Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 108
Filter
Add more filters










Publication year range
1.
Arterioscler Thromb Vasc Biol ; 21(12): 1915-20, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11742864

ABSTRACT

The role of extracellular signal-regulated kinase (ERK) was studied in the signaling pathway by which interleukin-1beta (IL-1beta) increases the expression of inducible NO synthase (iNOS) in rat vascular smooth muscle cells. IL-1beta induced a rapid and transient activation of nuclear factor-kappaB (NF-kappaB), followed by a prolonged activation of NF-kappaB that was required to induce iNOS expression. Either PD98059 or U0126, selective inhibitors of ERK activation, did not influence IL-1beta-induced early activation but effectively reduced the prolonged activation of NF-kappaB and significantly reduced IL-1beta induction of iNOS. Transfection with antisense, but not sense, phosphorothioate-modified oligodeoxynucleotides directed toward ERK also reduced IL-1beta-induced prolonged NF-kappaB activation and iNOS expression. IkappaBbeta, but not IkappaBalpha degradation, induced by IL-1beta was markedly attenuated when ERK activation was inhibited and could be partially responsible for the persistent NF-kappaB activation. These data suggest that ERK activity is required for persistent NF-kappaB activation by IL-1beta that is necessary for iNOS gene expression.


Subject(s)
Interleukin-1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/metabolism , NF-kappa B/metabolism , Nitric Oxide Synthase/genetics , Animals , Aorta, Thoracic , Butadienes/pharmacology , Cells, Cultured , DNA/metabolism , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Gene Expression/physiology , Nitric Oxide Synthase Type II , Nitriles/pharmacology , Rats , Transcription, Genetic
2.
J Biol Chem ; 275(16): 12223-30, 2000 Apr 21.
Article in English | MEDLINE | ID: mdl-10766859

ABSTRACT

These studies describe inhibitory effects of N-acetylcysteine on several biochemical events associated with the activation of extracellular signal-regulated kinases (ERK) by angiotensin II in the cardiac fibroblast and compare these effects with those of the nitric oxide donor, S-nitroso-N-acetylpenicillamine, an agent we showed previously to inhibit angiotensin II-induced ERK activation and the concomitant phosphorylation of proline-rich tyrosine kinase 2 (Wang, D., Yu, X., and Brecher, P. (1999) J. Biol. Chem. 274, 24342-24348). The transactivation of the epidermal growth factor receptor by angiotensin II, a process required for the activation of ERK, was inhibited by N-acetylcysteine but not by nitric oxide. The transactivation of the epidermal growth factor receptor by angiotensin II was shown to be independent of intracellular calcium increases. Nitric oxide, but not N-acetylcysteine, inhibited the angiotensin II-induced increase in intracellular Ca(2+). Neither nitric oxide nor N-acetylcysteine inhibited either phospholipase C activation or inositol triphosphate generation in response to angiotensin II. N-Acetylcysteine did inhibit the phosphorylation of the calcium sensitive tyrosine kinases PYK2 and Src, effects that also occurred using nitric oxide. These studies describe a novel effect of N-acetylcysteine on cross-talk between a G protein-linked receptor and a tyrosine kinase receptor and offer additional molecular insight to explain how N-acetylcysteine and nitric oxide act at different sites and might have an additive effect on specific hormonal responses.


Subject(s)
Acetylcysteine/pharmacology , Angiotensin II/pharmacology , Calcium/metabolism , ErbB Receptors/metabolism , Nitric Oxide/pharmacology , Acetylcysteine/metabolism , Animals , Cells, Cultured , Epidermal Growth Factor/metabolism , Fibroblasts/metabolism , Heart/drug effects , Inositol 1,4,5-Trisphosphate/metabolism , Myocardium/metabolism , Nitric Oxide Donors/metabolism , Oxidative Stress , Penicillamine/analogs & derivatives , Penicillamine/metabolism , Phosphorylation , Rats , Transcriptional Activation
3.
Hypertension ; 35(4): 914-8, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10775561

ABSTRACT

We have reported previously that N-acetyl-L-cysteine facilitated interleukin-1beta-induced nitric oxide synthase (iNOS) expression in rat vascular smooth muscle cells. The present study compares the effect of N-acetyl-L-cysteine with other antioxidants and tested the possibility that N-acetyl-L-cysteine potentiates iNOS induction by a mechanism involving activation of p44/42 mitogen-activated protein kinases (MAPKs). The effect of N-acetyl-L-cysteine on potentiating interleukin-1beta-induced nitrite production and iNOS expression was mimicked either by the enantiomers, L-cysteine and D-cysteine, or by a non-thiol-containing antioxidant, L-ascorbic acid. Interleukin-1beta activated p44/42 MAPK, and this activation was enhanced in the presence of N-acetyl-L-cysteine. Inhibition of p44/42 MAPK phosphorylation by the selective inhibitor PD98059 clearly inhibited iNOS expression induced by interleukin-1beta either in the absence or in the presence of N-acetyl-L-cysteine. These observations, combined with previous results, indicate that p44/42 MAPK activation is required for interleukin-1beta induction of iNOS and that N-acetyl-L-cysteine may act as a reducing agent and facilitate interleukin-1beta-induced iNOS expression through a reduction/oxidation-related mechanism involving potentiation of cytokine activation of the p44/42 MAPK signaling pathway.


