Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
Add more filters










Publication year range
1.
Am J Physiol Cell Physiol ; 294(4): C1012-20, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18287333

ABSTRACT

We have shown previously that extracellular cysteine is necessary for cellular responses to S-nitrosoalbumin. In this study we have investigated mechanisms involved in accumulation of extracellular cysteine outside vascular smooth muscle cells and characterized the role of cystine-cysteine release in transfer of nitric oxide (NO)-bioactivity. Incubation of cells with cystine led to cystine uptake, reduction, and cysteine release. The process was inhibitable by extracellular glutamate, suggesting a role for system x(c)(-) amino acid transporters. Smooth muscle cells express this transporter constitutively and induction of the light chain component (xCT) by either diethyl maleate or 3-morpholino-sydnonimine (SIN-1) led to glutamate-inhibitable cystine uptake and an increased rate of cysteine release from cells. Likewise, overexpression of xCT in smooth muscle cells or endothelial cells led to glutamate-inhibitable cysteine release. The resulting extracellular cysteine was found to be required for transfer of NO from extracellular S-nitrosothiols into cells via system L transporters leading to formation of cellular S-nitrosothiols. Cysteine release coupled to cystine uptake was also found to be required for cellular responses to S-nitrosoalbumin and facilitated S-nitrosoalbumin-mediated inhibition of epidermal growth factor signaling. These data show that xCT expression can constitute a cystine-cysteine shuttle whereby cystine uptake drives cysteine release. Furthermore, we show that extracellular cysteine provided by this shuttle mechanism is necessary for transfer of NO equivalents and cellular responses to S-nitrosoablumin.


Subject(s)
Amino Acid Transport System y+/metabolism , Cysteine/metabolism , Cystine/metabolism , Myocytes, Smooth Muscle/drug effects , Nitroso Compounds/pharmacology , Serum Albumin, Bovine/pharmacology , Amino Acid Transport System y+/genetics , Animals , Humans , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Rats
2.
Biochem Biophys Res Commun ; 307(4): 973-9, 2003 Aug 08.
Article in English | MEDLINE | ID: mdl-12878207

ABSTRACT

Vascular endothelial cells respond to nitric oxide by activating MAPK pathways and upregulating stress-activated proteins such as gamma-glutamylcysteine synthetase (gamma-GCS) and heme oxygenase-1 (HO-1). Since consensus sequences for the antioxidant response element (ARE) are found in the promoters of the gamma-GCS and HO-1 genes, we examined nuclear translocation of Nrf2, a CNC-bZIP protein which binds to and activates the ARE. We found a dramatic increase in Nrf2 nuclear translocation 1-8h following the nitric oxide donor spermine NONOate. Translocation was inhibited by pretreatment of cells with N-acetylcysteine suggesting involvement of an oxidative mechanism in this response. Translocation was also blocked by PD 98059 and SB 203580, inhibitors of ERK and p38 pathways, respectively. In addition to effects on Nrf2 subcellular localization, spermine NONOate increased Nrf2 protein levels by a mechanism which was inhibited by PD 98059. Pretreatment with N-acetylcysteine, PD 98059, and SB 203580 decreased HO-1 upregulation in spermine NONOate-treated cells. These results suggest that ERK and p38 pathways may regulate nitric oxide-mediated adaptive responses in vascular endothelium via translocation of Nrf2 and activation of the ARE.


Subject(s)
Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Endothelium, Vascular/metabolism , Nitric Oxide Donors/pharmacology , Spermine/analogs & derivatives , Spermine/pharmacology , Trans-Activators/metabolism , Active Transport, Cell Nucleus/drug effects , Animals , Cattle , Cells, Cultured , Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Enzyme Inhibitors/pharmacology , Heme Oxygenase (Decyclizing)/biosynthesis , Heme Oxygenase-1 , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , NF-E2-Related Factor 2 , Nitric Oxide Donors/antagonists & inhibitors , Nitrogen Oxides , Spermine/antagonists & inhibitors , Up-Regulation
3.
Brain Res ; 917(2): 253-61, 2001 Nov 02.
Article in English | MEDLINE | ID: mdl-11640911

ABSTRACT

Central nervous system oxygen toxicity (CNS O2 toxicity) is preceded by release of hyperoxic vasoconstriction, which increases regional cerebral blood flow (rCBF). These increases in rCBF precede the onset of O2-induced convulsions. We have tested the hypothesis that hyperbaric oxygen (HBO2) stimulates NO* production in the brain that leads to hyperemia and anticipates electrical signs of neurotoxicity. We measured rCBF and EEG responses in rats exposed at 4 to 6 atmospheres (ATA) of HBO2 and correlated them with brain interstitial NO* metabolites (NO(x)) as an index of NO* production. During exposures to hyperbaric oxygen rCBF decreased at 4 ATA, decreased for the initial 30 min at 5 ATA then gradually increased, and increased within 30 min at 6 ATA. Changes in rCBF correlated positively with NO(x) production; increases in rCBF during HBO2 exposure were associated with large increases in NO(x) at 5 and 6 ATA and always preceded EEG discharges as a sign of CNS O2 toxicity. In rats pretreated with L-NAME, rCBF remained maximally decreased throughout 75 min of HBO2 at 4, 5 and 6 ATA. These data provide the first direct evidence that increased NO* production during prolonged HBO2 exposure is responsible for escape from hyperoxic vasoconstriction. The finding suggests that NO* overproduction initiates CNS O2 toxicity by increasing rCBF, which allows excessive O2 to be delivered to the brain.


