Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
3.
Am J Transplant ; 12(11): 2938-48, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23016570

ABSTRACT

Ischemia/reperfusion injury (IRI) is the most common cause of early mortality following lung transplantation (LTx). We hypothesized that nitrite, an endogenous source of nitric oxide (NO), may protect lung grafts from IRI. Rat lung grafts were stored in preservation solution at 4°C for 6 hours. Both grafts and recipients were treated with nitrite. Nitrite treatment was associated with significantly higher levels of tissue oxygenation, lower levels of cytokines and neutrophil/macrophage infiltration, lower myeloperoxidase activity, reduced oxidative injury and increased cGMP levels in grafts than in the controls. Treatment with either a nitric oxide scavenger or a soluble guanylyl cyclase (sGC) inhibitor diminished the beneficial effects of nitrite and decreased cGMP concentrations. These results suggest that nitric oxide, generated from nitrite, is the molecule responsible for the effects of nitrite via the nitric oxide/sGC/cGMP pathway. Allopurinol, a xanthine oxidoreductase (XOR) inhibitor, abrogated the protective effects of nitrite, suggesting that XOR is a key enzyme in the conversion of nitrite to nitric oxide. In vitro experiments demonstrated that nitrite prevented apoptosis in pulmonary endothelial cells. Nitrite also exhibits longer survival rate in recipients than control. In conclusion, nitrite inhibits lung IRI following cold preservation and had higher survival rate in LTx model.


Subject(s)
Acute Lung Injury/prevention & control , Lung Transplantation/adverse effects , Nitrites/pharmacology , Oxidative Stress/physiology , Reperfusion Injury/prevention & control , Acute Lung Injury/etiology , Animals , Disease Models, Animal , Graft Rejection , Graft Survival/drug effects , Lung Transplantation/methods , Male , Nitric Oxide/metabolism , Peroxidase/metabolism , Random Allocation , Rats , Rats, Inbred Lew , Rats, Sprague-Dawley , Reference Values
4.
Proc Natl Acad Sci U S A ; 98(26): 15215-20, 2001 Dec 18.
Article in English | MEDLINE | ID: mdl-11752464

ABSTRACT

Plasma xanthine oxidase (XO) activity was defined as a source of enhanced vascular superoxide (O(2)( *-)) and hydrogen peroxide (H(2)O(2)) production in both sickle cell disease (SCD) patients and knockout-transgenic SCD mice. There was a significant increase in the plasma XO activity of SCD patients that was similarly reflected in the SCD mouse model. Western blot and enzymatic analysis of liver tissue from SCD mice revealed decreased XO content. Hematoxylin and eosin staining of liver tissue of knockout-transgenic SCD mice indicated extensive hepatocellular injury that was accompanied by increased plasma content of the liver enzyme alanine aminotransferase. Immunocytochemical and enzymatic analysis of XO in thoracic aorta and liver tissue of SCD mice showed increased vessel wall and decreased liver XO, with XO concentrated on and in vascular luminal cells. Steady-state rates of vascular O(2)( *-) production, as indicated by coelenterazine chemiluminescence, were significantly increased, and nitric oxide (( *)NO)-dependent vasorelaxation of aortic ring segments was severely impaired in SCD mice, implying oxidative inactivation of ( *)NO. Pretreatment of aortic vessels with the superoxide dismutase mimetic manganese 5,10,15,20-tetrakis(N-ethylpyridinium-2-yl)porphyrin markedly decreased O(2)( small middle dot-) levels and significantly restored acetylcholine-dependent relaxation, whereas catalase had no effect. These data reveal that episodes of intrahepatic hypoxia-reoxygenation associated with SCD can induce the release of XO into the circulation from the liver. This circulating XO can then bind avidly to vessel luminal cells and impair vascular function by creating an oxidative milieu and catalytically consuming (*)NO via O(2)( small middle dot-)-dependent mechanisms.


Subject(s)
Anemia, Sickle Cell/physiopathology , Endothelium, Vascular/physiopathology , Muscle Relaxation/physiology , Nitric Oxide/physiology , Superoxides/metabolism , Alanine Transaminase/blood , Animals , Endothelium, Vascular/metabolism , Erythrocytes/metabolism , Humans , In Vitro Techniques , Mice , Mice, Knockout , Xanthine Oxidase/blood
5.
Circ Res ; 89(3): 224-36, 2001 Aug 03.
Article in English | MEDLINE | ID: mdl-11485972

ABSTRACT

The evanescent nature of reactive oxygen and nitrogen species, the multiple cellular mechanisms evolved to maintain these substances at low (submicromolar) concentrations within the vascular system, and the often multifaceted nature of their reactivities have made measurement of these compounds within the vasculature problematic. This review attempts to provide a critical description of some of the most common approaches to quantification of nitric oxide, superoxide, hydrogen peroxide, and peroxynitrite, with attention to key issues that may influence the utility of a particular assay when adapted for use in vascular cells and tissues.


