological changes. Yet, in infarcted hearts MSCs remain in the site of injection forming clusters of round-shaped cells in the border-zone of the infarcted area. Both in normal and infarcted hearts, immuno-histochemistry and confocal imaging showed that, besides the proliferative marker proliferating cell nuclear agent (PCNA), some transplanted cells early express myoblastic maker GATA-4, and some of them show a VWF immunopositivity. Moreover, a few hours after injection connexin-43 is well evident between cardiomyocytes and injected cells. This study indicates for the first time that the isolated beating heart is a good model to study early features of MSC homing without external interferences. The results show (i) that MSCs start to change marker expression few hours after injection into a beating heart and (ii) that infarcted myocardium influences transplanted MSC morphology and mobility within the heart.


Subject(s)
Adult Stem Cells/physiology , Heart/physiology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/physiology , Myocardial Infarction/therapy , Adult Stem Cells/cytology , Animals , Biomarkers/metabolism , Cells, Cultured , Green Fluorescent Proteins/metabolism , Immunohistochemistry , In Vitro Techniques , Male , Mesenchymal Stem Cells/cytology , Models, Cardiovascular , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Rats , Rats, Wistar , Time Factors
8.
Am J Physiol Heart Circ Physiol ; 293(1): H719-27, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17416598

ABSTRACT

The acidic protein chromogranin A (CgA) is the precursor of several regulatory peptides generated by specific proteolytic processes. Human recombinant CgA NH(2)-terminal fragment STA-CgA(1-78) (hrSTA-CgA(1-78)), containing vasostatin-1 (CgA(1-76)) domain, exerts a negative inotropic effect and counteracts the beta-adrenergic positive inotropic effect on the rat heart. We hypothesized an involvement of nitric oxide (NO)-dependent pathway in both cardiodepression and cardioprotection by hrSTA-CgA(1-78). We also hypothesized an involvement of adenosine A(1) receptor and protein kinase C (PKC) in cardioprotection by hrSTA-CgA(1-78). Therefore, we evaluated whether 1) the cardioinhibition mediated by hrSTA-CgA(1-78) involves the G(i/o) proteins/NO-dependent signal transduction cascade, 2) hrSTA-CgA(1-78) induces ischemic preconditioning-like protective effects on the myocardium, and 3) inhibition of NO synthase (NOS), adenosine A(1) receptor, or PKC affects hrSTA-CgA(1-78) protection. Using the isolated rat heart, we found that the reduction of left ventricular pressure (LVP), rate-pressure product, and maximal values of the first derivative of LVP elicited by hrSTA-CgA(1-78) at 33 nM is abolished by blocking G(i/o) proteins with pertussis toxin, scavenging NO with hemoglobin, and blocking NOS activity with N(G)-monomethyl-l-arginine or N(5)-(iminoethyl)-l-ornithine, soluble guanylate cyclase with 1H-[1,2,4]oxadiazole-[4,4-a]quinoxalin-1-one, and protein kinase (PKG) with KT5823. Data suggest the involvement of the G(i/o) proteins/NO-cGMP-PKG pathway in the hrSTA-CgA(1-78)-dependent cardioinhibition. When given before 30 min of ischemia, hrSTA-CgA(1-78) significantly reduced the size of the infarct from 64 +/- 4 to 32 +/- 3% of the left ventricular mass. This protective effect was abolished by either NOS inhibition or PKC blockade and was attenuated, but not suppressed, by the blockade of A(1) receptors. These results suggest that hrSTA-CgA(1-78) activity triggers two different pathways: one of these pathways is mediated by A(1) receptors, and the other is mediated by NO release. As with repeated brief preconditioning ischemia, hrSTA-CgA(1-78) may be considered a stimulus strong enough to trigger both pathways, which may converge on PKC.