Subject(s)
Medulla Oblongata/cytology , Neurons/metabolism , Receptors, Opioid, mu/metabolism , Reticular Formation/metabolism , Spinal Cord/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Immunologic Techniques , Male , Microscopy, Electron , Nerve Endings/metabolism , Nerve Endings/physiology , Neurons/physiology , Neurons/ultrastructure , Rats , Rats, Sprague-Dawley , Reticular Formation/cytology , Reticular Formation/physiology , Reticular Formation/ultrastructure , Spinal Cord/cytology , Spinal Cord/physiology , Spinal Cord/ultrastructure
4.
Am J Physiol Cell Physiol ; 280(3): C659-76, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11171586

ABSTRACT

The transcription factor nuclear factor-kappaB (NF-kappaB) is constitutively activated in malignancies from enhanced activity of inhibitor of NF-kappaB (IkappaB) kinase, with accelerated IkappaBalpha degradation. We studied whether redox signaling might stimulate these events. Cultured melanoma cells generated superoxide anions (O(2)(-)) without serum stimulation. O(2)(-) generation was reduced by the NAD(P)H:quinone oxidoreductase (NQO) inhibitor dicumarol and the quinone analog capsaicin, suggesting that electron transfer from NQO through a quinone-mediated pathway may be an important source of endogenous reactive oxygen species (ROS) in tumor cells. Treatment of malignant melanoma cells with the H(2)O(2) scavenger catalase, the sulfhydryl donor N-acetylcysteine, the glutathione peroxidase mimetic ebselen, or dicumarol decreased NF-kappaB activation. Catalase, N-acetylcysteine, ebselen, dicumarol, and capsaicin also inhibited growth of melanoma and other malignant cell lines. These results raise the possibility that ROS produced endogenously by mechanisms involving NQO can constitutively activate NF-kappaB in an autocrine fashion and suggest the potential for new antioxidant strategies for interruption of oxidant signaling of melanoma cell growth.


Subject(s)
Melanoma/metabolism , NADH, NADPH Oxidoreductases/metabolism , NF-kappa B/physiology , Reactive Oxygen Species/physiology , Antioxidants/pharmacology , Capsaicin/pharmacology , Cell Division/drug effects , Dicumarol/pharmacology , Enzyme Inhibitors/pharmacology , Humans , Melanoma/pathology , NADP/physiology , NF-kappa B/drug effects , Ploidies , S Phase/drug effects , Tumor Cells, Cultured
5.
Am J Physiol Cell Physiol ; 279(4): C1168-76, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11003597

ABSTRACT

We and others recently demonstrated increased glutathione levels, stimulated cystine uptake, and induced gamma-glutamylcysteinyl synthase (gamma-GCS) in vascular cells exposed to nitric oxide donors. Here we report the effects of peroxynitrite on glutathione levels and cystine uptake. Treatment of bovine aortic endothelial and smooth muscle cells with 3-morpholinosydnonimine (SIN-1), a peroxynitrite donor, resulted in transient depletion of glutathione followed by a prolonged increase beginning at 8-9 h. Concentration-dependent increases in glutathione of up to sixfold occurred 16-18 h after 0.05-2.5 mM SIN-1. Responses to SIN-1 were inhibited by copper-zinc superoxide dismutases and manganese(III)tetrakis(1-methyl-4-pyridyl)porphyrin pentachloride, providing evidence for peroxynitrite involvement. Because glutathione synthesis is regulated by amino acid availability, we also studied cystine uptake. SIN-1 treatment resulted in a prolonged increase in cystine uptake beginning at 6-9 h. Increases in cystine uptake after SIN-1 were blocked by inhibitors of protein and RNA synthesis, by extracellular glutamate but not by extracellular sodium. These studies suggest induction of the x(c)(-) pathway of amino acid uptake. A close correlation over time was observed for increases in cystine uptake and glutathione levels. In summary, vascular cells respond to chronic peroxynitrite exposure with adaptive increases in cellular glutathione and cystine transport.


Subject(s)
Cystine/pharmacokinetics , Endothelium, Vascular/metabolism , Glutathione/metabolism , Molsidomine/analogs & derivatives , Muscle, Smooth, Vascular/metabolism , Nitrates/pharmacology , Penicillamine/analogs & derivatives , Adaptation, Physiological/physiology , Animals , Cattle , Cells, Cultured , Cystine/metabolism , Dose-Response Relationship, Drug , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Free Radical Scavengers/pharmacology , Glutamic Acid/metabolism , Glutamic Acid/pharmacology , Metalloporphyrins/pharmacology , Molsidomine/pharmacology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Nitric Oxide Donors/pharmacology , Nucleic Acid Synthesis Inhibitors/pharmacology , Penicillamine/pharmacology , Protein Synthesis Inhibitors/pharmacology , S-Nitroso-N-Acetylpenicillamine , Sodium/metabolism , Sodium/pharmacology , Superoxide Dismutase/metabolism
6.
Nitric Oxide ; 4(6): 597-608, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11139368