Subject(s)
Blood Vessels/chemistry , Hydrogen Peroxide/analysis , Nitrates/analysis , Nitric Oxide/analysis , Superoxides/analysis , Blood Vessels/metabolism , Electrochemistry , Free Radicals/chemistry , Oxidation-Reduction
6.
Circulation ; 103(9): 1282-8, 2001 Mar 06.
Article in English | MEDLINE | ID: mdl-11238274

ABSTRACT

BACKGROUND: Altered endothelial cell nitric oxide (NO(*)) production in atherosclerosis may be due to a reduction of intracellular tetrahydrobiopterin, which is a critical cofactor for NO synthase (NOS). In addition, previous literature suggests that inactivation of NO(*) by increased vascular production superoxide (O(2)(*-)) also reduces NO(*) bioactivity in several disease states. We sought to determine whether these 2 seemingly disparate mechanisms were related. METHODS AND RESULTS: Endothelium-dependent vasodilation was abnormal in aortas of apoE-deficient (apoE(-/-)) mice, whereas vascular superoxide production (assessed by 5 micromol/L lucigenin) was markedly increased. Treatment with either liposome-entrapped superoxide dismutase or sepiapterin, a precursor to tetrahydrobiopterin, improved endothelium-dependent vasodilation in aortas from apoE(-/-) mice. Hydrogen peroxide had no effect on the decay of tetrahydrobiopterin, as monitored spectrophotometrically. In contrast, superoxide modestly and peroxynitrite strikingly increased the decay of tetrahydrobiopterin over 500 seconds. Luminol chemiluminescence, inhibitable by the peroxynitrite scavengers ebselen and uric acid, was markedly increased in apoE(-/-) aortic rings. In vessels from apoE(-/-) mice, uric acid improved endothelium-dependent relaxation while having no effect in vessels from control mice. Treatment of normal aortas with exogenous peroxynitrite dramatically increased vascular O(2)(*-) production, seemingly from eNOS, because this effect was absent in vessels lacking endothelium, was blocked by NOS inhibition, and did not occur in vessels from mice lacking eNOS. CONCLUSIONS: Reactive oxygen species may alter endothelium-dependent vascular relaxation not only by the interaction of O(2)(*-) with NO(*) but also through interactions between peroxynitrite and tetrahydrobiopterin. Peroxynitrite oxidation of tetrahydrobiopterin may represent a pathogenic cause of "uncoupling" of NO synthase.


Subject(s)
Apolipoproteins E/deficiency , Biopterins/analogs & derivatives , Endothelium, Vascular/physiology , Pterins , Vasodilation/physiology , Acetylcholine/pharmacology , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Aorta, Thoracic/physiology , Apolipoproteins E/genetics , Biopterins/metabolism , Calcimycin/pharmacology , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Female , In Vitro Techniques , Ionophores/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitrates/pharmacology , Nitric Oxide Synthase/deficiency , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Nitroglycerin/pharmacology , Pteridines/pharmacology , Superoxides/metabolism , Superoxides/pharmacology , Vasodilation/drug effects , Vasodilator Agents/pharmacology
7.
Free Radic Biol Med ; 28(3): 437-46, 2000 Feb 01.
Article in English | MEDLINE | ID: mdl-10699756

ABSTRACT

Inhibition of nitric oxide synthesis prevents rat embryonic motor neurons from undergoing apoptosis when initially cultured without brain-derived neurotrophic factor. Using an improved cell culture medium, we found that the partial withdrawal of trophic support even weeks after motor neurons had differentiated into a mature phenotype still induced apoptosis through a process dependent upon nitric oxide. However, nitric oxide itself was not directly toxic to motor neurons. To investigate whether intracellular superoxide contributed to nitric oxide-dependent apoptosis, we developed a novel method using pH-sensitive liposomes to deliver Cu, Zn superoxide dismutase intracellularly into motor neurons. Intracellular superoxide dismutase prevented motor neuron apoptosis from trophic factor withdrawal, whereas empty liposomes, inactivated superoxide dismutase in liposomes or extracellular superoxide dismutase did not. Neither hydrogen peroxide nor nitrite added separately or in combination affected motor neuron survival. Our results suggest that a partial reduction in trophic support induced motor neuron apoptosis by a process requiring the endogenous production of both nitric oxide and superoxide, irrespective of the extent of motor neuron maturation in culture.


