Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Biochemistry ; 39(32): 9901-8, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10933809

ABSTRACT

Nitric oxide (NO) and related species serve as cellular messengers in various physiological and pathological processes. The monomeric G protein, Ras, transduces multiple signaling pathways with varying biological responses. We have previously reported that NO triggers Ras activation and recruitment of an effector, phosphatidylinositol 3'-kinase (PI3K) and Ras-dependent activation of mitogen-activated protein (MAP) kinases which include extracellular signal regulated kinases (ERKs), c-Jun NH(2)-terminal kinase (JNK), and p38 MAP kinase. In this study, we further defined NO-activated Ras signaling pathways. We have identified Raf-1 as another effector recruited by NO-activated Ras in T lymphocytes. NO activation results in association of Ras and Raf-1 and is biologically significant, as we observe an NO-induced increase in Raf-1 kinase activity. Downstream to Raf-1 kinase lie MAP kinases and their subsequent downstream targets, transcription factors. We found that treatment of T lymphocytes with NO yielded phosphorylation of the transcription factor, Elk-1. This phoshorylation is dependent on NO binding to the cysteine 118 residue of Ras. By further delineating the pathway with pharmacological inhibitors, Elk-1 phosphorylation was also found to be dependent on PI3K and ERK. Moreover, NO triggered an increase in mRNA levels of the proinflammatory cytokine, tumor necrosis factor-alpha (TNF-alpha), which was ERK dependent. Thus, we have defined an NO-induced signaling pathway in T lymphocytes arising at the membrane where NO-activated Ras recruits Raf-1 and culminating in the nucleus where Elk-1 is phosphorylated and TNF-alpha messenger RNA is induced. This NO-activated Ras-mediated signaling pathway may play a critical role in Elk-1-induced transcriptional activation of T lymphocytes, host defense and inflammation.


Subject(s)
DNA-Binding Proteins , Nitric Oxide/metabolism , Proto-Oncogene Proteins c-raf/metabolism , T-Lymphocytes/metabolism , Transcription Factors , ras Proteins/metabolism , Androstadienes/pharmacology , Cell Membrane/metabolism , Cell Nucleus/metabolism , Chromones/pharmacology , Enzyme Activation , Humans , Jurkat Cells , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Precipitin Tests , Protein Binding , Proto-Oncogene Proteins/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Wortmannin , ets-Domain Protein Elk-1
3.
J Biol Chem ; 274(52): 37315-20, 1999 Dec 24.
Article in English | MEDLINE | ID: mdl-10601298

ABSTRACT

p21(c-Ha-Ras) (Ras) can be activated by the guanine nucleotide exchange factor mSOS1 or by S-nitrosylation of cysteine 118 via nitric oxide (NO). To determine whether these two Ras-activating mechanisms modulate distinct biological effects, a NO-nonresponsive Ras mutant (Ras(C118S)) was stably expressed in the PC12 cells, a cell line that generates NO upon nerve growth factor treatment. We report here that Ras(C118S) functions indistinguishably from wild type Ras in activating and maintaining the mSOS1- and Raf-1-dependent mitogen-activated protein kinase cascade necessary for neuronal differentiation. However, continuous (>5 days) exposure to nerve growth factor reveals that, in contrast to parental or wild-type Ras-overexpressing PC12 cells, Ras(C118S)-expressing PC12 cells cannot sustain the basal interaction between Ras and phosphatidylinositol 3-kinase. This results in spontaneous apoptosis of these cells despite the presence of nerve growth factor and serum. Thus unique downstream effector interactions and biological outcomes can be differentially modulated by distinct modes of Ras activation.


Subject(s)
Nitric Oxide/physiology , Proto-Oncogene Proteins p21(ras)/physiology , Animals , Cell Survival , Nerve Growth Factor/pharmacology , PC12 Cells , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-raf/physiology , Rats
4.
Biochemistry ; 38(40): 13407-13, 1999 Oct 05.
Article in English | MEDLINE | ID: mdl-10529217