ABSTRACT

Based on recent evidence that nitric oxide (NO(.)) is involved in hyperoxic vasoconstriction, we tested the hypothesis that decreases in NO(.) availability in brain tissue during hyperbaric oxygen (HBO(2)) exposure contribute to decreases in regional cerebral blood flow (rCBF). rCBF was measured in rats exposed to HBO(2) at 5 atmospheres (ATA) and correlated with interstitial brain levels of NO(.) metabolites (NO(X)) and production of hydroxyl radical ((.)OH). Changes in rCBF were also correlated with the effects of NO(.) synthase inhibitor (l-NAME), NO(.) donor PAPANONOate, and intravascular superoxide dismutase (MnSOD) during HBO(2). After 30 min of O(2) exposure at 5 ATA, rCBF had decreased in the substantia nigra, caudate putamen, hippocampus, and parietal cortex by 23 to 37%. These reductions in rCBF were not augmented by exposure to HBO(2) in animals pre-treated with l-NAME. After 30 min at 5 ATA, brain NO(X) levels had decreased by 31 +/- 9% and correlated with the decrease in rCBF, while estimated (.)OH production increased by 56 +/- 8%. The decrease in rCBF at 5 ATA was completely abolished by MnSOD administration into the circulation before HBO(2) exposure. Doses of NO(.) donor that significantly increased rCBF in animals breathing air had no effect at 5 ATA of HBO(2). These results indicate that decreases in rCBF with HBO(2) are associated with a decrease in effective NO(.) concentration and an increase in ROS production in the brain. The data support the hypothesis that inactivation of NO(.) antagonizes basal relaxation of cerebral vessels during HBO(2) exposure, although an effect of HBO(2) on NO(.) synthesis has not been excluded.


Subject(s)
Cerebrovascular Circulation/physiology , Hyperbaric Oxygenation , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/metabolism , Animals , Blood Flow Velocity/drug effects , Blood Gas Analysis , Blood Pressure/drug effects , Cerebrovascular Circulation/drug effects , Corpus Striatum/metabolism , Electroencephalography , Enzyme Inhibitors/pharmacology , Hydrazines/administration & dosage , Hydroxybenzoates/metabolism , Hydroxyl Radical/metabolism , Injections, Intravenous , Microdialysis , NG-Nitroarginine Methyl Ester/administration & dosage , Nitrates/metabolism , Nitric Oxide/administration & dosage , Nitrites/metabolism , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/administration & dosage
7.
J Biol Chem ; 274(28): 20017-26, 1999 Jul 09.
Article in English | MEDLINE | ID: mdl-10391952

ABSTRACT

Reactive oxygen species have been recently identified as important mediators of mitogenic signaling in a number of cell types. We therefore explored their role in mediating mitogenesis of airway smooth muscle. The antioxidants catalase, N-acetylcysteine, and probucol significantly reduced proliferation in primary cultures of rat tracheal smooth muscle stimulated with fetal bovine serum or platelet-derived growth factor, without affecting cell viability or inducing apoptosis. N-Acetylcysteine also significantly reduced serum-stimulated elevation of c-Fos but did not prevent the normal mitogen-induced increase in c-fos mRNA. Fractionation of ribosomes by sucrose density centrifugation and subsequent dot-blot Northern analysis revealed that antioxidants reduced incorporation of c-fos mRNA into the heaviest polyribosomes, suggesting redox regulation of c-fos mRNA translation. Serum treatment of monolayers produced a small but reproducibly significant rise in superoxide dismutase-inhibitable reduction of ferricytochrome c by myocyte monolayers. Serum-induced ferricytochrome c reduction, cellular proliferation, and c-Fos elevation were decreased by the flavoprotein-dependent enzyme inhibitor dipheyleneiodonium. Growth responses to fetal bovine serum and superoxide dismutase-inhibitable reduction of ferricytochrome c were not different between cultured tracheal myocytes from wild-type versus gp91 phagocyte oxidase null mice. These results suggest that mitogen stimulation of airway smooth muscle induces signal transduction of cell proliferation that is in part dependent on generation of partially reduced oxygen species, generated by an NADH or NADPH oxidoreductase that is different from the oxidase in phagocytic cells.


Subject(s)
Muscle, Smooth/drug effects , NADPH Oxidases , Platelet-Derived Growth Factor/pharmacology , Reactive Oxygen Species/physiology , Animals , Antioxidants/pharmacology , Cell Division , Cells, Cultured , Male , Membrane Glycoproteins/genetics , Mice , Mice, Transgenic , Mitogens/pharmacology , Muscle Development , Muscle, Smooth/growth & development , NADPH Oxidase 2 , Nitric Oxide Synthase/metabolism , Onium Compounds/pharmacology , Rats , Rats, Sprague-Dawley , Trachea/drug effects , Xanthine Oxidase/metabolism
8.
Am J Physiol ; 276(4): C803-11, 1999 04.
Article in English | MEDLINE | ID: mdl-10199810

