Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 118
Filtrar
1.
Nat Med ; 7(1): 108-13, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11135624

RESUMEN

Diabetic patients frequently suffer from retinopathy, nephropathy, neuropathy and accelerated atherosclerosis. The loss of endothelial function precedes these vascular alterations. Here we report that activation of poly(ADP-ribose) polymerase (PARP) is an important factor in the pathogenesis of endothelial dysfunction in diabetes. Destruction of islet cells with streptozotocin in mice induced hyperglycemia, intravascular oxidant production, DNA strand breakage, PARP activation and a selective loss of endothelium-dependent vasodilation. Treatment with a novel potent PARP inhibitor, starting after the time of islet destruction, maintained normal vascular responsiveness, despite the persistence of severe hyperglycemia. Endothelial cells incubated in high glucose exhibited production of reactive nitrogen and oxygen species, consequent single-strand DNA breakage, PARP activation and associated metabolic and functional impairment. Basal and high-glucose-induced nuclear factor-kappaB activation were suppressed in the PARP-deficient cells. Our results indicate that PARP may be a novel drug target for the therapy of diabetic endothelial dysfunction.


Asunto(s)
Diabetes Mellitus Experimental/fisiopatología , Endotelio Vascular/fisiopatología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Animales , Diabetes Mellitus Experimental/enzimología , Endotelio Vascular/enzimología , Activación Enzimática , Glucosa/farmacología , Inmunohistoquímica , Técnicas In Vitro , Ratones , FN-kappa B/metabolismo
2.
J Exp Med ; 186(7): 1041-9, 1997 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-9314553

RESUMEN

A cytotoxic cycle triggered by DNA single-strand breakage and poly (ADP-ribose) synthetase activation has been shown to contribute to the cellular injury during various forms of oxidant stress in vitro. The aim of this study was to investigate the role of poly (ADP-ribose) synthetase (PARS) in the process of neutrophil recruitment and in development of local and systemic inflammation. In pharmacological studies, PARS was inhibited by 3-aminobenzamide (10-20 mg/kg) in rats and mice. In other sets of studies, inflammatory responses in PARS-/- mice were compared with the responses in corresponding wild-type controls. Inhibition of PARS reduced neutrophil recruitment and reduced the extent of edema in zymosan- and carrageenan-triggered models of local inflammation. Moreover, inhibition of PARS prevented neutrophil recruitment, and reduced organ injury in rodent models of inflammation and multiple organ failure elicited by intraperitoneal injection of zymosan. Inhibition of PARS also reduced the extent of neutrophil emigration across murine mesenteric postcapillary venules. This reduction was due to an increased rate of adherent neutrophil detachment from the endothelium, promoting their reentry into the circulation. Taken together, our results demonstrate that PARS inhibition reduces local and systemic inflammation. Part of the antiinflammatory effects of PARS inhibition is due to reduced neutrophil recruitment, which may be related to maintained endothelial integrity.


Asunto(s)
Inflamación/enzimología , Neutrófilos/fisiología , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Animales , Benzamidas/farmacología , Carragenina/farmacología , Movimiento Celular/efectos de los fármacos , Edema , Inhibidores Enzimáticos/farmacología , Histocitoquímica , Inflamación/inmunología , Pulmón/patología , Masculino , Ratones , Ratones Noqueados , Insuficiencia Multiorgánica/enzimología , Insuficiencia Multiorgánica/inmunología , Peritonitis/enzimología , Peritonitis/inmunología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Ratas , Ratas Wistar , Zimosan/farmacología
3.
J Clin Invest ; 100(3): 723-35, 1997 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-9239421

RESUMEN

DNA single strand breakage and activation of the nuclear enzyme poly (ADP-ribose) synthetase (PARS) contribute to peroxynitrite-induced cellular injury. We investigated the role of PARS activation in the pathogenesis of endothelial dysfunction. In human umbilical vein endothelial cells (HUVEC), DNA strand breakage (alkaline unwinding assay), PARS activation (incorporation or radiolabeled NAD+ into proteins), mitochondrial respiration [conversion of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide to formazan] and apoptotic index (cytoplasmatic release of histones) were measured. Endotoxin shock was induced in rats by bacterial lipopolysaccharide. Vascular reactivity of thoracic aortic rings were measured in organ chambers. In HUVEC, peroxynitrite caused a dose-dependent suppression of mitochondrial respiration, induced DNA strand breakage and caused an activation of PARS. Pharmacological inhibition of PARS reduced the acute and delayed suppression of mitochondrial respiration when cells were exposed to intermediate, but not high doses of peroxynitrite. Similarly, protection against the intermediate, but not high doses of peroxynitrite was seen in fibroblasts from the PARS-/- mice, when compared to wild-type controls. These data suggest that PARS plays a role in peroxynitrite-induced cytotoxicity, but at very high levels of oxidant exposure, PARS-independent cytotoxic mechanisms become predominant. Peroxynitrite-induced apoptosis was not affected by PARS inhibition. Vascular rings exposed to peroxynitrite and rings taken from rats subjected to endotoxic shock exhibited reduced endothelium-dependent relaxant responses in response to acetylcholine. The development of this endothelial dysfunction was ameliorated by the PARS inhibitor 3-aminobenzamide. Activation of PARS by peroxynitrite, therefore, may be involved in the development of endothelial dysfunction in endotoxemia.