Subject(s)
Cell Survival/drug effects , Motor Neurons/cytology , Nitric Oxide/pharmacology , Spinal Cord/cytology , Superoxide Dismutase/pharmacology , Animals , Brain-Derived Neurotrophic Factor/pharmacology , Caspases/metabolism , Cell Death/drug effects , Cells, Cultured , Cellular Senescence/drug effects , Cysteine Proteinase Inhibitors/pharmacology , Drug Carriers , Embryo, Mammalian , Humans , Hydrogen Peroxide/pharmacology , Liposomes , Motor Neurons/drug effects , Motor Neurons/physiology , NG-Nitroarginine Methyl Ester/pharmacology , Nerve Degeneration/prevention & control , Rats , Recombinant Proteins/administration & dosage , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Superoxide Dismutase/administration & dosage , Superoxide Dismutase/metabolism , Time Factors , omega-N-Methylarginine/pharmacology
8.
Anesthesiology ; 93(6): 1446-55, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11149440

ABSTRACT

BACKGROUND: Recent evidence implicates nitric oxide (*NO) in the pathogenesis of preeclampsia. The authors tested the hypothesis that administration of low-dose endotoxin to pregnant rats mimics the signs of preeclampsia in humans and that *NO and *NO-derived species play a role in that animal model. METHODS: Endotoxin was infused at doses of 1, 2 and 10 microg/kg over 1 h to rats on day 14 of pregnancy. Mean arterial pressure, urinary protein, urinary and plasma nitrite plus nitrate (NO2- + NO3-) concentrations, and platelet count were measured before and after the endotoxin infusion. In another group of pregnant rats, the nitric oxide synthase inhibitor L-nitroarginine methyl ester (L-NAME) was administered in drinking water at a dose of 3 mg x kg(-1) x d(-1) starting on day 7 of pregnancy. Endotoxin was then infused at 10 microg/kg on day 14 of pregnancy. Kidneys and uteroplacental units were examined histologically and analyzed immunohistochemically for 3-nitrotyrosine. RESULTS: Endotoxin administration at doses of 2 and 10 microg/kg caused proteinuria and thrombocytopenia in pregnant rats, but did not result in hypertension. Urinary NO2- + NO3- concentration, reflective of tissue *NO production rates, was significantly elevated in pregnant rats that received endotoxin at 10 microg/kg. Ingestion of L-NAME caused hypertension. Tissues from pregnant rats treated with L-NAME, endotoxin at 10 microg/kg, and a combination of L-NAME and endotoxin had increased 3-nitrotyrosine immunoreactivity. CONCLUSION: Nitric oxide either directly or through secondary species plays a significant role in the biochemical and physiologic changes that occur in a rodent model of endotoxin-induced injury.


Subject(s)
Disease Models, Animal , Endotoxins/administration & dosage , Nitric Oxide/physiology , Pre-Eclampsia/etiology , Animals , Blood Pressure/drug effects , Blood Pressure Determination , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Enzyme Inhibitors/pharmacology , Female , Kidney/drug effects , NG-Nitroarginine Methyl Ester/pharmacology , Nitrates/urine , Nitrites/urine , Pre-Eclampsia/blood , Pre-Eclampsia/physiopathology , Pregnancy , Rats , Rats, Sprague-Dawley
9.
Circ Res ; 84(10): 1203-11, 1999 May 28.
Article in English | MEDLINE | ID: mdl-10347095

ABSTRACT

Lucigenin-amplified chemiluminescence has frequently been used to assess the formation of superoxide in vascular tissues. However, the ability of lucigenin to undergo redox cycling in purified enzyme-substrate mixtures has raised questions concerning the use of lucigenin as an appropriate probe for the measurement of superoxide production. Addition of lucigenin to reaction mixtures of xanthine oxidase plus NADH resulted in increased oxygen consumption, as well as superoxide dismutase-inhibitable reduction of cytochrome c, indicative of enhanced rates of superoxide formation. Additionally, it was revealed that lucigenin stimulated oxidant formation by both cultured bovine aortic endothelial cells and isolated rings from rat aorta. Lucigenin treatment resulted in enhanced hydrogen peroxide release from endothelial cells, whereas exposure to lucigenin resulted in inhibition of endothelium-dependent relaxation in isolated aortic rings that was superoxide dismutase inhibitable. In contrast, the chemiluminescent probe coelenterazine had no significant effect on xanthine oxidase-dependent oxygen consumption, endothelial cell hydrogen peroxide release, or endothelium-dependent relaxation. Study of enzyme and vascular systems indicated that coelenterazine chemiluminescence is a sensitive marker for detecting both superoxide and peroxynitrite.