Subject(s)
Adenosine/metabolism , Chromogranin A/administration & dosage , Ischemic Preconditioning/methods , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/physiopathology , Nitric Oxide/metabolism , Peptide Fragments/administration & dosage , Signal Transduction/drug effects , Animals , Chromogranin A/genetics , Humans , In Vitro Techniques , Male , Peptide Fragments/genetics , Rats , Rats, Wistar , Recombinant Proteins/administration & dosage
9.
Cardiovasc Res ; 75(1): 168-77, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17400201

ABSTRACT

OBJECTIVE: Postconditioning (PostC) maneuvers allow post-ischemic accumulation of autacoids, which trigger protection. We tested if PostC-triggering includes bradykinin (BK) B2 receptor activation and its downstream pathway. METHODS AND RESULTS: Isolated rat hearts underwent 30 min ischemia and 120 min reperfusion. Infarct size was evaluated using nitro-blue tetrazolium staining. In Control hearts infarct size was 61+/-5% of risk area. PostC (5 cycles of 10 s reperfusion/ischemia) reduced infarct size to 22+/-4% (p<0.01). PostC protection was abolished by B2 BK receptor-antagonists (HOE140 or WIN64338), nitric oxide synthase-inhibitor (L-nitro-arginine-methylester), protein kinase G (PKG)-blocker (8-bromoguanosine-3',5'-cyclic-monophosphorothioate), and mitochondrial K(ATP) (mK(ATP))-blocker (5-hydroxydecanoate) each given for 3 min only. Since 3 min of BK-infusion (100 nM) did not reproduce PostC protection, protocols with Intermittent-BK infusion were used to mimic PostC: a) 5 cycles of 10 s oxygenated-no-BK/oxygenated+BK buffer; b) 5 cycles of 10 s oxygenated-no-BK/hypoxic+BK buffer. Both protocols with Intermittent-BK attenuated infarct size (36+/-5% and 38+/-4%, respectively; p<0.05 vs Control and NS vs PostC for both; NS vs each other). Intermittent-BK protection was abolished by the same antagonists used to prevent PostC protection. Intermittence of re-oxygenation only (5 cycles of 10 s oxygenated/hypoxic buffer) did not reproduce PostC. Yet, cardioprotection was triggered by intermittent mK(ATP) activation with diazoxide, but not by intermittent reactive oxygen species (ROS) generation with purine/xanthine oxidase. ROS scavengers (N-acetyl-L-cysteine or 2-mercaptopropionylglycine), given for 3 min only, abolished PostC-, Intermittent BK-and diazoxide-induced protection. CONCLUSIONS: Intermittent targeting of specific cellular sites (i.e. BK B2 receptors and mK(ATP) channels) during early reperfusion triggers PostC protection via ROS signaling. Since neither intermittent oxygenation nor exogenous ROS generators can trigger protection, it is likely that intermittent autacoid accumulation and ROS compartmentalization may play a pivotal role in PostC-triggering.


Subject(s)
Mitochondria, Heart/metabolism , Myocardial Infarction/prevention & control , Potassium Channels/metabolism , Receptor, Bradykinin B2/metabolism , Signal Transduction/physiology , Acetylcysteine/pharmacology , Adenosine Triphosphate/metabolism , Animals , Bradykinin/analogs & derivatives , Bradykinin/metabolism , Bradykinin/pharmacology , Bradykinin B2 Receptor Antagonists , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinases/metabolism , Decanoic Acids/pharmacology , Hydroxy Acids/pharmacology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Oxidation-Reduction , Perfusion , Potassium Channel Blockers/pharmacology , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
10.
J Cell Biochem ; 100(1): 86-99, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-16888800

ABSTRACT

In order to assess, in a controlled in vitro model, the differentiation potential of adult bone marrow derived stem cells we have developed a coculture procedure using adult rat cardiomyocytes and mesenchymal stem cells (MSCs) from transgenic GFP positive rats. We investigated in the cocultured MSCs the time course of cellular processes that are difficult to monitor in in vivo experiments. Adult rat cardiomyocytes and adult rat MSCs were cocultured for up to 7 days and analyzed by confocal microscopy. Several markers were studied by immunofluorescence technique. The fluorescent ST-BODIPY-Dihydropyridine was used to label calcium channels in living cells. Intracellular calcium was monitored with the fluorescent probe X-Rhod-1. Immunofluorescence experiments showed the presence of connexin-43 between cardiomyocytes and MSCs and between MSCs, while no sarcomeric structures were observed at any time of the coculture. We looked at the expression of calcium channels and development of voltage-dependent calcium signaling in cocultured MSCs. MSCs showed a time-dependent increase of labeling of ST-BODIPY-Dihydropyridine, reaching a relatively strong level after 72 h of coculture. The treatment with a non-fluorescent DHP, Nifedipine, completely abolished ST-BODIPY labeling. We investigated whether depolarization could modulate intracellular calcium. Depolarization-induced calcium transients increased in MSCs in relation to the coculture time. We conclude that MSCs cocultured with adult cardiomyocytes present preliminary evidence of voltage-dependent calcium modulation uncoupled with the development of nascent or adult myofibrils, thus showing a limited lineage specification and a low plasticity to differentiate in a full cardiomyocyte-like phenotype.