Asunto(s)
Endotelio Vascular/fisiopatología , Nitratos/farmacología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Choque Séptico/fisiopatología , Animales , Células Cultivadas , Endotelio Vascular/metabolismo , Activación Enzimática/efectos de los fármacos , Humanos , Lipopolisacáridos , Ratones , Ratas , Choque Séptico/metabolismo
4.
Cardiovasc Res ; 36(2): 205-15, 1997 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9463632

RESUMEN

OBJECTIVE: Peroxynitrite and hydroxyl radical, reactive oxidants produced during reperfusion, are potent triggers of DNA single strand breakage. DNA injury triggers the activation of the nuclear enzyme poly (ADP-ribose) synthetase (PARS), which contributes to cellular energetic depletion. Using 3-aminobenzamide, an inhibitor of PARS, we investigated the role of PARS in the pathogenesis of myocardial reperfusion injury in a rat model. METHODS AND RESULTS: Occlusion of the left main coronary artery (one hour) followed by reperfusion (one hour) in the anesthetized rat caused severe cardiac necrosis, neutrophil infiltration, and increased plasma creatine phosphokinase activity. There was significant peroxynitrite production during reperfusion, as indicated by a massive increase in nitrotyrosine in the necrotic myocardium. Reperfusion was also associated with a significant loss of myocardial ATP. In vivo administration of the PARS inhibitor 3-aminobenzamide (10 mg/kg i.v.) to rats subjected to myocardial ischemia and reperfusion, reduced myocardial infarct size and blunted the increase in plasma creatine phosphokinase activity and myeloperoxidase activity in infarcted hearts. In addition, 3-aminobenzamide partially preserved the myocardial ATP levels. In vitro, pharmacological inhibition of PARS also ameliorated peroxynitrite-induced cytotoxicity in rat cardiac myocytes and human endothelial cells. CONCLUSION: 3-aminobenzamide has significant protective effects in myocardial reperfusion injury. We hypothesize that activation of PARS activation plays a role in the pathophysiology of acute myocardial infarction.


Asunto(s)
Benzamidas/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Daño por Reperfusión Miocárdica/prevención & control , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Adenosina Trifosfato/metabolismo , Animales , Células Cultivadas , Creatina Quinasa/sangre , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/patología , Técnicas In Vitro , Masculino , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocardio/enzimología , Miocardio/metabolismo , Miocardio/patología , Peroxidasa/metabolismo , Ratas , Ratas Wistar , Tirosina/análogos & derivados , Tirosina/metabolismo
5.
FEBS Lett ; 372(2-3): 229-32, 1995 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-7556674

RESUMEN

To quantify peroxynitrite production during shock, we measured oxidation of dihydrorhodamine 123 in rats. In endotoxic and hemorrhagic shock and splanchic ischemia-reperfusion, dihydrorhodamine oxidation rapidly increased, which was prevented by inhibition of endothelial nitric oxide (.NO) synthase (ecNOS). Thus, peroxynitrite is already formed at early stages of shock from ecNOS-derived .NO. Overproduction of .NO by the inducible NOS at late shock was not associated with additional increases in dihydrorhodamine oxidation. ecNOS inhibition enhanced dihydrorhodamine oxidation in control rats. These latter findings may be explained by .NO-mediated inhibition of peroxynitrite-induced dihydrorhodamine oxidation, a phenomenon also observed in vitro.


Asunto(s)
Nitratos/farmacología , Daño por Reperfusión/metabolismo , Rodaminas/metabolismo , Choque Hemorrágico/metabolismo , Choque Séptico/metabolismo , Animales , Masculino , Oxidación-Reducción , Ratas , Ratas Wistar
6.
FEBS Lett ; 363(3): 235-8, 1995 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-7537701

RESUMEN

The free radicals nitric oxide (.NO) and superoxide (O2-) are known to react to form peroxynitrite (ONOO-), a highly reactive species. Peroxynitrite has been suggested to play an important role in the cellular damage associated with the overproduction of .NO, but there are very limited data regarding its in vivo formation. Here we demonstrate that injection of endotoxin into rats leads to the expression of an inducible isoform of .NO synthase (iNOS) in the thoracic aorta at 6 h and an increase in the circulating levels of nitrite/nitrate. Moreover, at the same time point, there is a marked increase in the immunoreactivity of nitrotyrosine, a marker of peroxynitrite in the aorta. The formation of nitrotyrosine was prevented by inhibiting the activity of NOS by NG-methyl-L-arginine in vivo. Our data suggest that during endotoxin shock, part of .NO, produced following the induction of iNOS, is converted into peroxynitrite in the vicinity of large blood vessels. The demonstration of the in vivo formation of peroxynitrite at sites of .NO overproduction may necessitate the development of novel and additional approaches for limiting or preventing .NO-related cytotoxic or vasodilatory actions during circulatory shock.


Asunto(s)
Aminoácido Oxidorreductasas/metabolismo , Aorta/metabolismo , Endotoxinas/farmacología , Lipopolisacáridos/farmacología , Nitratos/metabolismo , Animales , Calcio/metabolismo , Técnica del Anticuerpo Fluorescente , Masculino , Óxido Nítrico Sintasa , Ratas , Ratas Wistar , Tirosina/análogos & derivados , Tirosina/química
7.
FEBS Lett ; 409(2): 147-50, 1997 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-9202135

RESUMEN

Here we investigated the role of endogenous nitric oxide (NO) and peroxynitrite in the process of protein oxidation (as measured by the detection of 2,4-dinitrophenylhydrazine-reactive carbonyls) in immunostimulated macrophages. Immunostimulation of the macrophages by bacterial lipopolysaccharide and gamma-interferon (LPS/IFNgamma) resulted in a marked increase in the oxidation of a large number of mitochondrial and nuclear proteins. The inhibitor of NO synthase, N(G)-methyl-L-arginine (3 mM), and the cell-permeable superoxide dismutase mimetic Mn(III)tetrakis(4-benzoic acid)porphyrin (300 microM) both reduced the extent of protein oxidation in response to LPS/IFNgamma. These results support the view that endogenously produced peroxynitrite induces protein oxidation in the mitochondria and nucleus of immunostimulated macrophages.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Macrófagos/inmunología , Macrófagos/metabolismo , Mitocondrias/metabolismo , Nitratos/fisiología , Proteínas Nucleares/metabolismo , Animales , Línea Celular , Interferón gamma/farmacología , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Ratones , Mitocondrias/efectos de los fármacos , NG-Nitroarginina Metil Éster/farmacología , Nitratos/metabolismo , Óxido Nítrico Sintasa/antagonistas & inhibidores , Proteínas Nucleares/efectos de los fármacos , Oxidación-Reducción/efectos de los fármacos , Fracciones Subcelulares/inmunología , Fracciones Subcelulares/metabolismo , Superóxido Dismutasa/antagonistas & inhibidores
8.
Free Radic Biol Med ; 25(8): 914-25, 1998 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-9840736