Subject(s)
Acridines/pharmacology , Endothelium, Vascular/metabolism , Imidazoles , Oxidants/analysis , Pyrazines/pharmacology , Superoxides/analysis , Acetylcholine/pharmacology , Animals , Cells, Cultured , Cytochrome c Group/metabolism , Endothelium, Vascular/chemistry , Endothelium, Vascular/cytology , Firefly Luciferin/pharmacology , Hydrogen Peroxide/metabolism , Luminescent Measurements , Male , NAD/metabolism , Nitrates/analysis , Nitrates/metabolism , Oxidants/metabolism , Oxygen Consumption/drug effects , Oxygen Consumption/physiology , Rats , Rats, Sprague-Dawley , Superoxides/metabolism , Vasodilator Agents/pharmacology , Xanthine Oxidase/pharmacology
10.
Circulation ; 99(1): 53-9, 1999.
Article in English | MEDLINE | ID: mdl-9884379

ABSTRACT

BACKGROUND: In a variety of disease states, endothelium-dependent vasodilation is abnormal. Reduced nitric oxide (NO) production, increased destruction of NO by superoxide, diminished cellular levels of L-arginine or tetrahydrobiopterin, and alterations in membrane signaling have been implicated. We examined these potential mechanisms in human vessels. METHODS AND RESULTS: Relaxations to acetylcholine, the calcium ionophore A23187, and nitroglycerin, as well as superoxide production and NO synthase expression, were examined in vascular segments from patients with identified cardiovascular risk factors. Endothelium-dependent relaxations were also studied after incubation with L-arginine, L-sepiapterin, and liposome-entrapped superoxide dismutase (SOD) and after organoid culture with cis-vaccenic acid. Relaxations to acetylcholine and to a lesser extent the calcium ionophore A23187 were highly variable and correlated with the number of risk factors present among the subjects studied. Treatment of vessels with L-arginine, L-sepiapterin, liposome-entrapped SOD, or cis-vaccenic acid did not augment endothelium-dependent relaxations. Hypercholesterolemia was the only risk factor associated with high levels of superoxide; however, there was no correlation between superoxide production and the response to either endothelium-dependent vasodilator used. CONCLUSIONS: In human internal mammary arteries, depressed endothelium-dependent relaxations could not be attributed to increases in vascular superoxide production, deficiencies in either L-arginine or tetrahydrobiopterin, or reduced membrane fluidity. Variability in signaling mechanisms may contribute to the differences in responses to acetylcholine and the calcium ionophore A23187.


Subject(s)
Endothelium, Vascular/drug effects , Mammary Arteries/drug effects , Superoxides/metabolism , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Acetylcholine/pharmacology , Calcimycin/pharmacology , Endothelium, Vascular/metabolism , Female , Humans , Male , Mammary Arteries/metabolism , Middle Aged , Nitroglycerin/pharmacology , Organ Culture Techniques , Risk Factors , Vasodilation/physiology
11.
Science ; 286(5449): 2498-500, 1999 Dec 24.
Article in English | MEDLINE | ID: mdl-10617463

ABSTRACT

Mutations in copper, zinc superoxide dismutase (SOD) have been implicated in the selective death of motor neurons in 2 percent of amyotrophic lateral sclerosis (ALS) patients. The loss of zinc from either wild-type or ALS-mutant SODs was sufficient to induce apoptosis in cultured motor neurons. Toxicity required that copper be bound to SOD and depended on endogenous production of nitric oxide. When replete with zinc, neither ALS-mutant nor wild-type copper, zinc SODs were toxic, and both protected motor neurons from trophic factor withdrawal. Thus, zinc-deficient SOD may participate in both sporadic and familial ALS by an oxidative mechanism involving nitric oxide.