Subject(s)
Acetylcysteine/pharmacology , Free Radical Scavengers/pharmacology , Interleukin-1/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/enzymology , Nitric Oxide Synthase/metabolism , Animals , Cells, Cultured , Drug Synergism , Enzyme Induction/drug effects , MAP Kinase Signaling System/drug effects , Male , Mitogen-Activated Protein Kinase 3 , Nitric Oxide Synthase Type II , Rats , Rats, Wistar
4.
Arterioscler Thromb Vasc Biol ; 20(1): 27-34, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10634797

ABSTRACT

This study was performed to investigate whether the expression of p21(Sdi1/Cip1/Waf1), one of the cyclin-dependent kinase inhibitor proteins, could be regulated by nitric oxide (NO) and might account for the antiproliferative effect of NO. Quiescent adventitial fibroblasts were stimulated to proliferate by serum addition and by NO donors added during different phases of the cell cycle. [(3)H]Thymidine incorporation was markedly reduced by S-nitroso-N-acetyl-penicillamine (SNAP) added either with serum at quiescence or at later time point in the cell cycle. Northern and Western blot analyses showed that addition of SNAP either at quiescence or 15 hours after serum addition induced a rapid induction of p21 mRNA and protein. Immunoprecipitation studies and electrophoretic mobility shift analysis indicate that the treatment of cells with SNAP induced the phosphorylation of p53 (a tumor suppressor protein) and enhanced the ability of p53 to bind DNA when SNAP was added during the cell cycle. The increased expression of p21 mRNA or p53 activation during late G(1) or S phase was also caused by addition of 8-bromo-cGMP and effectively blocked by a specific inhibitor of the soluble guanylate cyclase. Furthermore, this response to SNAP was blocked by an inhibitor of protein kinase G. These studies implicate NO as a potential regulator of the cell cycle in aortic adventitial fibroblasts through a cGMP-mediated transcriptional mechanism involving the induction of p21.


Subject(s)
Aorta/metabolism , Cell Cycle/physiology , Cyclins/metabolism , Nitric Oxide/metabolism , Animals , Aorta/cytology , Aorta/drug effects , Base Sequence , Cell Cycle/drug effects , Cyclic GMP/metabolism , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , DNA/biosynthesis , DNA Primers/genetics , Fibroblasts/drug effects , Fibroblasts/metabolism , Nitric Oxide Donors/pharmacology , Penicillamine/analogs & derivatives , Penicillamine/pharmacology , Phosphorylation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Thymidine/metabolism , Tumor Suppressor Protein p53/metabolism
5.
J Biol Chem ; 275(15): 11389-96, 2000 Apr 14.
Article in English | MEDLINE | ID: mdl-10753954

ABSTRACT

Nitric oxide (NO) regulates the expression of p21(Waf1/Cip1) in several cell types. The present study examined the role of both the extracellular signal-regulated kinase (ERK) and p70 S6 kinase (p70(S6k)) in the NO-induced increase in p21 expression that occurred in adventitial fibroblasts during the cell cycle. Both ERK and p70(S6k) were phosphorylated in response to the NO donor S-nitroso-N-acetylpenicillamine (SNAP) and the activation was rapid, transient, and preceded increased p21 expresion under defined conditions where serum was present. Addition of a selective inhibitor of ERK phosphorylation (PD98059) prevented the subsequent phosphorylation of p70(S6k) and the increase in p21 protein. Both cGMP and cAMP activated both ERK and p70(S6k), whereas only selective inhibitors of protein kinase G prevented the activation of the kinases by SNAP. A complex between ERK and p70(S6k) was documented by immunoprecipitation procedures. Rapamycin blocked p70(S6k) phosphorylation induced by NO and also inhibited p53 phosphorylation and p21 expression whereas PD98059 only prevented the NO-induced increase in p21 protein without influencing either p53 activation or p21 mRNA expression. The studies show a unique relationship between NO, ERK, and p70(S6k) and also provide evidence for a novel role of p70(S6k) in the activation of p53.


Subject(s)
Cyclic GMP/physiology , Cyclins/biosynthesis , Mitogen-Activated Protein Kinases/physiology , Nitric Oxide/physiology , Ribosomal Protein S6 Kinases/physiology , Animals , Cyclic AMP-Dependent Protein Kinases/physiology , Cyclic GMP-Dependent Protein Kinases , Cyclin-Dependent Kinase Inhibitor p21 , Enzyme Activation , Male , Penicillamine/analogs & derivatives , Penicillamine/pharmacology , Phosphorylation , Platelet-Derived Growth Factor/pharmacology , Protein Kinases/physiology , Rats , Rats, Wistar , Tumor Suppressor Protein p53/metabolism
6.
Hypertension ; 34(6): 1259-64, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10601128