RESUMEN

Hydroxyguanidines (OHGs), including the endogenously formed NG-hydroxy-L-arginine (OH-arg), can react with nitric oxide (NO) and nitrogen oxides (NOx) in vitro. Therefore, we have tested OHGs and related compounds for their ability to scavenge peroxynitrite and to protect against peroxynitrite-induced oxidative processes in cells. Hydroxyguanidine, NG-hydroxy-L-arginine and other N-substituted OHGs, dose-dependently inhibited the in vitro oxidation of dihydrorhodamine (DHR) by peroxynitrite (PN), with similar or better efficacy than glutathione or cysteine. Amidoximes, aminoguanidines and O-substituted OHGs were less effective, and guanidines were without effect. In contrast to their effects on DHR oxidation, OHGs exerted only minimal inhibitory effects on the hydroxylation of benzoate by PN, suggesting that OHGs do not react with the activated isomer of peroxynitrous acid. Selected compounds were tested for protection against PN-induced suppression of mitochondrial respiration and protein oxidation in cultured J774 murine macrophages. Aminoguanidines afforded some protection against the effects of PN, but substituted-phenyl OHGs were considerably more effective. Analysis of the products of the reaction of 4-methoxybenzyl-OHG with PN showed rapid formation of nitrosated derivatives, as well as 4-methoxybenzylcyanamide and a small amount of 4-methoxybenzylurea. Nitric oxide and nitrous oxide were also evolved, but indirectly, arising from the decomposition of one of the nitrosation products. The current results demonstrate that hydroxyguanidines react with PN to protect cells against PN-mediated injury and may be more effective than the endogenous antioxidants cysteine and glutathione.


Asunto(s)
Depuradores de Radicales Libres , Guanidinas/farmacología , Nitratos/farmacología , Arginina/análogos & derivados , Arginina/farmacología , Ácido Benzoico/química , Línea Celular , Guanidinas/química , Hidroxilaminas , Hidroxilación , Macrófagos/efectos de los fármacos , Nitratos/química , Oxidación-Reducción , Rodaminas/química
9.
FEBS Lett ; 381(1-2): 82-6, 1996 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-8641445

RESUMEN

Here we report that the cell-permeable superoxide dismutase mimetic Mn(III)tetrakis (4-benzoic acid) porphyrin (MnTBAP) inhibits the oxidation of dihydrorhodamine-123 by peroxynitrite, but does not scavenge nitric oxide (NO). MnTBAP protects against the suppression of mitochondrial respiration in J774 cells exposed to peroxynitrite or to NO donors. MnTBAP and N(G)-methyl-L-arginine provide additive protective effect against the suppression of respiration in immunostimulated cells. Our data suggest separate contributions of NO and peroxynitrite to the suppression of mitochondrial respiration and support the role of oxidative stress in the expression of the inducible isoform of NO synthase.


Asunto(s)
Depuradores de Radicales Libres/farmacología , Macrófagos/metabolismo , Manganeso/farmacología , Metaloporfirinas/farmacología , Mitocondrias/metabolismo , Músculo Liso Vascular/fisiología , Nitratos/farmacología , Óxido Nítrico/farmacología , Consumo de Oxígeno/efectos de los fármacos , Superóxido Dismutasa , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/fisiología , Arginina/análogos & derivados , Arginina/farmacología , Línea Celular , Dietilaminas/farmacología , Escherichia coli , Técnicas In Vitro , Cinética , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Oxidación-Reducción , Penicilamina/análogos & derivados , Penicilamina/farmacología , Conejos , S-Nitroso-N-Acetilpenicilamina , Vasodilatación , omega-N-Metilarginina
10.
Free Radic Biol Med ; 24(3): 450-9, 1998 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9438558

RESUMEN

In vitro studies have demonstrated that mercaptoethylguanidine (MEG), a selective inhibitor of the inducible NO synthase (iNOS), is also effective as a scavenger of peroxynitrite (a potent cytotoxic oxidant produced by the reaction of NO and superoxide). In the present study, we evaluated the antiinflammatory potential of MEG treatment in two models of acute inflammation (carrageenan-induced paw edema and pleurisy), where oxyradicals, NO, and peroxynitrite play a crucial role in the inflammatory process. Our data show that MEG (given at 25 microg/paw in the paw edema model or 10 mg/kg in the pleurisy model) inhibits the inflammatory response (paw swelling, pleural exudate formation, mononuclear cell infiltration, histological injury) in both models. Furthermore, MEG reduced nitrite/nitrate concentrations in the exudate and reduced the activity of the inducible isoform of NO synthase in the lung ex vivo. MEG also reduced the appearance of nitrotyrosine immunoreactivity in the inflamed tissues. Taken together, the present results demonstrate that MEG exerts potent antiinflammatory effects. Part of these antiinflammatory effects may be related to an inhibition of the expression/activity of the inducible NO synthase, another part may be related to oxyradical and peroxynitrite scavenging.


Asunto(s)
Inhibidores Enzimáticos/uso terapéutico , Depuradores de Radicales Libres , Guanidinas/uso terapéutico , Inflamación/tratamiento farmacológico , Nitratos/metabolismo , Óxido Nítrico Sintasa/antagonistas & inhibidores , Animales , Carragenina , Modelos Animales de Enfermedad , Edema/inducido químicamente , Edema/tratamiento farmacológico , Edema/patología , Guanidinas/administración & dosificación , Inflamación/inducido químicamente , Pulmón/patología , Masculino , Neutrófilos/patología , Óxido Nítrico Sintasa de Tipo II , Nitritos/metabolismo , Oxidantes/metabolismo , Pleuresia/inducido químicamente , Pleuresia/tratamiento farmacológico , Pleuresia/patología , Ratas , Ratas Sprague-Dawley
11.
J Neuroimmunol ; 88(1-2): 57-61, 1998 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-9688324