Subject(s)
Amyotrophic Lateral Sclerosis/enzymology , Apoptosis , Motor Neurons/cytology , Nitric Oxide/metabolism , Superoxide Dismutase/metabolism , Zinc/metabolism , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Brain-Derived Neurotrophic Factor/pharmacology , Cells, Cultured , Chelating Agents/pharmacology , Copper/metabolism , Fluoresceins/metabolism , Liposomes , Motor Neurons/metabolism , Mutation , Nitrates/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type I , Oxidation-Reduction , Rats , Superoxide Dismutase/chemistry , Superoxide Dismutase/genetics , Superoxide Dismutase/toxicity , Superoxides/metabolism
14.
Circulation ; 95(3): 588-93, 1997 Feb 04.
Article in English | MEDLINE | ID: mdl-9024144

ABSTRACT

BACKGROUND: The major source of superoxide (.O2-) in vascular tissues is an NADH/NADPH-dependent, membrane-bound oxidase. We have previously shown that this oxidase is activated in angiotensin II-but not norepinephrine-induced hypertension. We hypothesized that hypertension associated with chronically elevated angiotensin II might be caused in part by vascular .O2- production. METHODS AND RESULTS: We produced hypertension in rats by a 5-day infusion of angiotensin II or norepinephrine. Rats were also treated with liposome-encapsulated superoxide dismutase (SOD) or empty liposomes. Arterial pressure was measured in conscious rats under baseline conditions and during bolus injections of either acetylcholine or nitroprusside. Vascular .O2- production was assessed by lucigenin chemiluminescence. In vitro vascular relaxations were examined in organ chambers. Norepinephrine infusion increased blood pressure to a similar extent as angiotensin II infusion (179 +/- 5 and 189 +/- 4 mm Hg, respectively). In contrast, angiotensin II-induced hypertension was associated with increased vascular .O2- production, whereas norepinephrine-induced hypertension was not. Treatment with liposome-encapsulated SOD reduced blood pressure by 50 mm Hg in angiotensin II-infused rats while having no effect on blood pressure in control rats or rats with norepinephrine-induced hypertension. Similarly, liposome-encapsulated SOD enhanced in vivo hypotensive responses to acetylcholine and in vitro responses to endothelium-dependent vasodilators in angiotensin II-treated rats. CONCLUSIONS: Hypertension caused by chronically elevated angiotensin II is mediated in part by .O2-, likely via degradation of endothelium-derived NO. Increased vascular .O2- may contribute to vascular disease in high renin/angiotensin II states.


Subject(s)
Angiotensin II , Hypertension/chemically induced , Hypertension/physiopathology , Norepinephrine , Superoxides/metabolism , Acetylcholine/pharmacology , Animals , Aorta/pathology , Blood Pressure/drug effects , Blood Vessels/metabolism , Drug Carriers , Hypertension/pathology , Liposomes , Macrophages/pathology , Male , Nitroprusside/pharmacology , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/administration & dosage , Superoxide Dismutase/pharmacology , Vasodilation
15.
Br J Pharmacol ; 119(3): 511-8, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8894171

ABSTRACT

1. This study was designed (i) to assess the effect of S-nitrosoglutathione monoethyl ester (GSNO-MEE), a membrane-permeable analogue of S-nitrosoglutathione (GSNO), on rat isolated heart during cardioplegic ischaemia, and (ii) to monitor the release of nitric oxide (.NO) from GSNO-MEE in intact hearts using endogenous myoglobin as an intracellular .NO trap and the hydrophilic N-methyl glucamine dithiocarbamate-iron (MGD-Fe2+) complex as an extracellular .NO trap. 2. During aerobic perfusion of rat isolated heart with GSNO-MEE (20 mumol 1(-1), there was an increase in cyclic GMP from 105 +/- 11 to 955 +/- 193 pmol g-1 dry wt. (P < 0.05), and a decrease in glycogen content from 119 +/- 3 to 96 +/- 2 mumol g-1 dry wt. (P < 0.05), and glucose-6-phosphate concentration from 258 +/- 22 in control to 185 +/- 17 nmol g-1 dry wt. (P < 0.05). During induction of cardioplegia, GSNO-MEE caused the accumulation of cyclic GMP (100 +/- 6 in control vs. 929 +/- 168 pmol g-1 dry wt. in GSNO-MEE-treated group, P < 0.05), and depletion of glycogen from 117 +/- 3 to 103 +/- 2 mumol g-1 dry wt. (P < 0.05) in myocardial tissue. 3. Inclusion of GSNO-MEE (20 mumol l-1) in the cardioplegic solution improved the recovery of developed pressure (46 +/- 8 vs. 71 +/- 3% of baseline, P < 0.05), and rate-pressure product from 34 +/- 6 to 63 +/- 5% of baseline (P < 0.05), and reduced the diastolic pressure during reperfusion from 61 +/- 7 in control to 35 +/- 5 mmHg (P < 0.05) after 35 min ischaemic arrest. GSH-MEE (20 mumol l-1) in the cardioplegic solution did not elicit the protective effect. 4. During cardioplegic ischaemia, GSNO-MEE (20-200 mumol l-1) induced the formation of nitrosylmyoglobin (MbNO), which was detected by electron spin resonance (ESR) spectroscopy. Inclusion of MGD-Fe2+ (50 mumol l-1 Fe2+ and 500 mumol l-1 MGD) in the cardioplegic solution along with GSNO-MEE yielded an ESR signal characteristic of the MGD-Fe2+ -NO adduct. However, the MGD-Fe2+ trap did not prevent the formation of the intracellular MbNO complex in myocardial tissue. During aerobic reperfusion, denitrosylation of the MbNO complex slowly occurred as shown by the decrease in ESR spectral intensity. GSNO-MEE treatment did not affect ubisemiquinone radical formation during reperfusion. 5. GSNO-MEE (20 microliters l-1) treatment elevated the myocardial cyclic GMP during ischaemia (47 +/- 3 in control vs. 153 +/- 34 pmol g-1 dry wt. after 35 min ischaemia, P < 0.05). The cyclic GMP levels decreased in the control group during ischaemia from 100 +/- 6 after induction of cardioplegia to 47 +/- 3 pmol g-1 dry wt. at the end of ischaemic duration. 6. Glycogen levels were lower in GSNO-MEE (20 mumol l-1)-treated hearts throughout the ischaemic duration (26.7 +/- 3.1 in control vs. 19.7 +/- 2.4 mumol g dry-t wt. in GSNO-MEE-treated group at the end of ischaemic duration), because of rapid depletion of glycogen during induction of cardioplegia. During ischaemia, the amounts of glycogen consumed in both groups were similar. Equivalent amounts of lactate were produced in both groups (148 +/- 4 in control vs. 141 +/- 4 mumol g-1 dry wt. in GSNO-MEE-treated group after 35 min in ischaemia). 7. The mechanism(s) of myocardial protection by GSNO-MEE against ischaemic injury may involve preischaemic glycogen reduction and/or elevated cyclic GMP levels in myocardial tissue during ischaemia.