ABSTRACT

The expression of inducible nitric oxide synthase (iNOS) is a characteristic response to inflammation and can be inhibited with sodium salicylate. We used the cytokine-induced iNOS induction in cardiac fibroblasts as a model system in which to test the hypothesis that effects on mitogen-activated protein kinases (MAPKs) may explain the mechanism by which salicylate exerts its anti-inflammatory effects. Tumor necrosis factor-alpha (TNF-alpha) alone can induce extracellular signal-regulated kinase (ERK), p38 MAPK, and c-Jun N-terminal kinase activity in a rapid and transient manner, whereas interferon-gamma (IFN-gamma) can induce only ERK. The inhibition of either the ERK pathway or p38 MAPK activity with selective inhibitors blocked cytokine-induced iNOS protein and nitrite production. Salicylate treatment inhibited iNOS expression induced by TNF-alpha and IFN-gamma and attenuated the phosphorylation of ERK by TNF-alpha and IFN-gamma either alone or in combination. Salicylate had no obvious effect on the activation of p38 MAPK or c-Jun N-terminal kinase. The results showed that salicylate inhibited the phosphorylation of ERK and iNOS expression induced by cytokines in a dose-dependent manner and suggested that salicylate exerts its anti-inflammatory action in part through inhibition of the ERK pathway and iNOS induction.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Fibroblasts/drug effects , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Nitric Oxide Synthase/antagonists & inhibitors , Sodium Salicylate/pharmacology , Animals , Blotting, Western , Cells, Cultured , Cytokines/pharmacology , Enzyme Induction/drug effects , Enzyme Inhibitors/pharmacology , Fibroblasts/cytology , Fibroblasts/enzymology , Interferon-gamma/pharmacology , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/metabolism , Myocardium/cytology , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , RNA, Messenger/metabolism , Rats , Tumor Necrosis Factor-alpha/pharmacology , p38 Mitogen-Activated Protein Kinases
7.
Hypertension ; 34(4 Pt 1): 574-9, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10523329

ABSTRACT

The effect of N-acetyl-L-cysteine on interleukin-1beta-induced nitric oxide synthase expression was studied in rat vascular smooth muscle cells to determine if the reduction/oxidation state would modulate cytokine-induced changes. Interleukin-1beta induced the production of nitrite, a stable metabolite of nitric oxide in a time- and dose-dependent manner. Cytokine-induced nitrite production was enhanced by the addition of N-acetyl-L-cysteine in a dose-dependent manner, with a >50% increase produced by the addition of 1 mmol/L N-acetyl-L-cysteine. There was no influence on nitrite production when the cells were treated with N-acetyl-L-cysteine alone. Northern and Western blot analyses revealed that the upregulation of interleukin-1beta-induced nitric oxide production by N-acetyl-L-cysteine resulted from an enhanced expression of inducible nitric oxide synthase. Interferon-gamma or tumor necrosis factor-alpha when used alone had no influence on nitrite production in the absence or presence of N-acetyl-L-cysteine. Nitrite accumulation was higher by the cells treated with interleukin-1beta combined with either interferon-gamma or tumor necrosis factor-alpha compared with those treated with interleukin-1beta alone. N-Acetyl-L-cysteine upregulated nitrite production and inducible nitric oxide synthase expression induced by combination treatment with interleukin-1beta and either interferon-gamma or tumor necrosis factor-alpha. However, N-acetyl-L-cysteine had no significant influence in cytokine-induced activation of nuclear factor-kappaB or signal transducer and activator of transciption-1, as assessed by electrophoretic mobility shift assays. These results demonstrate that N-acetyl-L-cysteine possibly acted as a thiol-containing reducing agent and facilitated the expression of inducible nitric oxide synthase in rat vascular smooth muscle cells by cytokines through a mechanism that is independent of nuclear factor-kappaB or signal transducer and activator of transciption-1.


Subject(s)
Acetylcysteine/pharmacology , Interleukin-1/pharmacology , Muscle, Smooth, Vascular/drug effects , Nitric Oxide Synthase/biosynthesis , Animals , Cells, Cultured , Drug Synergism , Electrophoresis , Male , Muscle, Smooth, Vascular/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Nitrites/metabolism , Rats , Rats, Wistar
8.
J Biol Chem ; 274(34): 24342-8, 1999 Aug 20.
Article in English | MEDLINE | ID: mdl-10446212

ABSTRACT

In a previous study, we showed that nitric oxide donors and N-acetylcysteine, either alone or in combination, inhibited the activation of several mitogen-activated protein kinases by angiotensin II in rat cardiac fibroblasts (Wang, D., Yu, X., and Brecher, P. (1998) J. Biol. Chem. 273, 33027-33034). In the present study, we have focused on the mechanism by which nitric oxide exerts this effect on the activation of extracellular signal-regulated kinase (ERK). We contrasted the effects of nitric oxide on ERK activation by angiotensin II and epidermal growth factor (EGF), since the transactivation of the EGF receptor has been implicated as a response to angiotensin II. We found that nitric oxide inhibited ERK activation by angiotensin II but did not inhibit the relatively slight but significant transactivation of the EGF receptor by angiotensin II. The tyrphostin AG1478, known to inhibit EGF receptor phosphorylation, also inhibited the angiotensin II and EGF-induced activation of ERK, the phosphorylation of the EGF receptor, and the subsequent association of Shc and Grb2. Nitric oxide did not affect either EGF receptor phosphorylation or Shc-Grb2 activation induced by either Ang II or EGF. However, the activation of the calcium-sensitive tyrosine kinase PYK2, which occurred in response to angiotensin II, but not EGF, was inhibited by nitric oxide. The data suggested that PYK2 activation may be an important inhibitory site in signaling pathways affected by nitric oxide.


Subject(s)
Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport , Angiotensin II/pharmacology , ErbB Receptors/metabolism , Mitogen-Activated Protein Kinases , Nitric Oxide/physiology , Protein-Tyrosine Kinases/antagonists & inhibitors , Transcriptional Activation , Animals , Calcium/physiology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cells, Cultured , Focal Adhesion Kinase 2 , Indoles/pharmacology , Maleimides/pharmacology , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Nitroprusside/pharmacology , Phosphorylation , Proteins/metabolism , Quinazolines , Rats , Shc Signaling Adaptor Proteins , Src Homology 2 Domain-Containing, Transforming Protein 1 , Tyrphostins/pharmacology
9.
Am J Hypertens ; 12(5): 467-75, 1999 May.
Article in English | MEDLINE | ID: mdl-10342784

ABSTRACT

In the current study we used in vitro and in vivo models to determine the sites of nitric oxide production in rat aortic tissue following cytokine stimulation. In vitro studies in which intact rat aortic rings were incubated with endotoxin (1 microg/mL) or interferon-gamma (600 U/mL) indicated that the expression of inducible nitric oxide synthase (iNOS) activity was increased as measured by Northern blot analysis or determination of nitrite production. In situ hybridization showed iNOS mRNA in the endothelium and adventitia of the incubated aortic rings but not in the media. Immunohistochemical staining showed a similar localization for iNOS protein in the incubated rings. Additional studies were performed in which bacterial endotoxin (4 mg/kg) was administered to rats, and iNOS expression was assayed using in situ hybridization and immunohistochemistry. Clear increases in iNOS mRNA and protein were found in aortic tissue. Endothelial and adventitial cells were the major source of iNOS, with relatively low amounts of iNOS mRNA present in medial smooth muscle, consistent with in vitro findings. These studies indicate that the aortic adventitia is a potential source of NO, and suggest that the adventitial fibroblast may have an important paracrine role in regulating arterial structure and function.


Subject(s)
Aorta, Thoracic/enzymology , Nitric Oxide Synthase/metabolism , Tunica Intima/enzymology , Animals , Aorta, Thoracic/drug effects , Blotting, Northern , Blotting, Western , Culture Media, Serum-Free , Enzyme Inhibitors/pharmacology , Fibroblasts/drug effects , Fibroblasts/enzymology , Gene Expression , In Situ Hybridization , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Male , Muscle, Smooth, Vascular/enzymology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , RNA, Messenger/metabolism , Rats , Rats, Wistar , Tunica Intima/cytology , Tunica Intima/drug effects
10.
Am J Hypertens ; 12(1 Pt 1): 28-34, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10075381

ABSTRACT

Angiotensin converting enzyme (ACE) inhibitors reduce the development of atherosclerosis in hypercholesterolemic animals across a wide range of species. Although the mechanism for these effects has not been well delineated, it has been assumed generally that both angiotensin II suppression and interference with the breakdown of bradykinin are involved. To determine whether angiotensin II receptor blockade provides similar effects as those observed with ACE inhibition, we examined the influence of irbesartan, an AT1 receptor antagonist, on aortic atherosclerosis in Watanabe heritable hyperlipidemic rabbits using the identical protocol that was employed in our earlier studies involving ACE inhibitors. At a dose of irbesartan (30 mg/kg/day), which was selected because it appeared to block most of the pressor effects of infused angiotensin in rabbits, no effect on atherosclerosis was observed. However, a higher dose of irbesartan (75 mg/kg/day) caused reductions in blood pressure and aortic atherosclerosis similar to those seen in earlier studies with ACE inhibitors. The decrease in aortic intimal surface involvement with irbesartan was from 38.9 +/- 3.8% in controls to 24.1 +/- 3.0% in the treated group (P < .01). Aortic cholesterol content was also significantly reduced in those animals (P < .02). The findings indicate that suppression of the renin-angiotensin system by AT1 receptor blockade in a genetically hypercholesterolemic rabbit model causes comparable inhibition of aortic atherosclerosis as that achieved by ACE inhibition, and that a mild reduction of blood pressure induced by both classes of agents may contribute to their antiatherosclerotic action in this model.


Subject(s)
Angiotensin Receptor Antagonists , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Antihypertensive Agents/therapeutic use , Arteriosclerosis/drug therapy , Biphenyl Compounds/therapeutic use , Peptidyl-Dipeptidase A/drug effects , Tetrazoles/therapeutic use , Animals , Antihypertensive Agents/administration & dosage , Aorta/drug effects , Aorta/metabolism , Aorta/pathology , Aortic Diseases/drug therapy , Aortic Diseases/metabolism , Aortic Diseases/physiopathology , Arteriosclerosis/metabolism , Arteriosclerosis/physiopathology , Biphenyl Compounds/administration & dosage , Blood Pressure/drug effects , Bradykinin/drug effects , Bradykinin/metabolism , Cholesterol/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Follow-Up Studies , Irbesartan , Peptidyl-Dipeptidase A/metabolism , Rabbits , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Tetrazoles/administration & dosage
11.
Hypertension ; 32(6): 1044-8, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9856971

ABSTRACT

-We have used the apolipoprotein E (apoE)-deficient mouse model to determine whether both the angiotensin II type 1 (AT1) and the alpha1-adrenergic receptors influence arteriosclerotic changes in this hyperlipidemic animal model. Mice were treated with antihypertensive drugs beginning at 9 weeks of age, and aortic atherosclerosis was measured after 12 weeks of treatment. Systolic blood pressure in the untreated apoE-deficient mouse averaged 104 mm Hg throughout the treatment period. Prazosin at a dose of 7.5 mg. kg-1. d-1 was ineffective in attenuating atherosclerosis and did not significantly reduce blood pressure. Losartan, at dosages of either 20 or 30 mg. kg-1. d-1, also did not influence atherosclerosis and had only a slight blood pressure-lowering effect. However, combined treatment with both prazosin and losartan markedly reduced atherosclerotic lesion development from an average lesion size per section of 2.6 to 1.5x10(5) microm2 (P<0.001) and significantly reduced blood pressure to 85+/-5 mm Hg. Treatment with NG-nitro-L-arginine methyl ester (40 mg. kg-1. d-1) produced significant elevations of blood pressure (127+/-3.8 mm Hg) but had no effect on the development of atherosclerosis. None of the treatments used affected plasma cholesterol throughout the 12-week period. These studies suggest that the vascular changes associated with atherosclerosis are influenced by a combination of AT1 and alpha1-adrenergic receptor activation.


Subject(s)
Adrenergic alpha-Antagonists/therapeutic use , Angiotensin Receptor Antagonists , Apolipoproteins E/deficiency , Arteriosclerosis/prevention & control , Animals , Antihypertensive Agents/therapeutic use , Arteriosclerosis/metabolism , Arteriosclerosis/pathology , Blood Pressure/drug effects , Drug Therapy, Combination , Hypercholesterolemia/complications , Hypercholesterolemia/drug therapy , Losartan/therapeutic use , Mice , Mice, Inbred C57BL , NG-Nitroarginine Methyl Ester/pharmacology , Prazosin/therapeutic use , Propranolol/pharmacology , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Receptors, Angiotensin/drug effects , Time Factors
12.
J Biol Chem ; 273(49): 33027-34, 1998 Dec 04.
Article in English | MEDLINE | ID: mdl-9830056

ABSTRACT

Angiotensin II acts on the cardiac fibroblast to produce a mitogenic response. Nitric oxide and N-acetylcysteine have been used to determine if oxidative stress influenced the effects of angiotensin II on the cardiac fibroblast. Angiotensin II activated the mitogen-activated protein kinases designated extracellular signal-regulated kinases within 5 min by interacting with the AT1 receptor. This activation was completely independent of protein kinase C and was inhibited when farnesylation was blocked, implicating Ras involvement. Pretreatment of cardiac fibroblasts with either N-acetylcysteine for 8 h or nitric oxide for 10 min suppressed this activation by angiotensin II in a dose-dependent manner. However, when both agents were added, inhibition was essentially complete. This combined effect of N-acetylcysteine and nitric oxide to block ERKs activation also was found if the activity was stimulated by either another growth factor (platelet-derived growth factor) or by the addition of phorbol ester, suggesting the effect was not limited to the receptor site alone. The results are consistent with the hypothesis that hormonal activation of mitogenic steps such as ERKs is influenced by increased oxidative stress, which is reduced by the combined effects of N-acetylcysteine and nitric oxide.


Subject(s)
Acetylcysteine/pharmacology , Angiotensin II/pharmacology , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Heart/drug effects , Myocardium/enzymology , Nitric Oxide/pharmacology , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cells, Cultured , Enzyme Activation , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/enzymology , Glutathione/metabolism , Myocardium/cytology , Phosphorylation , Protein Kinase C/metabolism , Rats
13.
Circ Res ; 82(7): 810-8, 1998 Apr 20.
Article in English | MEDLINE | ID: mdl-9562441

ABSTRACT

The purpose of this study was to determine whether superoxide anion is produced endogenously in the rat aortic adventitia and whether sufficient superoxide anion is produced to interfere with the response of the rat aorta to nitric oxide. Relaxation was measured in rings of the rat thoracic aorta, which were oriented so that the adventitial or luminal surface could be preferentially exposed to nitric oxide or sodium nitroprusside. To accomplish this, the rings were mounted (1) with the adventitia facing outward, (2) with the adventitia facing inward after inverting, or (3) with the adventitia facing outward after inverting twice (to control for the inverting procedure). The relaxation to nitric oxide, but not to sodium nitroprusside, was less in rings with the adventitia facing outward compared with those in which it faced inward. In contrast, the response to nitric oxide via either surface was similar when extracellular superoxide anion was scavenged with superoxide dismutase. Incubation of rings with nitro blue tetrazolium (NBT) resulted in blue formazan staining of the adventitia, and lucigenin chemiluminescence was significantly greater when detected from the adventitial compared with the intimal aspect of the artery. The reduction of NBT in intact aortic rings was 30+/-2 pmol x min(-1) x mg(-1) and was significantly decreased by superoxide dismutase to 19+/-2 pmol x min(-1) x mg(-1) and by a synthetic superoxide dismutase mimic, Euk-8, to 11+/-2 pmol x min(-1) x mg(-1). The NADPH oxidase inhibitor, diphenyleneiodonium, decreased NBT reduction to 9+/-1 pmol x min(-1) x mg(-1), whereas inhibitors of xanthine oxidase, mitochondrial oxidases, and nitric oxide synthase were ineffective. Immunohistochemical staining indicated the localization of NADPH oxidase proteins gp91phox, p22phox, p47phox, and p67phox almost exclusively in the adventitia of the rat aorta with no substantial staining in the media. These results indicate that NADPH oxidase located in the adventitia of rat thoracic aorta generates sufficient extracellular superoxide anion to constitute a barrier capable of inactivating nitric oxide. This study suggests that adventitial superoxide anion can play a role in the pathophysiology of the arterial wall.


Subject(s)
Aorta, Thoracic/metabolism , Nitric Oxide/antagonists & inhibitors , Superoxides/metabolism , Tunica Intima/metabolism , Vasodilation/physiology , Acridines , Animals , Indicators and Reagents , Isometric Contraction/physiology , Luminescent Measurements , Male , NADPH Oxidases/analysis , Nitroblue Tetrazolium , Phenylephrine/pharmacology , Rats , Rats, Wistar , Vasoconstrictor Agents/pharmacology
14.
Proc Assoc Am Physicians ; 110(1): 22-31, 1998.
Article in English | MEDLINE | ID: mdl-9460080

ABSTRACT

Increased plasma levels of homocysteine are an independent risk factor for atherothrombosis. While the endothelial cytotoxicity of homocysteine has been attributed to oxidative stress associated with the reactivity of the thiol group, the oxidative effect of homocysteine on vascular smooth-muscle cells has not been investigated. Recent evidence suggests that expression of inducible nitric oxide synthase (iNOS), or Nos2 gene product, in vascular smooth-muscle cells may, in part, promote atherosclerosis by increasing local oxidative stress. We therefore hypothesized that homocysteine contributes to atherosclerosis by affecting cytokine-induced production of nitric oxide (NO) by vascular smooth-muscle cells. Confluent rat aortic smooth-muscle cells were exposed to a range of concentrations of homocysteine for 4 hr, then were treated with interferon-gamma, interleukin-1 beta, and lipopolysaccharide to induce iNOS. Media NOx content (nitrite plus S-nitrosothiol) was measured over 24 hr using the Saville reaction. As compared to controls, 5, 50, and 500 microM homocysteine produced a dose-dependent increase in media NOx content, an effect that was primarily a consequence of increased S-nitrosothiol production. iNOS enzyme activity and iNOS protein levels were increased significantly in the homocysteine-treated cells as compared with controls. Northern analysis showed that homocysteine treatment increased steady-state Nos2 mRNA levels by 61% at 6 hr as compared with controls, an effect that was not caused by changes in message stability. By electrophoretic mobility shift assay, homocysteine activated NF-kappa B and also potentiated cytokine activation of NF-kappa B. These data demonstrate that exposure of vascular smooth-muscle cells to pathophysiologically relevant concentrations of homocysteine prior to cytokine stimulation leads both to an increase in NO production and to an NF-kappa B-mediated increase in Nos2 transcription. Upregulation of Nos2 may contribute to the inflammatory response that characterizes early atherogenesis and may, in part, account for the adverse vascular effects of hyperhomocysteinemia.


Subject(s)
Homocysteine/metabolism , Muscle, Smooth, Vascular/metabolism , NF-kappa B/metabolism , Nitric Oxide Synthase/genetics , Nitric Oxide/metabolism , Transcriptional Activation , Animals , Cells, Cultured , Homocysteine/pharmacology , Hydrogen Peroxide/metabolism , Muscle, Smooth, Vascular/cytology , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , RNA, Messenger , Rats
15.
Hypertension ; 30(5): 1035-40, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9369252

ABSTRACT

Our previous in vivo studies (Hou et al. J Clin Invest. 1995;96:2469-2477.) demonstrated that chronic inhibition of nitric oxide synthase led to an exaggerated response to relatively low doses of angiotensin II, resulting in a rapid and marked cardiac fibrosis. To examine further the importance of angiotensin II in inducing cardiac fibrosis and the possibility that nitric oxide serves as a modulator of the proliferative effects of angiotensin II, we used cultured rat cardiac fibroblasts to study the interrelationships between these substances. Angiotensin II induced a delayed DNA synthetic response in quiescent cells that occurred 30 hours after exposure to the hormone. The most pronounced effect of angiotensin II on thymidine uptake occurred 36 to 42 hours after the addition to cells. This response was inhibited in a dose-dependent manner by the addition of either S-nitroso-N-acetylpenicillamine or sodium nitroprusside, each a source of nitric oxide. The nitric oxide donor was most effective in reducing thymidine incorporation when added 12 hours after angiotensin II, whereas the metabolite N-acetylpenicillamine had no effect at any time. The inhibitory effect of S-nitroso-N-acetylpenicillamine was mimicked by 8-bromoguanosine 3':5'-cyclic monophosphate but not by 8-bromoadenosine 3':5'-cyclic monophosphate. Nitric oxide donors did not appear to inhibit the induction of c-fos, Egr-1, or other immediate-early genes in response to angiotensin II. The results suggest that nitric oxide affects the cell cycle following the transition into G, and modulates the proliferation of fibroblasts during cardiac fibrosis induced by angiotensin II.


Subject(s)
Angiotensin II/pharmacology , DNA Replication/drug effects , DNA Replication/physiology , Immediate-Early Proteins , Myocardium/metabolism , Nitric Oxide/pharmacology , Animals , Animals, Newborn , Cells, Cultured , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , DNA-Binding Proteins/genetics , Early Growth Response Protein 1 , Enzyme Inhibitors/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Myocardium/cytology , Nitroprusside/pharmacology , Penicillamine/analogs & derivatives , Penicillamine/pharmacology , Proto-Oncogene Proteins c-fos/genetics , RNA, Messenger/metabolism , Rats , S-Nitroso-N-Acetylpenicillamine , Thymidine/antagonists & inhibitors , Thymidine/metabolism , Transcription Factors/genetics
16.
J Biol Chem ; 271(49): 31585-92, 1996 Dec 06.
Article in English | MEDLINE | ID: mdl-8940176

ABSTRACT

We have previously reported that salicylate inhibits the inducible NO synthase (NOS 2) in cytokine-induced cardiac fibroblasts (Farivar, R. S., Chobanian, A. V., and Brecher, P. (1996) Circ. Res. 78, 759-768). To define further the mechanism of inhibition of NOS 2 by salicylate, we investigated NOS 2 mRNA induction by cytokines and determined the kinetics of inhibition by salicylate as compared to dexamethasone. Interferon-gamma plus tumor necrosis factor-alpha induced NOS 2 mRNA synergistically in a time- and dose-dependent manner. Both dexamethasone and salicylate equally inhibited the induction of NOS 2 mRNA in a time- and dose-dependent fashion, both before and after cytokine induction. Salicylate also inhibited interferon-gamma plus interleukin-1beta-induced NOS 2 mRNA. After 24 h of cytokine stimulation, salicylate stopped the induction of NOS 2 mRNA, whereas dexamethasone delayed the accumulation of transcript. In half-life experiments of NOS 2 mRNA, we found that dexamethasone reduced the half-life of NOS 2 mRNA from 7 to 4 h, whereas salicylate had no effect on mRNA stability. Tumor necrosis factor-alpha and interferon-gamma induced NF-kappaB (p50/p65) and STAT-1, respectively, as assessed by gel shift assays. Salicylate did not inhibit the cytokine induction of NF-kappaB or STAT-1. This study suggests that the anti-inflammatory mechanism of salicylate involves inhibition of NOS 2 transcription and shows that the effect is independent of NF-kappaB activation.


Subject(s)
Myocardium/enzymology , Nitric Oxide Synthase/biosynthesis , Salicylates/pharmacology , Transcription, Genetic/drug effects , Animals , Blotting, Northern , Cells, Cultured , DNA-Binding Proteins/metabolism , Dexamethasone/pharmacology , Dose-Response Relationship, Drug , Enzyme Induction/drug effects , Fibroblasts/enzymology , Interferon-gamma/pharmacology , Mice , NF-kappa B/metabolism , STAT1 Transcription Factor , Salicylic Acid , Sodium Salicylate/pharmacology , Time Factors , Trans-Activators/metabolism , Tumor Necrosis Factor-alpha/pharmacology
17.
Am J Physiol ; 271(3 Pt 2): F527-38, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8853414

ABSTRACT

In the present study, we provide immunohistochemical and immunologic evidence to localize an abundant, 15.5-kDa protein to the soluble protein fraction of the proximal tubule. This 15.5-kDa protein binds fatty acids in vitro and has identity with amino acids 10-117 of alpha 2 mu-globulin (A2 fragment), a 19-kDa protein synthesized predominantly in the male liver. With reverse transcription-polymerase chain reaction, mRNA for A2 was detected in male liver but not in the male kidney. De novo accumulation of the 15.5-kDa protein was observed in the renal cortex of female rats given intravenous injections of purified 19-kDa protein (A2), suggesting intrarenal processing of the larger protein. The potential role of this protein in the proximal tubule, a site that utilizes fatty acids as an important metabolic substrate, was determined in isolated proximal tubule segments. Fatty acid and glucose oxidation rates were measured in three experimental models in which the 15.5-kDa protein was virtually absent: 1) uninephrectomized male rats treated with deoxycorticosterone acetate and salt, 2) male rats subjected to bilateral adrenalectomy, and 3) normal female rats. In the absence of the 15.5-kDa protein, fatty acid oxidation rates decreased by 30-55%, whereas glucose oxidation significantly increased in all three models. In female renal cortex, depletion of the 15.5-kDa protein was associated with a rise in heart fatty acid binding protein, an alternative intracellular transporter of fatty acids. These data support the hypothesis that a proteolytic cleavage product of hepatic alpha 2 mu-globulin may facilitate the oxidation of oleate, a hydrophobic ligand, in the proximal tubule.


Subject(s)
Alpha-Globulins/metabolism , Kidney Tubules, Proximal/metabolism , Liver/metabolism , Neoplasm Proteins , Nerve Tissue Proteins , Peptide Fragments/metabolism , Tumor Suppressor Proteins , Adrenalectomy , Alpha-Globulins/chemistry , Animals , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Desoxycorticosterone/pharmacology , Fatty Acid-Binding Protein 7 , Fatty Acid-Binding Proteins , Fatty Acids/metabolism , Female , Glucose/metabolism , Humans , Male , Molecular Weight , Myelin P2 Protein/chemistry , Myelin P2 Protein/metabolism , Oxidation-Reduction , Peptide Fragments/chemistry , Rats , Rats, Wistar , Sex Characteristics , Tissue Distribution
18.
Hypertension ; 28(2): 153-8, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8707375

ABSTRACT

In previous studies, we showed that in vivo infusion of angiotensin II (Ang II) to adult rats induced vascular changes in gene expression, and this effect did not depend solely on blood pressure elevation. To determine whether nitric oxide can influence the effects of Ang II on the vessel wall, we administered to rats Ang II separately or in combination with the arginine analogue N omega-nitro-L-arginine methyl ester, which inhibits nitric oxide synthase chronically when given in vivo. We measured changes in aortic medial thickness, the association of macrophages with the endothelial surface of the aorta, the presence of proliferating cell nuclear antigen in the intima and adventitia as an index of aortic cell cycle changes, and the expression of immunodetectable fibronectin as an index of changes in the extra-cellular matrix. After 18 days of nitric oxide inhibition, the major changes were increased medial thickness and a 3.5-fold increase in the number of adherent macrophages. Rats treated with two different doses of Ang II for 3 days had a fivefold and threefold increase in the number of proliferating cells from the intimal and adventitial regions, respectively. Combined treatment resulted in increased medial thickness, intimal and adventitial cell proliferation, and macrophage adherence. An increased and altered pattern of fibronectin distribution was found in all treatment groups. Losartan administration prevented the effects of Ang II but not of nitric oxide inhibition, whereas administration of L-arginine, prevented both intimal macrophage adherence and increased adventitial proliferation in rats given combined treatment. The data suggest that nitric oxide selectively influences macrophage association with the arterial wall, whereas Ang II and nitric oxide may have opposing effects on arterial cell proliferation.


Subject(s)
Angiotensin II/pharmacology , Aorta/drug effects , Arginine/analogs & derivatives , Enzyme Inhibitors/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Angiotensin II/administration & dosage , Angiotensin II/antagonists & inhibitors , Animals , Antihypertensive Agents/pharmacology , Aorta/cytology , Arginine/pharmacology , Biphenyl Compounds/pharmacology , Cell Division/drug effects , Dose-Response Relationship, Drug , Drug Interactions , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Imidazoles/pharmacology , Infusion Pumps, Implantable , Losartan , Macrophages/drug effects , Male , NG-Nitroarginine Methyl Ester , Proliferating Cell Nuclear Antigen/drug effects , Rats , Rats, Wistar , Tetrazoles/pharmacology
19.
Circ Res ; 78(5): 759-68, 1996 May.
Article in English | MEDLINE | ID: mdl-8620595

ABSTRACT

To determine if fibroblasts are a source of NO inflammatory myocardial diseases, we have studied the effect of cytokines on the inducible NO synthase (iNOS) in neonatal cardiac fibroblasts and tested whether nonsteroidal anti-inflammatory drugs can diminish the induction of iNOS. In primary cultures, interferon gamma (IFN), interleukin-1 beta (IL-1), or tumor necrosis factor-alpha (TNF) separately did not stimulate nitrite production, whereas IFN combined with IL-1 or TNF synergistically induced iNOS, both at the level of steady state mRNA and nitrite accumulation. Steady state mRNA levels for iNOS were obvious as early as 3 hours after the addition of IFN + TNF and remained elevated for at least 72 hours. Sodium salicylate inhibited cytokine-induced nitrite accumulation in a time- and dose-dependent manner (IC50, 750 mumol/L). The inhibition was reversible and occurred when salicylate was added either before or after cytokine induction. Aspirin (1 mmol/L) also inhibited nitrite production, whereas indomethacin (25 mumol/L) or acetaminophen (100 mumol/L) did not. TNF, either alone or combined with IFN, significantly stimulated prostaglandin E2, which was inhibited by either salicylate (4 mmol/L) or indomethacin (25 mumol/L). Salicylate, when given either before or after IFN + TNF, reduced mRNA levels of iNOS induced by cytokines. Salicylate did not affect iNOS enzymatic activity when added to the cytosolic lysate, although it was able to reduce enzymatic activity to 32% of induced levels when given to intact cells. These studies implicate cardiac fibroblasts as a source of NO in inflammatory cardiac diseases and suggest a possible therapeutic role for salicylate and aspirin in diminishing the steady state levels of iNOS mRNA.


Subject(s)
Aspirin/pharmacology , Myocardium/enzymology , Nitric Oxide Synthase/antagonists & inhibitors , Salicylates/pharmacology , Animals , Cyclooxygenase Inhibitors/pharmacology , Enzyme Induction/drug effects , Fibroblasts/enzymology , Interleukin-1/pharmacology , Myocardium/cytology , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Salicylic Acid , Tumor Necrosis Factor-alpha/pharmacology
20.
Am J Physiol ; 270(1 Pt 2): H81-7, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8769737

ABSTRACT

We have recently shown that inhibition of endogenous Cu,Zn superoxide dismutase (SOD) by diethyldithiocarbamate (DDC) increased superoxide anion levels in isolated rabbit aortic rings, describing a useful experimental model to examine the effects of oxidative stress on the vessel wall. The present study examined the effects of oxidative stress on the steady-state mRNA levels of glyceraldehyde-3-phosphate dehydrogenase (GAPDH; EC 1.2.1.12). Aortic rings were incubated in physiological salt solution at 37 degrees C for up to 6 h. DDC (2 mM) decreased total SOD activity to < 5% of control levels and increased superoxide anion level ninefold. Steady-state mRNA levels of GAPDH were increased under comparable conditions. Although decreased biological activity of endothelium-derived nitric oxide was indicated by lower basal guanosine 3',5'-cyclic monophosphate levels in aortic rings treated with DDC compared with those in control rings (1.2 +/- 0.1 vs. 1.9 +/- 0.3 fmol/microgram protein, P < 0.05), neither endothelium denudation nor NG-nitro-L-arginine methyl ester had any effects on the steady-state mRNA levels of GAPDH. The cell.permeable iron chelator 1,10-phenanthroline completely prevented the increases in GAPDH mRNA levels induced by DDC. These results suggest that oxidative stress resulting from inhibition of endogenous Cu,Zn SOD causes induction of GAPDH gene expression and that the hydroxyl radical, produced through the iron-catalyzed Haber-Weiss reaction, is the intracellular reactive oxygen species responsible for the DDC-stimulated increase in GAPDH mRNA.


Subject(s)
Aorta/metabolism , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Oxidative Stress , RNA, Messenger/metabolism , 1,2-Dihydroxybenzene-3,5-Disulfonic Acid Disodium Salt/pharmacology , Animals , Anions/metabolism , Cyclic GMP/metabolism , In Vitro Techniques , Iodoacetates/pharmacology , Iodoacetic Acid , Iron Chelating Agents/pharmacology , Male , Nitric Oxide Synthase/antagonists & inhibitors , Rabbits , Superoxide Dismutase/metabolism , Superoxides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...