RESUMEN

Stimulation of beta-adrenoceptors has been shown to regulate the production of various inflammatory mediators. In the present study, we investigated in mice whether ligation of beta-adrenoceptors, modulates lipopolysaccharide (LPS)-induced plasma levels of interleukin (IL)-12, interferon-gamma (IFN-gamma), and IL-10. In BALB/c mice, isoproterenol (1-10 mg kg(-1) , i.p.), a selective agonist of beta-adrenoceptors and also dexamethasone (10 mg kg(-1), i.p.) pretreatment 30 min before the administration of LPS suppressed plasma IL-12 (p40 and p70) concentrations as determined at various time points after the LPS challenge. The inhibition of IL-12 release by isoproterenol was prevented by the beta-adrenoceptor antagonist propranolol confirming the involvement of beta-adrenoceptors in the effect of isoproterenol. Furthermore, pretreatment of the animals with propranolol alone enhanced LPS-induced plasma IL-12, suggesting that endogenous catecholamines inhibit IL-12 production via the beta-adrenoceptors. In IL-10 deficient C57BL/6 IL-10(-/-) mice, plasma levels of IL-12 and IFN-gamma were significantly higher than in their counterparts, with more than 70-fold increase in IL-12. Furthermore, while augmenting the IL-10 response in C57BL/6 IL-10(+/+), isoproterenol inhibited the production of IL-12 in both the C57BL/6 IL-10(+/+) and C57BL/6 IL-10(-/-) mice, suggesting that the inhibition of IL-12 production by this compound is independent of the increased release of IL-10. Our results demonstrate, for the first time, that stimulation of beta-adrenoceptors by isoproterenol or endogenous catecholamines suppresses IL-12 production in vivo.


Asunto(s)
Endotoxinas/farmacología , Interleucina-10/deficiencia , Interleucina-12/biosíntesis , Receptores Adrenérgicos beta/fisiología , Agonistas Adrenérgicos beta/farmacología , Animales , Interleucina-12/sangre , Isoproterenol/farmacología , Masculino , Ratones , Ratones Endogámicos BALB C , Valores de Referencia
12.
J Histochem Cytochem ; 46(7): 787-91, 1998 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9632737

RESUMEN

Here we examined the changes in NADPH diaphorase (NADPHd) and inducible nitric oxide synthetase (iNOS) positivity in the medulla of the mouse thymus in response to treatment with the superantigen, Staphylococcal enterotoxin B (SEB). A few NADPHd+ and iNOS+ cells scattered in the medulla were detected in the thymi of naive mice. SEB induced the appearance of a large number of NADPHd+- and iNOS-immunoreactive cells in the thymic medulla. In the thymus of iNOS-deficient mice, a total absence of these NADPHd+ and iNOS+ medullary cells was found both under basal conditions and after SEB stimulation. With the NADPHd reaction, only endothelial staining was detected in the thymi of iNOS-deficient mice. Our data indicate that NADPHd+ cells in the thymic medulla express iNOS and that SEB induces iNOS expression in the mouse thymus.


Asunto(s)
Histocitoquímica/métodos , NADPH Deshidrogenasa/metabolismo , Óxido Nítrico Sintasa/metabolismo , Timo/enzimología , Animales , Enterotoxinas/farmacología , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo II , Ratas , Ratas Wistar , Superantígenos/farmacología , Timo/efectos de los fármacos
13.
Immunol Lett ; 51(3): 163-7, 1996 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-8832286

RESUMEN

The inducible isoform of nitric oxide synthase (iNOS) produces large quantities of nitric oxide (NO) during inflammation and shock. Recent studies show that the reaction of NO with superoxide yields the cytotoxic oxidant peroxynitrite (ONOO-). An important pathway of ONOO- cytotoxicity involves DNA strand breakage, activation of the nuclear repair enzyme poly(ADP) ribosyltransferase (PARS), and concomitant ADP-ribosylation, NAD+ consumption, and exhaustion of intracellular energy stores. Using quin-2, a calcium chelator, we have investigated the role of calcium in the cytotoxicity elicited by ONOO-. Quin-2 (10-100 microM) ameliorated the suppression of mitochondrial respiration in response to ONOO- (1 mM) in J774 macrophages. Quin-2 at 100 microM, but not at 10 microM, caused a small (20%) inhibition of PARS activity, and did not significantly affect NAD+ depletion. Quin-2 exhibited a slight protective effect against the decrease in mitochondrial respiration in immunostimulated macrophages which endogenously produce ONOO-. These results suggest that the protective effect of quin-2 against the ONOO(-)-induced cellular injury is not due to interference with PARS activation or NAD+ depletion, but rather due to interference with a delayed intracellular event, possibly terminal calcium overload due to inhibition of mitochondrial enzymes and membrane pumps. Inhibition of calcium overload may be a viable experimental strategy to limit ONOO- cytotoxicity.


Asunto(s)
Calcio/antagonistas & inhibidores , Calcio/metabolismo , Macrófagos/efectos de los fármacos , Nitratos/toxicidad , Aminoquinolinas/farmacología , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Respiración/efectos de los fármacos
14.
Br J Pharmacol ; 129(5): 909-14, 2000 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10696089

RESUMEN

Immune cell activation releases ATP into the extracellular space. ATP-sensitive P2 purinergic receptors are expressed on immune cells and activation of these receptors alters immune cell function. Furthermore, ATP is metabolized by ectonucleotidases to adenosine, which has also been shown to alter cytokine production. In the present study, we investigated how extracellular ATP affects interleukin (IL)-12 and tumour necrosis factor (TNF)-alpha production in bacterial lipopolysaccharide (LPS)-treated murine peritoneal macrophages and we also examined whether extracellular ATP alters the production of the T helper 1 cytokine interferon (IFN)-gamma. Pretreatment of the peritoneal macrophages with ATP or various ATP analogues decreased both IL-12 and TNF-alpha production induced by LPS (10 microgram ml(-1)). The effect of ATP was partially reversed by cotreatment with adenosine deaminase (0.1 - 1 u ml(-1)), suggesting that the suppressive effect of ATP on cytokine production is, in part, due to its degradation products. Immunoneutralization with an anti-IL-10 antibody demonstrated that although ATP increases IL-10 production, the inhibition of IL-12 and TNF-alpha production is independent of the increased IL-10. The effect of ATP was pretranslational, as it suppressed steady state levels of mRNAs for IL-12 (both p35 and p40). In spleen cells stimulated with either LPS (10 microgram ml(-1)) or anti-CD3 (2 microgram ml(-1)) antibody, ATP suppressed, in a concentration-dependent manner, the production of IFN-gamma. These results suggest that extracellular ATP has multiple anti-inflammatory effects and that release of ATP into the extracellular space may play a role in blunting the overactive immune response in autoimmune diseases.