ABSTRACT

Nitric oxide (*NO) is a short-lived free radical with many functions including vasoregulation, synaptic plasticity, and immune modulation and has recently been associated with AIDS pathology. Various pathophysiological conditions, such as viral infection, trigger inducible nitric oxide synthase (iNOS) to synthesize NO in the cell. NO-derived species can react with thiols of proteins and form nitrosothiol adducts. HIV-1 protease (HIV-PR) contains two cysteine residues, Cys67 and Cys95, which are believed to serve a regulatory function. We have found that HIV-PR is inactivated by nitric oxide produced in vitro by NO donors and by iNOS. Sodium nitroprusside inhibited HIV-PR by 70%, and S-nitroso-N-acetylpenicillamine completely inhibited the enzyme. Furthermore, iNOS generated sufficient NO to inhibit HIV-PR activity by almost 90%. This inactivation was reversed by the addition of reducing agents. Treatment of HIV-PR with NO donors and ritonavir (a competitive peptide inhibitor) indicates that NO exerts its effect through a site independent of the active site of HIV-PR. Using electrospray ionization mass spectrometry, we found that NO forms S-nitrosothiols on Cys67 and Cys95 of HIV-PR which directly correlate with a loss of activity. These data indicate that NO may suppress HIV-1 replication by directly inhibiting HIV-PR.


Subject(s)
HIV Protease Inhibitors/chemistry , HIV Protease/metabolism , Nitrates/metabolism , Binding, Competitive , Enzyme Activation/drug effects , Ethylmaleimide/chemistry , HIV Protease/chemistry , HIV Protease Inhibitors/pharmacology , Nitrates/chemistry , Nitric Oxide/chemistry , Nitrosation , Nitroso Compounds/chemistry , Nitroso Compounds/metabolism , Reactive Oxygen Species/metabolism , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Sulfhydryl Compounds/chemistry , Sulfhydryl Compounds/metabolism
5.
Antioxid Redox Signal ; 1(1): 105-12, 1999.
Article in English | MEDLINE | ID: mdl-11225727

ABSTRACT

Nitric oxide (NO) is a naturally occurring free radical with many functions. The oxidized form of NO, the nitrosonium ion, reacts with the thiol group of cysteine residues resulting in their modification to S-nitrosothiols. The human immunodeficiency virus type 1 (HIV-1) protease (HIV-PR) has two cysteine residues that are conserved amongst different viral isolates found in patients with acquired immunodeficiency syndrome (AIDS). In an active dimer, these residues are located near the surface of the protease. We have found that treatment of HIV-PR with different NO congeners results in loss of its proteolytic activity and simultaneous formation of S-nitrosothiols. Sodium nitroprusside inhibited HIV-PR up to 70% and S-nitroso-N-acetylpenicillamine completely inhibited the protease within 5 min of treatment. The pattern of inhibition by NO donors is comparable to its inhibition by N-acetyl pepstatin. Using electrospray ionization-mass spectrometry, we identified the modification of HIV-PR by NO as that of S-nitrosation. Our findings point towards a possible role of NO in mediating resistance to HIV-1 infection.


Subject(s)
Cysteine/metabolism , HIV Protease/metabolism , Nitric Oxide/metabolism , Bacteria/enzymology , Bacteria/genetics , Cysteine/chemistry , Electrophoresis, Polyacrylamide Gel , HIV Protease/chemistry , HIV Protease/isolation & purification , HIV Protease Inhibitors/metabolism , HIV Protease Inhibitors/pharmacology , Humans , Inclusion Bodies/enzymology , Nitric Oxide Donors/pharmacology , Nitroprusside/pharmacology , Penicillamine/analogs & derivatives , Penicillamine/pharmacology , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , S-Nitroso-N-Acetylpenicillamine , Spectrometry, Mass, Electrospray Ionization , Vasodilator Agents/pharmacology
6.
J Biol Chem ; 273(45): 29923-8, 1998 Nov 06.
Article in English | MEDLINE | ID: mdl-9792710

ABSTRACT

Reactive free radical species are known to trigger biochemical events culminating in transcription factor activation and modulation of gene expression. The cytosolic signaling events triggered by free radicals that result in nuclear responses are largely unknown. Here we identify a signaling cascade triggered immediately upon redox activation of Ras. We examined two physiologically relevant models of redox signaling: 1) nitric oxide in human T cells, and 2) advanced glycation end product in rat pheochromocytoma cells. Reactive free radical species generated by nitric oxide donors and the interaction of advanced glycation end product with its receptor led to the recruitment of p85/p110 phosphatidylinositol 3'-kinase (PI3K) to the plasma membrane, where it associated directly with the effector domain of Ras and became activated. Only the p110beta and p110delta (but not p110alpha) catalytic subunits were recruited by redox-activated Ras. Activation of downstream targets of PI3K such as protein kinase B/Akt and mitogen-activated protein kinase was found to be PI3K dependent. Our study demonstrates that nitrosative and oxidative stressors trigger Ras-dependent and PI3K-regulated events in cells and define a biochemical pathway that is triggered by redox signaling.