ABSTRACT

Nitric oxide (NO) is known to produce some of its biological activity through modification of cellular thiols. Return of cellular thiols to their basal state requires the activity of the GSH redox cycle, suggesting important interactions between NO signaling and regulation of cellular redox status. Because continuous exposure to NO may lead to adaptive responses in cellular redox systems, we investigated the effects of NO on cellular GSH levels in vascular endothelial cells. Acute exposure (1 h) of cells to >1 mM S-nitroso-N-acetyl-penicillamine (SNAP) led to depletion of GSH. On the other hand, chronic exposure to lower concentrations of SNAP (

Subject(s)
Cystine/metabolism , Endothelium, Vascular/metabolism , Glutathione/metabolism , Nitric Oxide/pharmacology , Penicillamine/analogs & derivatives , Animals , Aorta , Biological Transport/drug effects , Cattle , Cells, Cultured , Cycloheximide/pharmacology , Cytosol/metabolism , Dactinomycin/pharmacology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Glutamic Acid/pharmacology , Kinetics , Penicillamine/pharmacology , S-Nitroso-N-Acetylpenicillamine , Time Factors
9.
Proc Natl Acad Sci U S A ; 96(8): 4686-91, 1999 Apr 13.
Article in English | MEDLINE | ID: mdl-10200323

ABSTRACT

Estrogens and growth factors such as epidermal growth factor (EGF) act as mitogens promoting cellular proliferation in the breast and in the reproductive tract. Although it was considered originally that these agents manifested their mitogenic actions through separate pathways, there is a growing body of evidence suggesting that the EGF and estrogen-mediated signaling pathways are intertwined. Indeed, it has been demonstrated recently that 17beta-estradiol (E2) can induce a rapid activation of mitogen-activated protein kinase (MAPK) in mammalian cells, an event that is independent of both transcription and protein synthesis. In this study, we have used a pharmacological approach to dissect this novel pathway in MCF-7 breast cancer cells and have determined that in the presence of endogenous estrogen receptor, activation of MAPK by E2 is preceded by a rapid increase in cytosolic calcium. The involvement of intracellular calcium in this process was supported by the finding that the presence of EGTA and Ca2+-free medium did not affect the activation of MAPK by E2 and, additionally, that this response was blocked by the addition of the intracellular calcium chelator 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetate. Cumulatively, these data indicate that the estrogen receptor, in addition to functioning as a transcription factor, is also involved, through a nongenomic mechanism, in the regulation of both intracellular calcium homeostasis and MAPK-signaling pathways. Although nongenomic actions of estrogens have been suggested by numerous studies in the past, the ability to link estradiol and the estrogen receptor to a well defined signaling pathway strongly supports a physiological role for this activity.


Subject(s)
Calcimycin/pharmacology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Calcium/metabolism , Estradiol/pharmacology , Mitogen-Activated Protein Kinases , Breast Neoplasms , Cell Nucleus/enzymology , Cytoplasm/enzymology , Egtazic Acid/pharmacology , Enzyme Activation/drug effects , Epidermal Growth Factor/pharmacology , Estradiol/analogs & derivatives , Estradiol/physiology , Estrogen Antagonists/pharmacology , Female , Fulvestrant , HeLa Cells , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Recombinant Proteins/pharmacology , Tumor Cells, Cultured
10.
Arch Biochem Biophys ; 358(2): 232-42, 1998 Oct 15.
Article in English | MEDLINE | ID: mdl-9784235

ABSTRACT

Nitrosothiols, the product of the reaction of nitric oxide-derived species (NOx) with thiols, participate in both cell signaling and cytotoxic events. Glutathione has recently been shown to modulate nitrosothiol-mediated signal transduction and to protect against NOx-mediated cytotoxicity. We have investigated the role of protein S-thiolation/dethiolation as a potential mechanism by which glutathione regulates nitrosothiol signaling and toxicity. Our data show that exogenous sources of NOx decreased both free protein thiol and total glutathione levels in endothelial cells. The decrease in glutathione levels could not be accounted for by formation of S-nitrosoglutathione (GSNO) since borohydride treatment of the nonprotein fraction of cell extracts did not restore glutathione levels, whereas borohydride treatment of protein-containing cell extracts led to recovery of glutathione levels. The NOx-mediated decrease in glutathione and protein thiol content was correlated with an increase in protein mixed disulfide formation, as measured by the incorporation of [35S]glutathione into cellular proteins. [35S]glutathione was incorporated into proteins via a covalent disulfide bond since dithiothreitol removed the radiolabel from cellular proteins. The withdrawal of the exogenous NOx source led to recovery of free protein thiol and cellular glutathione levels, which correlated with the dethiolation of proteins. Dethiolation required the action of the glutathione redox system since 1, 3-bis(2-chloroethyl)-1-nitrosourea, an inhibitor of glutathione reductase, blocked both the recovery of glutathione levels and the dethiolation of proteins. These results suggest that exposure of cells to NOx does not lead to accumulation of GSNO but rather stimulates protein S-thiolation, a mechanism which may have important implications with respect to nitrosothiol signaling and toxicity.


Subject(s)
Glutathione/metabolism , Nitric Oxide/physiology , Sulfhydryl Compounds/metabolism , Animals , Aorta , Cattle , Endothelium, Vascular , Glutathione/analogs & derivatives , Glutathione Reductase/metabolism , Nitric Oxide Donors/metabolism , Nitroso Compounds/metabolism , Oxidation-Reduction , S-Nitrosoglutathione
11.
J Pharmacol Exp Ther ; 285(2): 876-83, 1998 May.
Article in English | MEDLINE | ID: mdl-9580639

ABSTRACT

The adrenal steroid dehydroepiandrosterone (DHEA) and its analogs reduce growth of immortalized and malignant cell lines. We therefore explored their effects on the growth of airway smooth muscle, whose hyperplasia may lead to fixed airways obstruction and enhanced airways hyperresponsiveness in severe chronic asthma. DHEA and its potent analog 16 alpha-bromoepiandrosterone dramatically reduced proliferation in primary cultures of rat tracheal smooth muscle stimulated with fetal bovine serum or platelet-derived growth factor. Growth inhibition was dose-dependent and could not be attributed to interference with glucose-6-phosphate dehydrogenase activity or cholesterol metabolism, as reported for immortalized or malignant cell lines, respectively. Expression of the early response gene c-fos remained intact, but DHEA and 16 alpha-bromoepiandrosterone decreased DNA binding of the transcription factor activator protein-1, a later response important for expression of genes that mediate DNA synthesis and cell cycle progression. These results suggest that the nonglucocorticoid steroid DHEA and its analogs may impair activation of secondary growth response genes in a fashion analogous to that reported for glucocorticoids and that they may prove useful for treatment of asthmatic airway remodeling in the human.


Subject(s)
Androsterone/analogs & derivatives , DNA/metabolism , Dehydroepiandrosterone/pharmacology , Muscle, Smooth/drug effects , Trachea/drug effects , Transcription Factor AP-1/metabolism , Androsterone/pharmacology , Animals , Cell Division/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Male , Muscle, Smooth/cytology , Rats , Rats, Sprague-Dawley , Trachea/cytology
12.
Shock ; 9(1): 40-5, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9466472

ABSTRACT

We hypothesized that endothelin (ET) may be released in response to tumor necrosis factor-alpha (TNF) and that platelet-activating factor (PAF) and cyclooxygenase products modulate TNF-induced ET-1 release in vivo. Anesthetized and instrumented pigs were randomly assigned to receive: 1) saline + TNF (n = 8); 2) saline + heat-inactivated TNF (control group, n = 5); 3) WEB 2086 (PAF receptor antagonist) + TNF (n = 7); or 4) indomethacin + TNF (n = 6). Infusion of TNF was associated with increases in mean aortic, mean pulmonary artery, and intratracheal pressures, increases in systemic and pulmonary vascular resistances, and elevated plasma thromboxane B2 concentration. Plasma ET-1 concentrations were unchanged in controls and significantly increased in TNF-treated pigs at 2 to 4 h. WEB 2086 did not modify plasma levels of ET-1 during exogenous infusion of TNF. In contrast, the cyclooxygenase inhibitor, indomethacin, mildly, but not significantly, reduced plasma ET-1 levels. In addition, indomethacin (but not WEB 2086) blocked or attenuated the TNF-induced increases in mean aortic pressure, systemic vascular resistance, mean pulmonary artery pressure, pulmonary vascular resistance, and intratracheal pressure. We conclude that in the pig, cyclooxygenase products modulate some of the cardiovascular responses to TNF and may mildly affect ET-1 biosynthesis. On the other hand, PAF neither significantly influences TNF-induced biosynthesis of ET-1 nor its associated cardiovascular responses.


Subject(s)
Arachidonic Acid/metabolism , Endothelin-1/metabolism , Lipids/physiology , Platelet Activating Factor/physiology , Prostaglandin-Endoperoxide Synthases/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Endothelin-1/biosynthesis , Heart/drug effects , Lung/drug effects , Recombinant Proteins/pharmacology , Swine , Thromboxane B2/biosynthesis
13.
Am J Physiol ; 273(4): C1298-305, 1997 10.
Article in English | MEDLINE | ID: mdl-9357774

ABSTRACT

Tunicamycin is a nucleoside antibiotic that inhibits protein glycosylation and palmitoylation. The therapeutic use of tunicamycin is limited in animals because of its toxic effects, particularly in cerebral vasculature. Tunicamycin decreases palmitoylation of the endothelial isoform of nitric oxide synthase, stimulates nitric oxide synthesis, and increases the concentration of intracellular calcium ([Ca2+]i) in bovine aortic endothelial cells (B. J. Buckley and A. R. Whorton. FASEB J. 11: A110, 1997). In the present study, we investigated the mechanism by which tunicamycin alters [Ca2+]i using the Ca2+-sensitive dye fura 2. We found that tunicamycin increased [Ca2+]i without increasing levels of inositol phosphates. When cells were incubated in the absence of extracellular Ca2+, [Ca2+]i rapidly rose in response to tunicamycin, although a full response was not achieved. The pool of intracellular Ca2+ mobilized by tunicamycin overlapped with that mobilized by thapsigargin. Extracellular nickel blocked a full response to tunicamycin when cells were incubated in the presence of extracellular Ca2+. The effects of tunicamycin on [Ca2+]i were partially reversed by washing out the drug, and the remainder of the response was inhibited by removing extracellular Ca2+. These results indicate that tunicamycin mobilizes Ca2+ from intracellular stores in a manner independent of phospholipase C activation and increases the influx of Ca2+ across the plasma membrane.


Subject(s)
Calcium/metabolism , Endothelium, Vascular/drug effects , Tunicamycin/pharmacology , Animals , Aorta , Bradykinin/pharmacology , Calcium/pharmacology , Cattle , Cells, Cultured , Egtazic Acid/pharmacology , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Inositol Phosphates/metabolism , Kinetics , Nitric Oxide Synthase/metabolism , Palmitic Acid/metabolism , Thapsigargin/pharmacology , Time Factors
14.
Am J Physiol ; 272(6 Pt 1): L1161-6, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9227518

ABSTRACT

Vascular endothelial cells synthesize nitric oxide (NO) in response to agonists that elevate cytosolic free Ca2+ concentrations. Once activated, NO synthase (NOS) requires arginine, NADPH, and O2 as cosubstrates. In this study, we investigated the role of O2 in regulating endothelial NOS activity in intact bovine aortic endothelial cells by measuring the rate of nitrite (NO2-) and nitrate (NO3-) production after conversion of NO2- to S-nitrosoglutathione before analysis or after reduction of NO2- and NO3- to NO using acidic vanadium chloride. The basal rate of NO2- production was 6.5 +/- 0.8 pmol.min-1.mg protein-1. Thapsigargin (TG, 1 microM) elevated free cytosolic Ca2+ concentration and increased the rate of NO2- synthesis. At maximal concentrations of TG, the rate of stimulated NO2- production was linear for at least 20 min and was eightfold higher than the basal rate (53.5 +/- 1.8 pmol.min-1.mg protein-1). Incubation of cells in gas mixtures chosen to produce PO2 values in the physiological range led to a progressive fall in the rate of TG-stimulated NO2- production, as O2 concentrations were reduced from that of room air. The half-maximal effective concentration for NO2- production by intact cells was found to occur at 38 Torr. PO2 values higher than that of room air did not lead to a change in the rate of TG-stimulated NO2- production. To confirm that measurement of NO2- accurately reflects total NO production, both NO2- plus NO3- were measured in buffer samples from cells incubated in either room air or N2. The sum of these NO oxidation products was inhibited similarly by hypoxia. These findings suggest that O2 is an important determinant of NOS activity in hypoxic tissues or in vascular beds such as the pulmonary arterial or fetal circulation where PO2 values in the range of 40 Torr are encountered normally.


Subject(s)
Calcium/metabolism , Endothelium, Vascular/physiology , Nitric Oxide Synthase/metabolism , Nitric Oxide/biosynthesis , Oxygen/pharmacology , Animals , Aorta , Cattle , Cell Hypoxia , Cells, Cultured , Egtazic Acid/pharmacology , Endothelium, Vascular/drug effects , Kinetics , Luminescent Measurements , Thapsigargin/pharmacology , omega-N-Methylarginine/pharmacology
15.
Am J Physiol ; 272(6 Pt 1): L1167-73, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9227519

ABSTRACT

Nitric oxide (NO.) has been proposed to modulate hypoxic vasoconstriction in the lung. The activity of nitric oxide synthase (NOS) can be inhibited by hypoxia because molecular oxygen is a necessary substrate for the enzyme. On the basis of this mechanism, we hypothesized that NOS activity has a key role in regulation of pulmonary vascular tone during hypoxia. We measured oxidation products of NO. released into the vasculature of isolated buffer-perfused rabbit lung ventilated with normoxic (21% O2), moderately hypoxic (5% O2), or anoxic (0% O2) gas using two methods. Mean PO2 in perfusate exiting the lung was 25 Torr during anoxic ventilation and 47 Torr during moderately hypoxic ventilation. We found that the amount of the NO. oxidation product nitrite released into the perfusate was suppressed significantly during ventilation with anoxic but not moderately hypoxic gas. During normoxic ventilation, nitrite release was inhibited by pretreatment with NG-monomethyl-L-arginine, a competitive inhibitor of NOS. To confirm that changes in nitrite concentration reflected changes in NO. release into the perfusate, major oxidation products of NO. (NOx) were assayed using a method for reduction of these products to NO. by vanadium(III) Cl. Release of NOx into the perfusate was suppressed by severe hypoxia (anoxic ventilation), and this effect was reversed by normoxia. Pulmonary vasoconstriction was observed during severe but not moderate hypoxia and was related inversely to the rate of nitrite release. These observations provide evidence that decreased NO. production contributes to the pulmonary vasoconstrictor response during severe hypoxia.


Subject(s)
Hypoxia , Lung/physiology , Nitric Oxide/biosynthesis , Pulmonary Artery/physiology , omega-N-Methylarginine/pharmacology , Animals , Blood Pressure/drug effects , Glutathione/analogs & derivatives , Glutathione/pharmacology , In Vitro Techniques , Lung/drug effects , Lung/physiopathology , Male , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Nitrates/metabolism , Nitrites/metabolism , Nitroso Compounds/pharmacology , Oxygen , Partial Pressure , Pulmonary Artery/drug effects , Rabbits , S-Nitrosoglutathione , Vasoconstriction/drug effects
16.
Am J Physiol ; 272(1 Pt 1): C99-108, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9038816

ABSTRACT

Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) has been identified as a potential target for nitric oxide (NO)-mediated cellular toxicity. We have previously shown that NO inhibits GAPDH by S-nitrosylation of the active site cysteine residue, which is reversed by low-molecular-weight thiols. Because endothelial cells contain high concentrations of low-molecular-weight thiols, principally glutathione, we investigated the effect of NO on GAPDH activity in intact endothelial cells and the influence that cellular glutathione has on GAPDH inhibition. Our results show that incubation of cells with an exogenous NO-generating system resulted in inhibition of GAPDH activity. The mechanism for inhibition appears to involve reversible modification of GAPDH because addition of thiols to cell extracts restored activity. Furthermore, cells were able to completely recover GAPDH activity after removal of the NO-generating system. Recovery did not require de novo protein synthesis. Depletion of cellular glutathione levels by treatment of cells with buthionine sulfoximine resulted in greater NO-mediated GAPDH inhibition as well as a lesser ability to recover activity. Finally, disruption of the glutathione redox cycle with the glutathione reductase inhibitor, 1,3-bis(2-chloroethyl)-1-nitrosourea, increased the extent of NO-mediated GAPDH inhibition and decreased both the rate and degree of recovery of GAPDH-activity. These results suggest that the glutathione redox cycle plays an important role not only in regulating the extent of NO-mediated GAPDH inhibition but also in the ability of endothelial cells to recover from NO-mediated GAPDH inhibition.


Subject(s)
Glutathione/metabolism , Glyceraldehyde-3-Phosphate Dehydrogenases/antagonists & inhibitors , Nitric Oxide/physiology , Animals , Buthionine Sulfoximine/pharmacology , Carmustine/pharmacology , Cattle , Cells, Cultured , Dithiothreitol/pharmacology , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Enzyme Inhibitors/pharmacology , Glutathione/antagonists & inhibitors , Glutathione Reductase/antagonists & inhibitors , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Oxidation-Reduction
17.
Am J Physiol ; 271(1 Pt 1): C332-7, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8760062

ABSTRACT

We have characterized the ability of several cell types associated with the microvasculature of solid tumors to release nitric oxide (NO.) in response to increases in cytosolic Ca2+ concentration ([Ca2+]c). EA.hy926 immortalized human umbilical vein endothelial cells (EC), rat fibroblasts (RFL), and tumorigenic cells isolated from R3230Ac rat mammary adenocarcinoma (MaC) were treated with thapsigargin (TG), an inhibitor of Ca(2+)-ATPase. NO. output was measured via a chemiluminescence detection system. Baseline NO. output was detectable only for EC. TG caused a significant increase in EC NO. output that could be blocked with NG-monomethyl-L-arginine and restored with L-arginine. TG did not stimulate NO. release from RFL or MaC cells, despite elevating [Ca2+]c in all cells. A Ca(2+)-dependent isoform of NO synthase (eNOS) was detected by immunoblot only in EC. These data indicate that EC, but not RFL or MaC, are capable of Ca(2+)-dependent NO. release and suggest that any Ca(2+)-dependent NO. release within this tumor is primarily of endothelial (and not tumorigenic cell) origin.


Subject(s)
Adenocarcinoma/metabolism , Calcium/physiology , Endothelium, Vascular/metabolism , Mammary Neoplasms, Experimental/metabolism , Nitric Oxide/metabolism , Adenocarcinoma/pathology , Animals , Arginine/pharmacology , Calcium-Transporting ATPases/antagonists & inhibitors , Cell Line , Female , Fibroblasts/metabolism , Humans , Immunoblotting , Isoenzymes/metabolism , Mammary Neoplasms, Experimental/pathology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Rats , Thapsigargin/pharmacology , omega-N-Methylarginine/pharmacology
18.
Am J Physiol ; 269(6 Pt 1): C1489-95, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8572178

ABSTRACT

Arachidonic acid and its metabolites are important cellular mediators. In this study, we report a novel role for arachidonic acid in vascular cell signaling. We tested the effects of exogenous arachidonic acid on protein tyrosine phosphorylation in cultured vascular endothelial and smooth muscle cells. Arachidonic acid stimulated the phosphorylation of tyrosine-containing proteins of approximately 58, 93, and 120 kDa in the three cell types studied. This response was dose dependent, with a maximum effect observed with 40 microM arachidonic acid. Phosphorylation was rapid and transient, reaching a peak 0.5 min after the addition of arachidonic acid and returning to baseline by 8 min. A common set of protein substrates was phosphorylated in smooth muscle cells treated with the Ca(2+)-mobilizing agonist endothelin, concomitant with an increase in endogenous unesterified arachidonic acid. To determine whether the protein tyrosine phosphorylation was due to arachidonic acid or to a metabolite, we used inhibitors of cyclooxygenase, lipoxygenase, and epoxygenase pathways. Ibuprofen, nordihydroguaiaretic acid, eicosatriynoic and eicosatetraynoic acids, and 8-methoxypsoralen failed to inhibit the arachidonic acid-mediated response. We also found increased protein tyrosine phosphorylation after treatment with oleic, linolenic and gamma-linoleic acid. These results suggest a mechanism of protein tyrosine phosphorylation that is directly stimulated by unmetabolized unsaturated fatty acids.


Subject(s)
Arachidonic Acid/pharmacology , Endothelium, Vascular/metabolism , Muscle, Smooth, Vascular/metabolism , Tyrosine/metabolism , Animals , Arachidonic Acid/metabolism , Cells, Cultured , Endothelium, Vascular/cytology , Fatty Acids, Unsaturated/pharmacology , Muscle, Smooth, Vascular/cytology , Phosphorylation/drug effects , Swine
19.
Am J Physiol ; 269(3 Pt 1): C739-49, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7573405

ABSTRACT

Nitric oxide (NO), produced by vascular endothelial cells, mediates both physiological and pathological responses. Although the molecular targets responsible for NO-mediated endothelial cell injury are not known, one candidate is the glycolytic enzyme, glyceraldehyde-3-phosphate dehydrogenase (GAPDH). In this study, we investigated the mechanism involved in NO-mediated GAPDH inhibition and found that S-nitrosoglutathione (GSNO) inhibited GAPDH activity in both purified enzyme preparations and endothelial cells. Furthermore, GSNO-mediated GAPDH inhibition occurred by modification of the active site cysteine residue in GAPDH, since increasing concentrations of the substrate, glyceraldehyde-3-phosphate, which interacts with the active site cysteine residue, protected GAPDH from inhibition by GSNO. Although under certain conditions both GSNO and the NO donor, sodium nitroprusside (SNP), led to the covalent NAD(+)-dependent modification of GAPDH, this putative ADP ribosylation was unlikely to be the primary mechanism for inhibition, since the stoichiometry was extremely low, and, in the case of GSNO, inhibition was completely reversed by thiol reagents. Furthermore, GSNO effectively S-nitrosylated GAPDH, and the extent of nitrosylation was linearly correlated with the degree of inhibition such that addition of 1 mole of NO per mole of GAPDH monomer was necessary to inhibit the enzyme. Consistent with this finding, GSNO-mediated GAPDH inhibition was reversible with low-molecular-weight thiols, and the reversal of inhibition correlated with the "denitrosylation" of GAPDH. These results suggest that endothelial GAPDH is a target for NO and that inhibition occurs principally by the reversible S-nitrosylation of the active site cysteine residue in GAPDH.


Subject(s)
Glutathione/analogs & derivatives , Glyceraldehyde-3-Phosphate Dehydrogenases/antagonists & inhibitors , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Nitroso Compounds/metabolism , Nitroso Compounds/pharmacology , Animals , Cattle , Cell Line , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Glutathione/metabolism , Glutathione/pharmacology , Glyceraldehyde-3-Phosphate Dehydrogenases/chemistry , Nitric Oxide/physiology , Pulmonary Artery/cytology , Pulmonary Artery/enzymology , S-Nitrosoglutathione
20.
Am J Physiol ; 269(3 Pt 1): C757-65, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7573407

ABSTRACT

Vascular endothelium responds to Ca(2+)-mobilizing agonists by producing nitric oxide (NO), a potent vasodilator and inhibitor of platelet aggregation. Regulation of constitutively expressed endothelial NO synthase (eNOS) in intact cells is not well understood. We investigated the kinetics of NO formation in response to Ca(2+)-mobilizing agonists, the requirement for extracellular L-arginine, and the role of NO in regulating eNOS activity. When endothelial cells were stimulated with bradykinin and ATP in the presence of 100 microM L-arginine, we observed a rapid and transient rise in intracellular Ca2+ concentration ([Ca2+]i) from 50 +/- 8 nM to 698 +/- 74 and 637 +/- 53 nM, respectively, and a rapid and transient rise in NO production from a basal level of 37 pmol.min-1.mg protein-1 to 256 and 275 pmol.min-1.mg protein-1, respectively. When cells were stimulated with A-23187 or thapsigargin in the presence of 100 microM L-arginine, we observed a sustained increase in [Ca2+]i and a sustained increase in NO production. The rate of NO synthesis was linear over 30 min, rising above control levels of 7 pmol/min to 53 pmol/min for A-23187 and 62 pmol/min for thapsigargin. Thapsigargin stimulated NO production and [Ca2+]i with 50% effective concentration values of 0.01 and 0.05 microM, respectively. Ca(2+)-stimulated NO production was attenuated by the NO synthase inhibitor NG-monomethyl-L-arginine, the removal of extracellular L-arginine, and the Ca(2+)-chelator ethylene glycol-bis(beta-aminoethyl ether)-N,N,N',N'-tetraacetic acid. When we exposed cells to NO gas (3.1 mM for 15 min) and S-nitrosoglutathione (10 mM for 1 h) thapsigargin-stimulated NO production was decreased by 50%.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Aorta/metabolism , Calcium/physiology , Endothelium, Vascular/metabolism , Nitric Oxide Synthase/metabolism , Animals , Aorta/cytology , Arginine/antagonists & inhibitors , Arginine/pharmacology , Biological Transport , Calcium/agonists , Cattle , Cells, Cultured , Endothelium, Vascular/cytology , Intracellular Membranes/metabolism , Nitric Oxide/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...