Asunto(s)
Adenosina Trifosfato/farmacología , Interleucina-12/metabolismo , Macrófagos Peritoneales/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Complejo CD3/inmunología , Interferón gamma/biosíntesis , Interleucina-10/metabolismo , Lipopolisacáridos/farmacología , Macrófagos Peritoneales/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Consumo de Oxígeno/efectos de los fármacos , Antagonistas del Receptor Purinérgico P2 , ARN/biosíntesis , ARN/aislamiento & purificación , Ribonucleasas/metabolismo , Bazo/citología , Bazo/efectos de los fármacos , Bazo/metabolismo
15.
Br J Pharmacol ; 121(6): 1065-74, 1997 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9249240

RESUMEN

1. Peroxynitrite, a potent cytotoxic oxidant formed by the reaction of nitric oxide with superoxide anion, and hydroxyl radical, formed in the iron-catalysed Fenton reaction, are important mediators of reperfusion injury. In in vitro studies, DNA single strand breakage, triggered by peroxynitrite or by hydroxyl radical, activates the nuclear enzyme poly (ADP-ribose) synthetase (PARS), with consequent cytotoxic effects. Using 3-aminobenzamide, an inhibitor of PARS, we investigated the role of PARS in the pathogenesis of splanchnic artery occlusion shock. 2. Splanchnic artery occlusion and reperfusion shock (SAO/R) was induced in rats by clamping both the superior mesenteric artery and the coeliac trunk for 45 min, followed by release of the clamp (reperfusion). At 60 min after reperfusion, animals were killed for histological examination and biochemical studies. 3. SAO/R rats developed a significant fall in mean arterial blood pressure, significant increase of tissue myeloperoxidase activity and marked histological injury to the distal ileum. SAO/R was also associated with a significant mortality (0% survival at 2 h after reperfusion). 4. There was a marked increase in the oxidation of dihydrorhodamine 123 to rhodamine (a marker of peroxynitrite-induced oxidative processes) in the plasma of the SAO/R rats, starting early after reperfusion, but not during ischaemia alone. Immunohistochemical examination demonstrated a marked increase in the immunoreactivity to nitrotyrosine, a specific 'footprint' of peroxynitrite, in the necrotic ileum in shocked rats, as measured at 60 min after the start of reperfusion. 5. In addition, in ex vivo studies in aortic rings from shocked rats, we found reduced contractions to noradrenaline and reduced responsiveness to a relaxant effect to acetylcholine (vascular hyporeactivity and endothelial dysfunction, respectively). 6. In a separate set of studies, using a 4000 Dalton fluorescent dextran tracer, we investigated the changes in epithelial permeability associated with SAO/R. Ten minutes of reperfusion, after 30 min of splanchnic artery ischaemia, resulted in a marked increase in epithelial permeability. 7. There was a significant increase in PARS activity in the intestinal epithelial cells, as measured 10 min after reperfusion ex vivo. 3-Aminobenzamide, a pharmacological inhibitor of PARS (applied at 10 mg kg(-1), i.v., 5 min before reperfusion, followed by an infusion of 10 mg kg(-1) h(-1)), significantly reduced ischaemia/reperfusion injury in the bowel, as evaluated by histological examination. Also it significantly improved mean arterial blood pressure, improved contractile responsiveness to noradrenaline, enhanced the endothelium-dependent relaxations and reduced the reperfusion-induced increase in epithelial permeability. 8. 3-Aminobenzamide also prevented the infiltration of neutrophils into the reperfused intestine, as evidenced by reduced myeloperoxidase activity. It improved the histological status of the reperfused tissues, reduced the production of peroxynitrite in the late phase of reperfusion and improved survival. 9. In conclusion, our study demonstrates that the PARS inhibitor 3-aminobenzamide exerts multiple protective effects in splanchnic artery occlusion/reperfusion shock. We suggest that peroxynitrite and/or hydroxyl radical, produced during the reperfusion phase, trigger DNA strand breakage, PARS activation and subsequent cellular dysfunction. The vascular endothelium is likely to represent an important cellular site of protection by 3-aminobenzamide in SAO shock.


Asunto(s)
Benzamidas/farmacología , Inhibidores Enzimáticos/farmacología , Arterias Mesentéricas/efectos de los fármacos , Oclusión Vascular Mesentérica/enzimología , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Daño por Reperfusión/enzimología , Animales , Presión Sanguínea , Permeabilidad de la Membrana Celular , Activación Enzimática , Técnicas In Vitro , Intestinos/patología , Masculino , Arterias Mesentéricas/enzimología , Arterias Mesentéricas/metabolismo , Oclusión Vascular Mesentérica/metabolismo , Oclusión Vascular Mesentérica/patología , Modelos Biológicos , Nitratos/metabolismo , Óxido Nítrico/biosíntesis , Poli(ADP-Ribosa) Polimerasas/metabolismo , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/patología , Tasa de Supervivencia
16.
Br J Pharmacol ; 120(3): 357-66, 1997 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9031736