Subject(s)
Oxidative Stress , Phosphatidylinositol 3-Kinases/metabolism , ras Proteins/metabolism , Animals , Humans , Jurkat Cells , Nitric Oxide/metabolism , Precipitin Tests , Rats , Signal Transduction , Tumor Cells, Cultured
7.
Circ Res ; 81(5): 774-84, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9351450

ABSTRACT

We determined whether local bradykinin production modulates cardiac adrenergic activity. Depolarization of guinea pig heart sympathetic nerve endings (synaptosomes) with 1 to 100 mmol/L K+ caused the release of endogenous norepinephrine (10% to 50% above basal level). This release was exocytotic, because it depended on extracellular Ca2+, was inhibited by the N-type Ca(2+)-channel blocker omega-conotoxin and the protein kinase C inhibitor Ro31-8220, and was potentiated by the neuronal uptake-1 inhibitor desipramine. Typical of adrenergic terminals, norepinephrine exocytosis was enhanced by activation of prejunctional angiotensin AT1-receptors and attenuated by adrenergic alpha 2-receptors, adenosine A1-receptors, and histamine H3-receptors. Exogenous bradykinin enhanced norepinephrine exocytosis by 7% to 35% (EC50, 17 nmol/L), without inhibiting uptake 1. B2-receptor, but not B1-receptor, blockade antagonized this effect. The kininase II/angiotensin-converting enzyme inhibitor enalaprilat and the addition of kininogen or kallikrein enhanced norepinephrine exocytosis by approximately equal to 6% to 40% (EC50, 20 nmol/L) and approximately equal to 25% to 60%, respectively. This potentiation was prevented by serine protease inhibitors and was antagonized by B2-receptor blockade. Therefore, norepinephrine exocytosis is augmented when bradykinin synthesis is increased or when its breakdown is inhibited. This is the first report of a local kallikrein-kinin system in adrenergic nerve endings capable of generating enough bradykinin to activate B2-receptors in an autocrine/paracrine fashion and thus enhance norepinephrine exocytosis. This amplification process may operate in disease states, such as myocardial ischemia, associated with severalfold increases in local kinin concentrations.


Subject(s)
Exocytosis/physiology , Heart/innervation , Heart/physiology , Norepinephrine/physiology , Receptors, Bradykinin/physiology , Sympathetic Nervous System/physiology , Animals , Autocrine Communication , Guinea Pigs , Male , Nerve Endings/physiology , Neuromuscular Junction/physiology , Paracrine Communication
8.
Biochemistry ; 36(31): 9523-31, 1997 Aug 05.
Article in English | MEDLINE | ID: mdl-9235998

ABSTRACT

Mechanisms contributing to altered heterotrimeric G-protein expression and subsequent signaling events during cholesterol accretion have been unexplored. The influence of cholesterol enrichment on G-protein expression was examined in cultured smooth muscle cells that resemble human atherosclerotic cells by exposure to cationized LDL (cLDL). cLDL, which increases cellular free and esterified cholesterol 2-fold and 10-fold, respectively, reduced the cell membrane content of Galphai-1, Galphai-2, Galphai-3, Gq/11, and Galphas. The following evidence supports the premise that the mechanism by which this occurs is due to reduced isoprenylation of the Ggamma-subunit. First, the inhibitory effect of cholesterol enrichment on the membrane content of Galphai subunits was found to be post-transcriptional, since the mRNA steady-state levels of Galphai(1-3) were unchanged following cholesterol enrichment. Second, the membrane expression of alpha and beta subunits was mimicked by cholesterol and 17-ketocholesterol, both of which inhibit HMG-CoA reductase. Third, inhibition of Galphai and Gbeta expression in cholesterol-enriched cells was overcome by mevalonate, the immediate product of HMG-CoA reductase. Fourth, pulse-chase experiments revealed that cholesterol enrichment did not reduce the degradation rate of membrane-associated Galphai subunits. Fifth, cholesterol enrichment also reduced membrane expression of Ggamma-5, Ggamma-7upper; these gamma subunits are responsible for trafficking of the heterotrimeric G-protein complex to the cell membrane as a result of HMG-CoA reductase-dependent post-translational lipid modification (geranylgeranylation) and subsequent membrane association. Cholesterol enrichment did not alter expression of G-gamma-5 mRNA, as assessed by reverse transcriptase polymerase chain reaction, supporting a post-transcriptional defect in Ggamma subunit expression. Fifth, cholesterol enrichment also reduced the membrane content of p21ras (a low molecular weight G-protein requiring farnesylation for membrane targeting) but did not alter the membrane content of the two proteins that do not require isoprenylation for membrane association&sbd;PDGF-receptor or p60-src. Reduced G-protein content in cholesterol-laden cells was reflected by reduced G-protein-mediated signaling events, including ATP-induced GTPase activity, thrombin-induced inhibition of cyclic AMP accumulation, and MAP kinase activity. Collectively, these results demonstrate that cholesterol enrichment reduces G-protein expression and signaling by inhibiting isoprenylation and subsequent membrane targeting. These results provide a molecular basis for altered G-protein-mediated cell signaling processes in cholesterol-enriched cells.