Subject(s)
Mesenchymal Stem Cells/physiology , Myocytes, Cardiac/physiology , Actinin/metabolism , Animals , Animals, Genetically Modified , Boron Compounds , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels/physiology , Cell Differentiation , Cells, Cultured , Coculture Techniques , Connexin 43/metabolism , Dihydropyridines , Fluorescent Dyes , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Ion Channel Gating , Mesenchymal Stem Cells/cytology , Myocytes, Cardiac/cytology , Myosins/metabolism , Nifedipine/pharmacology , Rats , Sarcomeres/metabolism
11.
G Ital Cardiol (Rome) ; 7(7): 464-73, 2006 Jul.
Article in Italian | MEDLINE | ID: mdl-16977786

ABSTRACT

Post-ischemic reperfusion worsens myocardial injury. Ischemic preconditioning limits the damage by ischemia and reperfusion. Both adenosine and nitric oxide (NO) pathways are involved in protection. Preconditioning, however, is of little, if any, practical use as the onset of an infarction is usually unpredictable. Recently, it has been shown that the heart can be protected against the extension of reperfusion injuries if 3 or 4 brief (10-30 s) coronary occlusions are performed just at the beginning of the reperfusion. This procedure has been called post-conditioning. Post-conditioning reduces the oxidant-induced injury; moreover, it attenuates the local inflammatory response to reperfusion. Post-conditioning also activates triggers, signaling pathways and effectors implicated in other cardioprotective maneuvers, such as ischemic and pharmacological preconditioning. Post-conditioning seems to trigger the up-regulation of survival kinases principally known to attenuate the pathogenesis of apoptosis and possibly necrosis. As regards the possibility of pharmacological post-conditioning, several agents have been tested. We are testing NO donor(s), which can reduce infarct size in the rat in the absence of post-conditioning. Since during reperfusion there is a large production of reactive oxygen species, also the effect of administration of an antioxidant compound during reperfusion was studied. In the rat, such a procedure reduced the infarct size to a greater extent than post-conditioning. Moreover, an additive effect of NO donors and antioxidant compounds is possible.


Subject(s)
Antioxidants/pharmacology , Myocardial Infarction/therapy , Myocardial Reperfusion Injury/prevention & control , Nitric Oxide Donors/pharmacology , Adenosine/metabolism , Animals , Antioxidants/therapeutic use , Coronary Circulation , Drug Synergism , Ischemic Preconditioning, Myocardial/methods , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/physiopathology , Nitric Oxide/metabolism , Nitric Oxide Donors/therapeutic use , Phosphotransferases/metabolism , Rats , Time Factors
12.
J Cardiovasc Med (Hagerstown) ; 7(9): 645-52, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16932076

ABSTRACT

Endothelins (ETs) exert a persistent constrictor effect on the vessels via an increase in intracellular Ca2+ concentration due to the activation of Na+/H+ and Na+/Ca2+ exchangers of the vascular smooth muscle fibres. They also produce a transient dilator effect via the activation of endothelial nitric oxide synthase mediated by protein kinase B/Akt. ETA and ETB2 receptors are involved in vasoconstriction, whereas transient vasodilatation depends on the activation of ETB1 receptors. Depending on animal species and experimental conditions, ETs can also play a role in cardiac muscle contraction and induce either an increase or a decrease in contractility. It is likely that only ETA, and not ETB, receptors are involved in the ET-induced increase in myocardial contractility. As in the case of vasoconstriction, this inotropic effect depends on an increase in intracellular Ca2+ concentration induced by Na+/H+ and Na+/Ca2+ exchangers. Activation of the Na+/H+ exchanger is stimulated by protein kinase C, which is activated by diacylglycerol released in response to ET activity. It has also been proposed that the positive inotropic effect can occur without the contribution of the Na+/Ca2+ exchanger, if the cell alkalinisation produced by the Na/H exchanger improves myofibrillar Ca2+ sensitivity. A reduction in contractility has been attributed to the involvement of the Gi protein/protein kinase G pathway or to the activation of protein kinase C without an increase in intracellular Ca2+ concentration or in myofibrillar Ca2+ sensitivity. The chronic effect of ETs on the myocardium results in hypertrophy and prevention of apoptosis, two processes that are together responsible for the contradictory effect of ETs in heart failure.


Subject(s)
Endothelins/physiology , Endothelium, Vascular/physiology , Muscle, Smooth, Vascular/physiology , Animals , Cells, Cultured , Heart/physiology , Humans , Muscle Cells/physiology , Muscle Contraction/physiology , Nitric Oxide Synthase/physiology , Receptors, Endothelin/physiology , Sodium-Hydrogen Exchangers/physiology , Vasoconstriction/physiology
13.
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
14.
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
15.
Ital Heart J Suppl ; 6(5): 272-8, 2005 May.
Article in Italian | MEDLINE | ID: mdl-15934423

ABSTRACT

Apelin, an endogenous peptide, is the ligand of APJ receptors. Although initially it was identified in the gastrointestinal tract, later its presence was found in several organs and tissues. On the cardiovascular system apelin induces an increase in myocardial contractility and a reduction of vasomotor tone. While the increase in contractility seems to depend on an activation of Na+/H+ and Na+/Ca2+ exchangers, vasodilation is attributed to a release of nitric oxide from the vascular endothelial cells. Apelin-induced vasodilation leads to a reduction of mean filling pressure which in turn causes a decrease of afterload and preload. When apelin is given acutely, the decrease in preload favors the reduction of stroke volume and cardiac output in spite of an increased contractility. On the contrary, when the peptide is administered for 2 weeks, cardiac output increases significantly without the occurrence of cardiac hypertrophy. It is not excluded that hypertrophy might occur after a longer administration.


Subject(s)
Carrier Proteins/pharmacology , Myocardial Contraction/drug effects , Receptors, G-Protein-Coupled , Vasomotor System/drug effects , Animals , Apelin , Blood Pressure/drug effects , Cardiac Output/drug effects , Carrier Proteins/administration & dosage , Carrier Proteins/metabolism , Dose-Response Relationship, Drug , Endothelium, Vascular/physiology , Heart Rate/drug effects , Humans , Intercellular Signaling Peptides and Proteins , Ligands , Mice , Nitric Oxide/metabolism , Rats , Receptors, Cell Surface/metabolism , Research , Time Factors , Vasoconstriction/drug effects , Vasodilation/drug effects
16.
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
18.
Biochim Biophys Acta ; 1659(1): 52-62, 2004 Nov 04.
Article in English | MEDLINE | ID: mdl-15511527

ABSTRACT

A method has been developed to allow the level of F(0)F(1)ATP synthase capacity and the quantity of IF(1) bound to this enzyme be measured in single biopsy samples of goat heart. ATP synthase capacity was determined from the maximal mitochondrial ATP hydrolysis rate and IF(1) content was determined by detergent extraction followed by blue native gel electrophoresis, two-dimensional SDS-PAGE and immunoblotting with anti-IF(1) antibodies. Anaesthetized open-chest goats were subjected to ischemic preconditioning and/or sudden increases of coronary blood flow (CBF) (reactive hyperemia). When hyperemia was induced before ischemic preconditioning, a steep increase in synthase capacity, followed by a deep decrease, was observed. In contrast, hyperemia did not affect synthase capacity when applied after ischemic preconditioning. Similar effects could be produced in vitro by treatment of heart biopsy samples with anoxia (down-regulation of the ATP synthase) or high-salt or high-pH buffers (up-regulation). We show that both in vitro and in vivo the same close inverse correlation exists between enzyme activity and IF(1) content, demonstrating that under all conditions tested the only significant modulator of the enzyme activity was IF(1). In addition, both in vivo and in vitro, 1.3-1.4 mol of IF(1) was predicted to fully inactivate 1 mol of synthase, thus excluding the existence of significant numbers of non-inhibitory binding sites for IF(1) in the F(0) sector.


Subject(s)
Mitochondria, Heart/metabolism , Myocardial Ischemia/metabolism , Myocardium/metabolism , Proteins/pharmacology , Proton-Translocating ATPases/antagonists & inhibitors , Proton-Translocating ATPases/metabolism , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Female , Goats , Homeostasis , In Vitro Techniques , Male , Mitochondria, Heart/drug effects , ATPase Inhibitory Protein
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...