RESUMEN

1. It has been proposed that in inflammatory conditions, in which both the inducible isoforms of nitric oxide synthase (iNOS) and cyclo-oxygenase (COX-2) are induced, inhibition of NOS also results in inhibition of arachidonic acid metabolism. In the present study we have investigated whether mercaptoalkylguanidines, a novel class of selective iNOS inhibitors, may also influence the activity of cyclo-oxygenase (COX). Therefore, the effect of mercaptoethylguanidine (MEG) and related compounds on the activity of the constitutive (COX-1) and the inducible COX (COX-2) was investigated in cells and in purified enzymes. Aminoguanidine, NG-methyl-L-arginine (L-NMA) and NG-nitro-L-arginine methyl ester (L-NAME) were also studied for comparative purposes. 2. Western blot analysis demonstrated a significant COX-1 activity in unstimulated J774 macrophages and in unstimulated human umbilical vein endothelial cells (HUVEC). Immunostimulation of the J774 macrophages by endotoxin (lipopolysaccharide of E. coli, LPS 10 micrograms ml-1) and interferon gamma (IFN gamma, 100 u ml-1) for 6 h resulted in a significant induction of COX-2, and a down-regulation of COX-1. No COX-2 immunoreactivity was detected in unstimulated HUVEC or unstimulated J774 cells. Therefore, in subsequent studies, the effect of mercaptoalkylguanidines on COX-1 activity was studied in HUVEC stimulated with arachidonic acid for 6 h, and in J774 cells stimulated with arachidonic acid for 30 min. The effect of mercaptoalkylguanidines on COX-2 activity was studied in immunostimulated J774 macrophages, both on prostaglandin production by endogenous sources, and on prostaglandin production in response to exogenous arachidonic acid stimulation. In addition, the effect of mercaptoalkylguanidines on purified COX-1 and COX-2 activities was also studied. 3. In experiments designed to measure COX-1 activity in HUVEC, the cells were stimulated by arachidonic acid (15 microM) for 6 h. This treatment induced a significant production of 6-keto-prostaglandin F1 alpha (6-keto-PGF1 alpha, the stable metabolite of prostacyclin), while nitrite production was undetectable by the Griess reaction. MEG (1 microM to 3 mM) caused a dose-dependent inhibition of the accumulation of 6-keto-PGF1 alpha, with an IC50 of 20 microM. However, aminoguanidine, L-NAME or L-NMA (up to 3 mM) did not affect the production of 6-keto-PGF1 alpha in this experimental system. In experiments designed to measure COX-1 activity in J774.2 macrophages, the cells were stimulated by arachidonic acid (15 microM) for 30 min; this also induced a significant production of 6-keto-PGF1 alpha and MEG (1 microM to 3 mM), aminoguanidine (at 1 and 3 mM), but neither L-NAME nor L-NMA inhibited the production of prostaglandins. 4. In experiments designed to measure prostaglandin production by COX-2 with endogenous arachidonic acid, J774.2 cells were immunostimulated for 6 h in the absence or presence of various inhibitors. In experiments designed to measure prostaglandin production by COX-2 with exogenous arachidonic acid, J774.2 cells were immunostimulated for 6 h, followed by a replacement of the culture medium with fresh medium containing arachidonic acid and various inhibitors. Both of these treatments induced a significant production of 6-keto-PGF1 alpha. Nitrite production, an indicator of NOS activity, was moderately increased after immunostimulation. MEG (1 microM to 3 mM) caused a dose-dependent inhibition of the accumulation of COX metabolites. Similar inhibition of LPS-stimulated 6-keto PGF1 alpha production was shown by other mercaptoalkylguanidines (such as N-methyl-mercaptoethylguanidine, N,N'-dimethyl-mercaptoethylguanidine, S-methyl-mercaptoethylguanidine and guanidino-ethyldisulphide), with IC50 values ranging between 34-55 microM. However, aminoguanidine, L-NAME and L-NMA (up to 3 mM) did not affect the production of prostaglandins.5. In comparative experiments indomethacin, a non selective COX inhibitor, and NS-398, a selective COX-2 inhibitor, reduced (LPS) stimulated 6-keto-PGF1alpha production in J774 macrophages in a dose-dependent manner without affecting nitrite release. Indomethacin, but not NS-398, inhibited 6-keto-PGF1alpha production in the HUVECs. 6.The inhibitory effect of MEG was due to direct inhibition of the catalytic activity of COX as indicated in experiments with purified COX-1 and COX-2. MEG dose-dependently inhibited the purified COX-1 and COX-2 activity with IC50 values of 33microM and 36microM, respectively. Aminoguanidine (at the highest concentrations) inhibited the formation of COX-1 metabolites, without affecting COX-2 activity. High doses of L-NAME (3mM) decreased COX-1 activity only, while L-NMA (up to 3mM) had no effect on the activity of either enzyme. 7.These results suggest that MEG and related compounds are direct inhibitors of the constitutive and the inducible cyclo-oxygenases, in addition to their effects on the inducible NOS. The additional effect of mercaptoalkylguanidines on COX activity may contribute to the beneficial effects of these agents in inflammatory conditions where both iNOS and COX-2 are expressed.


Asunto(s)
Inhibidores de la Ciclooxigenasa/síntesis química , Guanidinas/síntesis química , 6-Cetoprostaglandina F1 alfa/metabolismo , Ácido Araquidónico/farmacología , Western Blotting , Línea Celular , Supervivencia Celular/efectos de los fármacos , Inhibidores de la Ciclooxigenasa/farmacología , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/enzimología , Activación Enzimática/efectos de los fármacos , Guanidinas/farmacología , Humanos , Óxido Nítrico Sintasa/antagonistas & inhibidores , Nitritos/metabolismo , Estimulación Química , Especificidad por Sustrato , Tromboxano B2/metabolismo
17.
Br J Pharmacol ; 122(3): 493-503, 1997 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9351506

RESUMEN

1. Zymosan is a wall component of the yeast Saccharomyces Cerevisiae. Injection of zymosan into experimental animals is known to produce an intense inflammatory response. Recent studies demonstrated that the zymosan-induced inflammatory response in vivo can be ameliorated by inhibitors of nitric oxide (NO) biosynthesis. The cytotoxic effects of NO are, in part, mediated by the oxidant preoxynitrite and subsequent activation of the nuclear enzyme poly (ADP-ribose) synthetase (PARS). In the present in vitro study, we have investigated the cellular mechanisms of vascular failure elicited by zymosan-activated plasma and the contribution of peroxynitrite production and activation of PARS to the changes. 2. Incubation of rat aortic smooth muscle cells with zymosan-activated plasma (ZAP) induced the production of nitrite, the breakdown product of NO, due to the expression of the inducible isoform of NO synthase (iNOS) over 6 24 h. In addition, ZAP triggered the production of peroxynitrite in these cells, as measured by the oxidation of the fluorescent dye dihydrorhodamine 123 and by nitrotyrosine Western blotting. 3. Incubation of the smooth muscle cells with ZAP induced DNA single strand breakage and PARS activation. These effects were reduced by inhibition of NOS with NG-methyl-L-arginine (L-NMA, 3 mM), and by glutathione (3 mM), a scavenger of peroxynitrite. The PARS inhibitor 3-aminobenzamide (1 mM) inhibited the ZAP-induced activation of PARS. 4. Incubation of thoracic aortae with ZAP in vitro caused a reduction of the contractions of the blood vessels to noradrenaline (vascular hyporeactivity) and elicited a reduced responsiveness to the endothelium-dependent vasodilator acetylcholine (endothelial dysfunction). 5. Preincubation of the thoracic aortae with L-NMA (1 mM), glutathione (3 mM) or by the PARS inhibitor 3-aminobenzamide (1 mM) prevented the development of vascular hyporeactivity in response to ZAP. Moreover, glutathione and 3-aminobenzamide treatment protected against the ZAP-induced development of endothelial dysfunction. The PARS-related loss of the vascular contractility was evident at 30 min after incubation in endothelium-intact, but not in endothelium-denuded vessels and also manifested at 6 h after incubation with ZAP in endothelium-denuded rings. The acute response is probably related, therefore, to peroxynitrite formation (involving the endothelial NO synthase), whereas the delayed response may be related to the expression of iNOS in the smooth muscle. 6. The data obtained suggest that zymosan-activated plasma causes vascular dysfunction by inducing the simultaneous formation of superoxide and NO. These radicals combine to form peroxynitrite, which, in turn causes DNA injury and PARS activation. The protective effect of 3-aminobenzamide demonstrates that PARS activation contributes both to the development of vascular hyporeactivity and endothelial dysfunction during the vascular failure induced by ZAP.