Subject(s)
Cyclic GMP/metabolism , Glutathione/analogs & derivatives , Glycogen/metabolism , Heart Arrest, Induced , Heart/drug effects , Nitroso Compounds/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Analysis of Variance , Animals , Electron Spin Resonance Spectroscopy , Glutathione/pharmacology , Heart/physiopathology , Male , Myocardial Contraction/drug effects , Myocardial Reperfusion , Nitric Oxide/metabolism , Rats , S-Nitrosoglutathione
16.
Proc Natl Acad Sci U S A ; 93(16): 8745-9, 1996 Aug 06.
Article in English | MEDLINE | ID: mdl-8710942

ABSTRACT

Reactive oxygen species play a central role in vascular inflammation and atherogenesis, with enhanced superoxide (O2.-) production contributing significantly to impairment of nitric oxide (.NO)-dependent relaxation of vessels from cholesterol-fed rabbits. We investigated potential sources of O2.- production, which contribute to this loss of endothelium-dependent vascular responses. The vasorelaxation elicited by acetylcholine (ACh) in phenylephrine-contracted, aortic ring segments was impaired by cholesterol feeding. Pretreatment of aortic vessels with either heparin, which competes with xanthine oxidase (XO) for binding to sulfated glycosaminoglycans, or the XO inhibitor allopurinol resulted in a partial restoration (36-40% at 1 muM ACh) of ACh-dependent relaxation. Furthermore, O2.(-)-dependent lucigenin chemiluminescence, measured in intact ring segments from hypercholesterolemic rabbits, was decreased by addition of heparin, allopurinol or a chimeric, heparin-binding superoxide dismutase. XO activity was elevated more than two-fold in plasma of hypercholesterolemic rabbits. Incubation of vascular rings from rabbits on a normal diet with purified XO (10 milliunits/ml) also impaired .NO-dependent relaxation but only in the presence of purine substrate. As with vessels from hypercholesterolemic rabbits, this effect was prevented by heparin and allopurinol treatment. We hypothesize that increases in plasma cholesterol induce the release of XO into the circulation, where it binds to endothelial cell glycosaminoglycans. Only in hypercholesterolemic vessels is sufficient substrate available to sustain the production of O2.- and impair NO-dependent vasorelaxation. Chronically, the continued production of peroxynitrite, (ONOO-) which the simultaneous generation of NO and O2.- implies, may irreversibly impair vessel function.