Subject(s)
Cholesterol/metabolism , GTP-Binding Proteins/metabolism , Mitogen-Activated Protein Kinases , Muscle, Smooth, Vascular/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Membrane/metabolism , Cells, Cultured , Humans , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/enzymology , Oncogene Protein pp60(v-src)/metabolism , Oxygen/metabolism , Protein Prenylation , Protein-Tyrosine Kinases/metabolism , RNA, Messenger/metabolism , Rabbits , Rats , Receptors, Platelet-Derived Growth Factor/metabolism , Steroids/metabolism
9.
Biochemistry ; 36(31): 9532-9, 1997 Aug 05.
Article in English | MEDLINE | ID: mdl-9235999

ABSTRACT

PGI2 generation by the vessel wall is an agonist for cyclic-AMP-dependent cholesteryl ester hydrolysis. The process of enhanced PGI2 synthesis is stimulated, in part, by G-protein-coupled receptor ligands. Cellular cholesterol enrichment has been hypothesized to alter G-protein-mediated PGI2 synthesis. In the studies reported herein, cells generated PGI2 in response to AlF4-, GTPgammaS, and ATP in a dose-dependent manner. G-protein agonists stimulated eicosanoid production principally by activating phospholipase A2, but not phospholipase C. This is in contrast to PDGF, which stimulated phospholipase A2 and PLCgamma activities. Galphai subunits mediate G-protein agonist-induced PGI2 synthesis, since ATP- and PDGF-induced PGI2 synthesis was inhibited by pertussis toxin. Although cholesterol enrichment reduced arachidonic acid- and PDGF-induced PGI2 synthesis, cholesterol enrichment enhanced PGI2 release in response to AlF4-, GTPgammaS, and ATP. The enhancement of PGI2 release in cholesterol-enriched cells was augmented by mevalonate, which inhibits the ability of cholesterol enrichment to reduce membrane-associated G-protein subunits. Since cholesterol enrichment inhibited PDGF and AlF4--induced MAP kinase activity [Pomerantz, K., Lander, H. M., Summers, B., Robishaw, J. D., Balcueva, E. A., & Hajjar, D. P. (1997) Biochemistry 36, 9523-9531] (the major mechanism by which phospholipase A2 is activated), these results suggest that cholesterol enrichment induces other alternative signaling pathways leading to phospholipase A2 activation. A PKC-dependent pathway is described herein that is involved in enhanced eicosanoid production in cholesterol-enriched cells. This conclusion is supported by two observations: (1) G-protein-linked PGI2 production is inhibited by calphostin, and (2) cholesterol enrichment augments the specific translocation of the delta-isoform of PKC from the cytosol to the plasma membrane following treatment of cells with phorbol ester. These data support the concept that, in cells possessing normal levels of cholesterol, MAP-kinase-dependent pathways mediate eicosanoid synthesis in response to G-protein activation; however, under conditions of high cellular cholesterol levels, augmented G-protein-linked eicosanoid production results from enhanced PKCdelta activity.


Subject(s)
Cholesterol/metabolism , Epoprostenol/biosynthesis , GTP-Binding Proteins/metabolism , Isoenzymes/metabolism , Muscle, Smooth, Vascular/metabolism , Protein Kinase C/metabolism , Signal Transduction , Animals , Biological Transport , Cells, Cultured , Humans , Mevalonic Acid/pharmacology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/enzymology , Pertussis Toxin , Phospholipases A/metabolism , Phospholipases A2 , Protein Kinase C-delta , Rabbits , Rats , Tetradecanoylphorbol Acetate/pharmacology , Virulence Factors, Bordetella/pharmacology
10.
J Biol Chem ; 272(28): 17810-4, 1997 Jul 11.
Article in English | MEDLINE | ID: mdl-9211935

ABSTRACT

Advanced glycation end products (AGEs) exert their cellular effects on cells by interacting with specific cellular receptors, the best characterized of which is the receptor for AGE (RAGE). The transductional processes by which RAGE ligation transmits signals to the nuclei of cells is unknown and was investigated. AGE-albumin, a prototypic ligand, activated p21(ras) in rat pulmonary artery smooth muscle cells that express RAGE, whereas nonglycated albumin was without effect. MAP kinase activity was enhanced at concentrations of AGE-albumin, which activated p21(ras) and NF-kappaB. Depletion of intracellular glutathione rendered cells more sensitive to AGE-mediated activation of this signaling pathway. In contrast, signaling was blocked by preventing p21(ras) from associating with the plasma membrane or mutating Cys118 on p21(ras) to Ser. Signaling was receptor-dependent, because it was prevented by blocking access to RAGE with either anti-RAGE IgG or by excess soluble RAGE. These data suggest that RAGE-mediated induction of cellular oxidant stress triggers a cascade of intracellular signals involving p21(ras) and MAP kinase, culminating in transcription factor activation. The molecular mechanism that triggers this pathway likely involves oxidant modification and activation of p21(ras).


Subject(s)
Alkyl and Aryl Transferases , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Glycation End Products, Advanced/metabolism , Oxidative Stress , Transferases/metabolism , Animals , Enzyme Activation , Glutathione/metabolism , PC12 Cells , Rats , Receptor for Advanced Glycation End Products , Receptors, Immunologic/metabolism , Signal Transduction
11.
FASEB J ; 11(2): 118-24, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9039953

ABSTRACT

It is well accepted that extracellular ligands trigger nuclear signals through a cascade of protein-protein interactions. Many of these pathways have been carefully defined and provide an important framework by which we can understand and intervene in the processes they initiate. Recent data in the literature indicate that many extracellular ligands generate and/or require reactive free radicals or derived species to successfully transmit their signals to the nucleus. Thus, a novel signaling mechanism akin to one solely dependent on protein-protein interactions may exist. Here, we review this information, identify both the sources and targets of free radicals generated by various growth factors and cytokines, discuss how specificity can be achieved, and explore the pathophysiological implications.


Subject(s)
Reactive Oxygen Species/physiology , Signal Transduction/drug effects , Animals , Free Radicals , Humans , Nitrogen/physiology , Signal Transduction/physiology
12.
J Biol Chem ; 272(7): 4323-6, 1997 Feb 14.
Article in English | MEDLINE | ID: mdl-9020151

ABSTRACT

We have identified the site of molecular interaction between nitric oxide (NO) and p21(ras) responsible for initiation of signal transduction. We found that p21(ras) was singly S-nitrosylated and localized this modification to a fragment of p21(ras) containing Cys118. A mutant form of p21(ras), in which Cys118 was changed to a serine residue and termed p21(ras)C118S, was not S-nitrosylated. NO-related species stimulated guanine nucleotide exchange on wild-type p21(ras), resulting in an active form, but not on p21(ras)C118S. Furthermore, in contrast to parental Jurkat T cells, NO-related species did not stimulate mitogen-activated protein kinase activity in cells transfected with p21(ras)C118S. These data indicate that Cys118 is a critical site of redox regulation of p21(ras), and S-nitrosylation of this residue triggers guanine nucleotide exchange and downstream signaling.


Subject(s)
Nitric Oxide/chemistry , Oncogene Protein p21(ras)/chemistry , Amino Acid Sequence , DNA, Complementary , Humans , Jurkat Cells , Molecular Sequence Data , Nitric Oxide/metabolism , Oncogene Protein p21(ras)/genetics , Oncogene Protein p21(ras)/metabolism , Oxidation-Reduction , Signal Transduction , Transfection
13.
J Biol Chem ; 271(33): 19705-9, 1996 Aug 16.
Article in English | MEDLINE | ID: mdl-8702674

ABSTRACT

Many studies have identified nitric oxide (NO) and related chemical species (NOx) as having critical roles in neurotransmission, vasoregulation, and cellular signaling. Previous work in this laboratory has focused on elucidating the mechanism of NOx signaling in cells. We have demonstrated that NOx-induced activation of the guanine nucleotide-binding protein p21(ras) leads to nuclear translocation of the transcription factor NFkappaB. Here, we investigated whether intermediary signaling elements, namely the mitogen-activated protein (MAP) kinases, are involved in mediating NOx signaling. We found that NOx activates the extracellular signal-regulated kinase (ERK), p38, and c-Jun NH2-terminal kinase (JNK) subgroups of MAP kinases in human Jurkat T cells. JNK was found to be 100-fold more sensitive to NOx stimulation than p38 and ERK. In addition, the activation of JNK and p38 by NOx was more rapid than ERK activation. Depletion of intracellular glutathione augmented the NOx-induced increase in kinase activity. Furthermore, endogenous NO, generated from NO synthase, activated ERK, and NOx-induced MAP kinase activation was effectively blocked by the farnesyl transferase inhibitor alpha-hydroxyfarnesylphosphonic acid. These data support the hypothesis that critical signaling kinases, such as ERK, p38, and JNK, are activated by NO-related species and thus participate in NO signal transduction. These findings establish a role for multiple MAP kinase signaling pathways in the cellular response to NOx.


Subject(s)
Alkyl and Aryl Transferases , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Mitogen-Activated Protein Kinases , Nitric Oxide/chemistry , T-Lymphocytes/physiology , Enzyme Activation , Farnesyltranstransferase , GTP-Binding Proteins/physiology , Humans , Hydrogen Peroxide/metabolism , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Nitric Oxide/physiology , Nitrogen Oxides/pharmacology , Nitroprusside/metabolism , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Signal Transduction , Transferases/antagonists & inhibitors , Tumor Cells, Cultured , p38 Mitogen-Activated Protein Kinases
15.
Circ Res ; 78(3): 475-81, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8593706

ABSTRACT

We previously showed that prejunctional histamine H3-receptors downregulate norepinephrine exocytosis, which is markedly enhanced in early myocardial ischemia. In the present study, we investigated whether H3-receptors modulate nonexocytotic norepinephrine release during protracted myocardial ischemia. In this setting, decreased pH(i) in sympathetic nerve endings sequentially leads to a compensatory activation of the Na+-H+ antiporter (NHE), accumulation of intracellular Na+, reversal of the neuronal uptake of norepinephrine, and thus carrier-mediated release of norepinephrine. Accordingly, norepinephrine overflow from isolated guinea pig hearts undergoing 20-minute global ischemia and 45-minute reperfusion was attenuated approximately 80% by desipramine (10 nmol/L) and 70% by 5-(N-ethyl-N-isopropyl)-amiloride (EIPA, 10 micromol/L), inhibitors of norepinephrine uptake and NHE, respectively. The H3-receptor agonist imetit (0.1 micromol/L) decreased carrier-mediated norepinephrine release by approximately 50%. This effect was blocked by the H3-receptor antagonist thioperamide (0.3 micromol/L), indicating that H-receptor activation inhibits carrier-mediated norepinephrine release. At lower concentrations, imetit (10 nmol/L) or EIPA (3 micromol/L) did not inhibit carrier-mediated norepinephrine release. However, a 25% inhibition occurred with imetit (10 nmol/L) and EIPA (3 micromol/L) combined. This synergism suggests an association between H-receptors and NHE. Conceivably, activation of H-receptors may lead to inhibition of NHE. In fact, alpha2-adrenoceptor activation, which is known to stimulate NHE, enhanced norepinephrine release, whereas alpha2-adrenoceptor blockade attenuated it. Furthermore, activation of adenosine A1-receptors markedly attenuated norepinephrine release, whereas their inhibition potentiated it. Because norepinephrine directly correlated with the severity of reperfusion arrhythmia and imetit reduced the incidence of ventricular fibrillation by 50%, our findings with H-receptor agonists may further the development of novel pharmacological means to reduce reperfusion arrhythmias in the clinical setting.


Subject(s)
Myocardial Ischemia/metabolism , Myocardium/metabolism , Norepinephrine/metabolism , Receptors, Adrenergic, alpha-2/metabolism , Receptors, Histamine H3/metabolism , Receptors, Purinergic P1/metabolism , Adrenergic alpha-2 Receptor Agonists , Adrenergic alpha-2 Receptor Antagonists , Animals , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/metabolism , Guinea Pigs , Histamine/metabolism , Histamine Agonists/pharmacology , In Vitro Techniques , Ligands , Male , Myocardial Reperfusion Injury/complications , Myocardial Reperfusion Injury/metabolism , Neurotransmitter Uptake Inhibitors/pharmacology , Purinergic P1 Receptor Agonists
16.
J Biol Chem ; 270(36): 21195-8, 1995 Sep 08.
Article in English | MEDLINE | ID: mdl-7673152

ABSTRACT

Reactive free radicals have been implicated in mediating signal transduction by a variety of stimuli. We have investigated the role of p21ras in mediating free radical signaling. Our studies revealed that signaling by oxidative agents which modulate cellular redox status, such as H2O2, hemin, Hg2+, and nitric oxide was prevented in cells in which p21ras activity was blocked either through expression of a dominant negative mutant or by treating with a farnesyltransferase inhibitor, as assessed by NF-kappa B binding activity. Furthermore, the NF-kappa B response to these oxidative stress stimuli was found to be enhanced when cells from the human T cell line, Jurkat, were pretreated with L-buthionine-(S,R)-sulfoximine, an inhibitor of glutathione synthesis. We directly assayed p21ras and mitogen-activated protein kinase activities in Jurkat cells and found both of these signaling molecules to be activated in cells treated with the redox modulating agents. Blocking glutathione synthesis made cells 10- to 100-fold more sensitive to these agents. Finally, using recombinant p21ras in vitro, we found that redox modulators directly promoted guanine nucleotide exchange on p21ras. This study suggests that direct activation of p21ras may be a central mechanism by which a variety of redox stress stimuli transmit their signal to the nucleus.


Subject(s)
Oncogene Protein p21(ras)/metabolism , Oxidative Stress , Signal Transduction , Animals , Buthionine Sulfoximine , Cell Line , Enzyme Activation , Free Radicals , Glutamate-Cysteine Ligase/antagonists & inhibitors , Humans , Methionine Sulfoximine/analogs & derivatives , Methionine Sulfoximine/pharmacology , NF-kappa B/metabolism , Oxidation-Reduction , PC12 Cells , Protein Kinases/metabolism , Rats
17.
Circ Res ; 77(1): 206-10, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7788879

ABSTRACT

Norepinephrine release contributes to ischemic cardiac dysfunction and arrhythmias. Because activation of histamine H3-receptors inhibits norepinephrine release, we searched for the presence of H3-receptors directly in sympathetic nerve endings (cardiac synaptosomes) isolated from surgical specimens of human atria. Norepinephrine was released by depolarization with K+. The presence of H3-receptors was ascertained because the selective H3-receptor agonists (R) alpha-methylhistamine and imetit reduced norepinephrine release, and the specific H3-receptor antagonist thioperamide blocked this effect. Norepinephrine release was exocytotic, since it was inhibited by the N-type Ca(2+)-channel blocker omega-conotoxin and the protein kinase C inhibitor Ro31-8220. Functional relevance of these H3-receptors was obtained by showing that transmural electrical stimulation of sympathetic nerve endings in human atrial tissue increased contractility, an effect blocked by propranolol and attenuated in a concentration-dependent manner by (R) alpha-methylhistamine. Also, thioperamide antagonized the effect of (R) alpha-methylhistamine. Our findings are the first demonstration that H3-receptors are present in sympathetic nerve endings in the human heart, where they modulate adrenergic responses by inhibiting norepinephrine release. Since myocardial ischemia causes intracardiac histamine release, H3-receptor-induced attenuation of sympathetic neurotransmission may be clinically relevant.


Subject(s)
Heart/physiology , Myocardium/chemistry , Receptors, Histamine H3/analysis , Receptors, Histamine H3/physiology , Synaptosomes/chemistry , Synaptosomes/physiology , Cell Separation , Electric Stimulation , Exocytosis , Histamine Agonists/pharmacology , Histamine Antagonists , Humans , Imidazoles/pharmacology , In Vitro Techniques , Indoles/pharmacology , Methylhistamines/pharmacology , Myocardial Ischemia/physiopathology , Norepinephrine/metabolism , Piperidines/pharmacology , Protein Kinase C/antagonists & inhibitors , Receptors, Histamine H3/drug effects , Synaptosomes/metabolism , Thiourea/analogs & derivatives , Thiourea/pharmacology
18.
J Biol Chem ; 270(29): 17185-8, 1995 Jul 21.
Article in English | MEDLINE | ID: mdl-7615515

ABSTRACT

Recent studies have demonstrated the biological importance of the interaction of nitric oxide (NO) with proteins. Protein-associated targets of NO include heme, Cys, and Tyr. Electrospray ionization-mass spectrometry was used to monitor the results of exposure of model peptides and an enzyme to NO under different conditions and thus addressed aspects of NO-protein interactions. The molecular mass of a decapeptide containing a single Cys residue increased by 29 Da upon treatment with NO under aerobic and acidic conditions, consistent with the substitution of one NO moiety. The mass of reduced somatostatin, a peptide containing two Cys residues, increased by 58 Da, consistent with the substitution of two NO moieties. These substitutions were prevented by pretreatment of the peptides with N-ethylmaleimide. The strength of the nitrosothiol bond was examined by varying the amount of energy applied to the peptide ions and indicated a labile species. Cys residues were very rapidly nitrosated, while other reactions were observed to occur at much slower rates. These include the further oxidation of nitrosothiol to sulfonic acid and nitration of Tyr. Peptides treated with NO at physiological pH were observed to undergo dimerization as well as nitrosation. These studies were extended to the enzyme p21ras, whose activity has been postulated to be modulated by nitrosothiol formation, and revealed the formation of a single nitrosothiol on p21ras upon NO treatment. These data suggest that electrospray ionization-mass spectrometry allows for quantitation and characterization of nitrosothiol bonds in peptides and proteins.


Subject(s)
Mercaptoethanol , Nitric Oxide/metabolism , Nitroso Compounds/chemistry , Proteins/metabolism , S-Nitrosothiols , Amino Acid Sequence , Kinetics , Mass Spectrometry , Molecular Sequence Data , Proto-Oncogene Proteins p21(ras)/metabolism , Somatostatin/metabolism
19.
J Biol Chem ; 270(13): 7017-20, 1995 Mar 31.
Article in English | MEDLINE | ID: mdl-7706235

ABSTRACT

The protooncogene p21ras, a monomeric G protein family member, plays a critical role in converting extracellular signals into intracellular biochemical events. Here, we report that nitric oxide (NO) activates p21ras in human T cells as evidenced by an increase in GTP-bound p21ras. In vitro studies using pure recombinant p21ras demonstrate that the activation is direct and reversible. Circular dichroism analysis reveals that NO induces a profound conformational change in p21ras in association with GDP/GTP exchange. The mechanism of activation is due to S-nitrosylation of a critical cysteine residue which stimulates guanine nucleotide exchange. Furthermore, we demonstrate that p21ras is essential for NO-induced downstream signaling, such as NF-kappa B activation, and that endogenous NO can activate p21ras in the same cell. These studies identify p21ras as a target of the same cell. These studies identify p21ras as a target of NO in T cells and suggest that NO activates p21ras by an action which mimics that of guanine nucleotide exchange factors.


Subject(s)
GTP-Binding Proteins/metabolism , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , Nitric Oxide/pharmacology , Proto-Oncogene Proteins p21(ras)/metabolism , Antibodies/pharmacology , Carbon Monoxide/pharmacology , Cell Line , GTP Phosphohydrolases/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Hemoglobins/pharmacology , Humans , Kinetics , NF-kappa B/metabolism , Nitric Oxide/physiology , Signal Transduction , Sulfur Radioisotopes , T-Lymphocytes , Tritium , Tumor Cells, Cultured
20.
J Leukoc Biol ; 57(1): 174-9, 1995 Jan.
Article in English | MEDLINE | ID: mdl-7829969

ABSTRACT

The effects of bacterial lipopolysaccharide (LPS) on macrophage gene expression are mediated in part by its ability to induce activation of transcription factor NF-kappa B. We compared the ability of LPS-treated macrophages from Lpsn (LPS-responsive) C3H/HeN and Lpsd (LPS-hyporesponsive) C3H/HeJ mice to mobilize NF-kappa B by electrophoretic mobility shift assays with oligonucleotide probes containing a unique NF-kappa B sequence from the promoter of inducible nitric oxide synthase (iNOS). In response to ng/ml concentrations of LPS, this probe bound proteins that appeared rapidly in the nuclei of thioglycollate-elicited macrophages and bone marrow-derived macrophage cell lines from both Lpsn and Lpsd mice. Only in macrophages from Lpsn mice, however, was LPS able to induce iNOS or tumor necrosis factor alpha. NF-kappa B-containing DNA-protein complexes from Lpsd macrophages were formed in lesser amounts than from Lpsn macrophages but shared the same composition, insofar as they displayed the same electrophoretic mobilities and content of heterodimers of p50/RelA (p65) and p50/c-rel. Two conclusions emerge from these findings: (1) NF-kappa B activity alone is not sufficient for induction of certain LPS-responsive genes and (2) An LPS-response pathway involving activation of NF-kappa B is preserved in Lpsd mice. The inability of cells from Lpsd mice to induce gene expression in response to LPS thus cannot be attributed to inability to activate NF-kappa B.


Subject(s)
Gene Expression Regulation/drug effects , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/drug effects , Mice, Inbred C3H/genetics , NF-kappa B/physiology , Animals , Base Sequence , Bone Marrow Cells , Cell Line , Cells, Cultured , DNA/analysis , DNA/genetics , Female , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/metabolism , Mice , Molecular Sequence Data , NF-kappa B/genetics , NF-kappa B/metabolism , Nitrites/metabolism , Oligonucleotide Probes/chemistry , Protein Binding , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...