Asunto(s)
Músculo Liso Vascular/efectos de los fármacos , Nitratos/fisiología , Oxidantes/fisiología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Zimosan/farmacología , Animales , Aorta/efectos de los fármacos , Aorta/enzimología , Aorta/metabolismo , Aorta Torácica/efectos de los fármacos , Western Blotting , Células Cultivadas , Daño del ADN , Activación Enzimática/efectos de los fármacos , Técnicas In Vitro , Contracción Muscular/efectos de los fármacos , Músculo Liso Vascular/enzimología , Músculo Liso Vascular/metabolismo , Óxido Nítrico/biosíntesis , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/metabolismo , Poli(ADP-Ribosa) Polimerasas/sangre , Ratas , Tirosina/metabolismo , Zimosan/sangre
18.
Br J Pharmacol ; 123(3): 525-37, 1998 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9504394

RESUMEN

1. Peroxynitrite, a cytotoxic oxidant formed from the reaction of nitric oxide (NO) and superoxide is a mediator of cellular injury in ischaemia/reperfusion injury, shock and inflammation. Here we investigated whether L-buthionine-(S,R)-sulphoximine (BSO), an inhibitor of gamma-glutamylcysteine synthetase, alters endothelial and vascular smooth muscle injury in response to peroxynitrite in vitro and during endotoxic shock in vivo. 2. In human umbilical vein endothelial cells and in rat aortic smooth muscle cells, BSO (1 mM, for 24 h) enhanced, whereas glutathione (3 mM) or glutathione ethyl ester (3 mM) attenuated the peroxynitrite (100-1000 microM)-induced suppression of mitochondrial respiration (measured by the conversion of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) to formazan), formation of nitrotyrosine (detected by Western blotting), protein oxidation (measured by detection of 2,4 dinitrophenylhydrazine-reactive carbonyls), and DNA single strand breakage and activation of the nuclear enzyme poly (ADP-ribose) synthetase (PARS) (measured by the incorporation of radiolabelled NAD+ into nuclear proteins and by the alkaline unwinding assay, respectively). Glutathione ethyl ester treatment reduced the BSO-induced enhancement of peroxynitrite-induced cytotoxicity. 3. In rat isolated thoracic aortic rings, BSO treatment (in vivo, at 1 g kg(-1) intraperitoneally (i.p.) for 24 h) enhanced, whereas pretreatment with glutathione (in vitro, 3 mM) attenuated the peroxynitrite-induced reduction of the contractions to noradrenaline, and the peroxynitrite-induced impairment of the endothelium-dependent relaxations to acetylcholine. 4. In BSO-pretreated rats, treatment with bacterial lipopolysaccharide (LPS, 15 mg kg(-1), i.p., for 6 h) caused a more pronounced vascular hyporeactivity and endothelial dysfunction ex vivo. BSO pretreatment also increased the degree of nitrotyrosine staining (detected by imunohistochemistry) in the aorta after LPS treatment. 5. In conclusion, our results demonstrate that L-buthionine-(S,R)-sulphoximine, an inhibitor of gamma-glutamylcysteine synthetase enhances peroxynitrite- and endotoxic shock-induced vascular failure. Based on these findings, we suggest that endogenous glutathione plays an important protective role against peroxynitrite- and LPS-induced vascular injury.


Asunto(s)
Butionina Sulfoximina/farmacología , Endotelio Vascular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Glutamato-Cisteína Ligasa/antagonistas & inhibidores , Nitratos/farmacología , Choque Séptico/fisiopatología , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/fisiología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/fisiología , Inducción Enzimática , Glutatión/fisiología , Humanos , Técnicas In Vitro , Contracción Muscular/efectos de los fármacos , Óxido Nítrico Sintasa/biosíntesis , Ratas
19.
Br J Pharmacol ; 117(4): 619-32, 1996 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-8646406