Subject(s)
Endothelium, Vascular/metabolism , Glycosaminoglycans/metabolism , Hypercholesterolemia/enzymology , Xanthine Oxidase/blood , Acetylcholine/pharmacology , Animals , Hypercholesterolemia/blood , Hypercholesterolemia/physiopathology , In Vitro Techniques , Luminescent Measurements , Muscle, Smooth, Vascular/physiology , Nitric Oxide/physiology , Rabbits , Superoxides/metabolism , Vasodilation/physiology
17.
J Clin Invest ; 97(8): 1916-23, 1996 Apr 15.
Article in English | MEDLINE | ID: mdl-8621776

ABSTRACT

We tested the hypothesis that angiotensin II-induced hypertension is associated with an increase in vascular .O2- production, and characterized the oxidase involved in this process. Infusion of angiotensin II (0.7 mg/kg per d) increased systolic blood pressure and doubled vascular .O2- production (assessed by lucigenin chemiluminescence), predominantly from the vascular media. NE infusion (2.75 mg/kg per d) produced a similar degree of hypertension, but did not increase vascular .O2- production. Studies using various enzyme inhibitors and vascular homogenates suggested that the predominant source of .O2- activated by angiotensin II infusion is an NADH/NADPH-dependent, membrane-bound oxidase. Angiotensin II-, but not NE-, induced hypertension was associated with impaired relaxations to acetylcholine, the calcium ionophore A23187, and nitroglycerin. These relaxations were variably corrected by treatment of vessels with liposome-encapsulated superoxide dismutase. When Losartan was administered concomitantly with angiotensin II, vascular .O2- production and relaxations were normalized, demonstrating a role for the angiotensin type-1 receptor in these processes. We conclude that forms of hypertension associated with elevated circulating levels of angiotensin II may have unique vascular effects not shared by other forms of hypertension because they increase vascular smooth muscle .O2- production via NADH/NADPH oxidase activation.


Subject(s)
Angiotensin II/pharmacology , Hypertension/physiopathology , Muscle Tonus/physiology , Muscle, Smooth, Vascular/physiopathology , NADH, NADPH Oxidoreductases/metabolism , Superoxides/metabolism , Acetylcholine/pharmacology , Animals , Antihypertensive Agents/pharmacology , Aorta , Arginine/analogs & derivatives , Arginine/pharmacology , Biphenyl Compounds/pharmacology , Calcimycin/pharmacology , Endothelium, Vascular/physiology , Enzyme Activation , Enzyme Inhibitors/pharmacology , Hypertension/chemically induced , Imidazoles/pharmacology , In Vitro Techniques , Losartan , Male , Muscle Relaxation/drug effects , Muscle Tonus/drug effects , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Nitric Oxide Synthase/antagonists & inhibitors , Nitroglycerin/pharmacology , Norepinephrine/pharmacology , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/pharmacology , Tetrazoles/pharmacology , omega-N-Methylarginine
18.
J Pharmacol Exp Ther ; 274(1): 200-6, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7616400

ABSTRACT

The objective of this study was to assess the cardioprotective effect of the nitric oxide (.NO) donor, S-nitrosoglutathione (GSNO) and to investigate the mechanism of cardioprotection in a model of ischemia and reperfusion in isolated rat hearts. The role of .NO in myocardial protection was investigated by using nitronyl nitroxide as the .NO trap. Electron spin resonance spectroscopy was used to demonstrate that nitronyl nitroxide can trap .NO released from GSNO in a cardioplegic solution. .NO traps, oxyhemoglobin (4 mumol/l, n = 4) and nitronyl nitroxide (400 mumol/l, n = 5), inhibited the (2 mumol/l) GSNO-induced coronary vasodilation from the control value of 122% (n = 6) above base-line value to 73 and 60%, respectively. In the ischemia-reperfusion protocol, GSNO (20 mumol/l) was added to the cardioplegic solution during a 35-min ischemic arrest (n = 8). GSNO improved the functional recovery of ischemic hearts as compared to control (n = 6) as measured by the developed pressure (76 +/- 3 to 95 +/- 5% of base-line), rate pressure product (68 +/- 3 to 83 +/- 4% of base-line) and diastolic pressure (31 +/- 2 to 19 +/- 3 mm Hg). Reduction of coronary flow rate during reperfusion to control values in GSNO-treated hearts did not eliminate the improvement of functional recovery induced by GSNO. GSNO increased cyclic GMP production and slowed the accumulation of lactate (154 +/- 7 in control to 114 +/- 4 mumol/g dry wt.) and glucose-6-phosphate (3.66 +/- 0.19 in control to 2.18 +/- 0.10 mumol/g dry wt.) in myocardial tissue during ischemia.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Glutathione/analogs & derivatives , Heart Arrest/physiopathology , Heart/drug effects , Myocardial Ischemia/physiopathology , Nitric Oxide/physiology , Nitroso Compounds/pharmacology , Animals , Cyclic GMP/metabolism , Glutathione/chemistry , Glutathione/metabolism , Glutathione/pharmacology , Glycolysis , Heart/physiopathology , In Vitro Techniques , Male , Myocardium/metabolism , Nitroso Compounds/chemistry , Nitroso Compounds/metabolism , Rats , Rats, Sprague-Dawley , S-Nitrosoglutathione , Spin Labels , Vasodilator Agents/chemistry , Vasodilator Agents/metabolism , Vasodilator Agents/pharmacology
19.
FEBS Lett ; 364(3): 314-8, 1995 May 15.
Article in English | MEDLINE | ID: mdl-7758588

ABSTRACT

Peroxynitrite stimulated the synthesis of cyclic GMP by rat aortic smooth muscle in a time- and dose-dependent manner. Peak formation of cyclic GMP occurred at 1 min with 100 microM peroxynitrite and was inhibited by oxyhemoglobin. Peroxynitrite was less potent than nitric oxide in stimulating cyclic GMP synthesis. Peroxynitrite also enhanced endothelial-dependent cyclic GMP synthesis, via generation of a long-lived substance, which was prevented by inhibition of glutathione synthesis. These data show that peroxynitrite stimulates cyclic GMP synthesis, inferring production of low yields of nitric oxide or associated derivatives. Additionally, vascular exposure to peroxynitrite potentiates endothelial-dependent activation of guanylate cyclase.


Subject(s)
Cyclic GMP/biosynthesis , Muscle, Smooth, Vascular/metabolism , Nitrates/pharmacology , Animals , Aorta , Cattle , Cells, Cultured , Cyclic GMP/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Enzyme Activation , Glutathione/biosynthesis , Guanylate Cyclase/metabolism , Kinetics , Muscle, Smooth, Vascular/drug effects , Nitric Oxide/pharmacology , Oxyhemoglobins/pharmacology , Rats
20.
Free Radic Biol Med ; 18(2): 169-77, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7744299

ABSTRACT

Nitronyl nitroxides have been used to trap nitric oxide (.NO) produced during visible irradiation of nitrovasodilators such as sodium nitroprusside (Joseph et al., Biochem. Biophys. Res. Commun. 192:926-934; 1993). We have also shown that nitrone and nitroso spin traps exert a potent vasodilatory effect in the isolated perfused rat heart (Konorev et al., Free Radic. Biol. Med. 14:127-137, 1993). The objective of this study was to investigate the effect of nitronyl nitroxides on the vasodilatory action of sodium nitroprusside (SNP), S-nitroso-N-acetylpenicillamine (SNAP), alpha-(4-pyridyl-1-oxide)-N-tert-butyl nitrone (POBN) and 4-hydroxy-2,2,6,6-tetramethylpiperidinyloxy free radical (TEMPOL) in the isolated perfused rat heart model. In this study, we have used the following nitronyl nitroxides as nitric oxide traps: 2-(p-carboxyphenyl)-4,4,5,5-tetramethyl imidazoline-3-oxide 1-oxyl (SLI) and 2(1',1'-dimethyl-2'-hydroxyethyl)-4,4,5,5-tetramethyl imidazoline-3-oxide 1-oxyl (SLII). Under in vitro conditions, both SLI and SLII trapped .NO released from SNP/light treatment and from spontaneous decomposition of SNAP, forming the corresponding imino nitroxides, which were characterized by electron spin resonance (ESR) technique. In isolated hearts, SNP (2 mumol/l) and SNAP (20 mumol/l) increased coronary flow rate to a maximum of 185% and 190%, respectively. SNP-induced vasodilation was inhibited by SLI (0.05-3 mmol/l) from 162% to 131% of baseline, and SNAP-induced vasodilation was inhibited by SLII (0.05-1.2 mmol/l) from 190% to 136% of baseline. In contrast, neither SLI nor SLII inhibited the vasodilatory action elicited by POBN or TEMPOL.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Benzoates , Cyclic N-Oxides/pharmacology , Nitric Oxide/metabolism , Spin Labels , Vasodilator Agents/pharmacology , Animals , Coronary Circulation/drug effects , Electron Spin Resonance Spectroscopy , Imidazoles/pharmacology , Male , Nitrogen Oxides/pharmacology , Nitroprusside/pharmacology , Penicillamine/analogs & derivatives , Penicillamine/pharmacology , Pyridines , Rats , Rats, Sprague-Dawley , S-Nitroso-N-Acetylpenicillamine , Vasodilation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...