RESUMEN

1. The generation of nitric oxide (NO) from L-arginine by NO synthases (NOS) can be inhibited by guanidines, amidines and S-alkylisothioureas. Unlike most L-arginine based inhibitors, however, some guanidines and S-alkylisothioureas, in particular aminoethylisothiourea (AETU), show selectivity towards the inducible isoform (iNOS) over the constitutive isoforms (endothelial, ecNOS and brain isoform, bNOS) and so may be of therapeutic benefit. In the present study we have investigated the effects of AETU and other aminoalkylisothioureas on the activities of iNOS, ecNOS and bNOS. 2. AETU, aminopropylisothiourea (APTU) and their derivatives containing alkyl substituents on one of the amidino nitrogens, potently inhibit nitrite formation by immunostimulated J774 macrophages (a model of iNOS activity) with EC50 values ranging from 6-30 microM (EC50 values for NG-methyl-L-arginine (L-NMA) and NG-nitro-L-arginine were 159 and > 1000 microM, respectively). The inhibitory effects of these aminoalkylisothioureas (AATUs) were attentuated by L-arginine in the incubation medium, indicating that these agents may complete with L-arginine for its binding site on NOS. 3. The above AATUs undergo chemical conversion in neutral or basic solution (pH 7 or above) as indicated by (1) the disappearance of AATUs from solution as measured by h.p.l.c., (2) the generation of free thiols not previously present and (3) the isolation of species (as picrate and flavianate salts) from neutral or basic solutions of AATUs that are different from those obtained from acid solutions. 4. Mercaptoalkylguanidines (MAGs) were prepared and shown to be potent inhibitors of iNOS activity with EC50s comparable to those of their isomeric AATUs. 5. These findings suggest that certain AATUs exert their potent inhibitory effects through intramolecular rearrangement to mercaptoalkylguanidines (MAGs) at physiological pH. Those AATUs not capable of such rearrangement do not exhibit the same degree of inhibition of iNOS. 6. In contrast to their potent effects on iNOS, some AATUs and MAGs were 20-100 times weaker than NG-methyl-L-arginine and NG-nitro-L-arginine as inhibitors of ecNOS as assessed by their effects on the conversion of L-arginine to L-citrulline in homogenates of bovine endothelial cells and by their pressor effects in anaesthetized rats. Thus mercaptoalkylguanidines represent a new class of NOS inhibitors with preference towards iNOS. 7. AETU and mercaptoethylguanidine (MEG), when given as infusions, gave slight decreases in MAP in control rats. However, infusions of AETU or MEG to endotoxin-treated rats caused an increase in MAP and restored 80% of the endotoxin-induced fall in MAP. 8. High doses of MEG (30-60 mg kg-1) caused a decrease in MAP of normal rats. This depressor effect may be a consequence of the in vivo oxidation of MEG to the disulphide, guanidinoethyldisulphide (GED), which caused pronounced, transient hypotensive responses in anaesthetized rats and caused endothelium-independent vasodilator responses in precontracted rat aortic rings in vitro. 9. In some cases, slight differences were observed in the activities of AATUs and the corresponding MAGs. These may be explained by the formation of other species from AATUs in physiological media. For example, AETU can give rise to small amounts of the potent ecNOS inhibitor, 2-aminothiazoline, in addition to MEG. This may account for the differences in the in vitro and in vivo effects of AETU and MEG. 10. In conclusion, the in vitro and in vivo effects of AETU and related aminoalkylisothioureas can be explained in terms of their intramolecular rearrangement to generate mercaptoalkylguanidines, a novel class of selective inhibitors of iNOS.


Asunto(s)
Inhibidores Enzimáticos/química , Guanidinas/química , Isoenzimas/análisis , Óxido Nítrico Sintasa/antagonistas & inhibidores , Tiourea/análogos & derivados , Animales , Aorta/efectos de los fármacos , Aorta/fisiología , Arginina/farmacología , Presión Sanguínea/efectos de los fármacos , Bovinos , Línea Celular , Inducción Enzimática , Inhibidores Enzimáticos/farmacología , Guanidinas/farmacología , Técnicas In Vitro , Isoenzimas/biosíntesis , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Contracción Muscular/efectos de los fármacos , Óxido Nítrico Sintasa/biosíntesis , Nitritos/metabolismo , Ratas , Compuestos de Sulfhidrilo/metabolismo , Tiourea/química
20.
Br J Pharmacol ; 120(2): 259-67, 1997 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9117118

RESUMEN

1. Peroxynitrite is a toxic oxidant species produced from nitric oxide (NO) and superoxide. We have recently observed that the cell-permeable superoxide dismutase mimetic Mn(III)tetrakis(4-benzoic acid) porphyrin (MnTBAP) inhibits the suppression of mitochondrial respiration elicited by authentic peroxynitrite in vitro. Here we have investigated the relative potency of MnTBAP and a range of related compounds in terms of inhibition of peroxynitrite-induced oxidation and cytotoxicity. In addition, we tested the effects of MnTBAP on the vascular and the cellular energetic failure in rodent models of endotoxic shock. 2. We observed a dose-related inhibition of the peroxynitrite-induced oxidation of dihydrorhodamine 123 to rhodamine by MnTBAP, ZnTBAP and FeTBAP, but not by MnTMPyP [(5,10,15,20-tetrakis(N-methyl-4'-pirydyl)porphinato)-mangan ese (III)]. In addition, MnTBAP, ZnTBAP and FeTBAP, but not MnTMPyP prevented the suppression of mitochondrial respiration by authentic peroxynitrite in cultured J774 macrophages. 3. In rat cultured aortic smooth muscle cells, MnTBAP protected against the suppression of mitochondrial respiration in response to authentic peroxynitrite, immunostimulation and nitric oxide (NO) donor compounds. MnTBAP slightly reduced the amount of nitrite/nitrate produced in response to immunostimulation in these cells. 4. Administration of MnTBAP, 15 mg kg-1 i.v., before the administration of endotoxin (15 mg kg-1, i.v.) to rats ameliorated the development of vascular hyporeactivity and the development of endothelial dysfunction in the thoracic aorta ex vivo. 5. MnTBAP also prevented the endotoxin-induced decrease in mitochondrial respiration, the development of DNA single strand breaks, and the depletion of intracellular NAD+ in peritoneal macrophages ex vivo. 6. MnTBAP did not inhibit the expression by endotoxin of the inducible NO synthase in lung samples. 7. MnTBAP did not alter survival rate in mice challenged with high dose endotoxin. 8. Our findings, taken together with previous data demonstrating protective effects of NO synthase inhibitors against the endotoxin-induced contractile and energetic failure in the models of shock used in the current study, and with the known ability of peroxynitrite to cause cellular energy depletion, suggest a role for peroxynitrite in the pathogenesis of cellular energetic failure and contractile dysfunction in endotoxin shock.


Asunto(s)
Metaloporfirinas/farmacología , Nitratos/toxicidad , Consumo de Oxígeno/efectos de los fármacos , Choque Séptico/fisiopatología , Animales , Células Cultivadas , Lipopolisacáridos/farmacología , Masculino , Ratones , Mitocondrias/metabolismo , Músculo Liso Vascular/efectos de los fármacos , Óxido Nítrico/biosíntesis , Ratas , Ratas Wistar , Vasoconstricción
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda