Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Phytomedicine ; 14(5): 344-52, 2007 May.
Article in English | MEDLINE | ID: mdl-17084601

ABSTRACT

NO-synthase (NOS) is a heme-containing enzyme that catalyzes the oxidation of L-arginine to nitric oxide, an important cellular signaling molecule. Recently, it was found that aqueous extracts of tobacco cigarettes cause the inactivation of the neuronal isoform of NOS (nNOS) and that this may explain some of the toxicological effects of smoking. Although the exact identity of the chemical inactivator(s) is not known, we wondered if extracts prepared from other plants, including those closely related to tobacco, Nicotiana tabacum (Solanaceae), would similarly inactivate nNOS. We examined 33 plants, representing diverse members of the plant kingdom ranging from whisk fern, Psilotum nudum (Psilotaceae) to tobacco and discovered 18 plants that contain a chemical inactivator(s) of nNOS. Of these plants, 16 are members of the core asterids flowering plant group. Of these asterids, 6 are members of the Solanaceae family, of which tobacco is a member. Based on the phylogenetic relationship of the plants, it is possible that the same chemical or related chemical inactivator(s) exist. This, in turn, may help elucidate the structure of the chemical(s), as well as provide a source of a potentially novel inactivator of nNOS. The alkaloid nicotine can be excluded as putative nNOS inhibitor.


Subject(s)
Enzyme Inhibitors/pharmacology , Nicotiana , Nitric Oxide Synthase Type I/antagonists & inhibitors , Phytotherapy , Plant Extracts/pharmacology , Chromatography, High Pressure Liquid , Enzyme Inhibitors/chemistry , Humans , Inhibitory Concentration 50 , Nitric Oxide Synthase Type I/chemistry , Plant Components, Aerial , Plant Extracts/chemistry
2.
Drug Metab Dispos ; 34(9): 1448-56, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16738031

ABSTRACT

It is established that guanabenz inhibits neuronal nitric-oxide (NO) synthase (nNOS) and causes the enhanced proteasomal degradation of nNOS in vivo. Although the time- and NADPH-dependent inhibition of nNOS has been reported in studies where guanabenz was incubated with crude cytosolic preparations of nNOS, the exact mechanism for inhibition is not known. Moreover, even less is known about how the inhibition of nNOS triggers its proteasomal degradation. In the current study, we show, with the use of purified nNOS, that guanabenz treatment leads to the oxidation of tetrahydrobiopterin and formation of a pterin-depleted nNOS, which is not able to form NO. With the use of 14C-labeled guanabenz, we were unable to detect any guanabenz metabolites or guanabenz-nNOS adducts, indicating that reactive intermediates of guanabenz probably do not play a role in the inhibition. Superoxide dismutase, however, prevents the guanabenz-mediated oxidation of tetrahydrobiopterin and inhibition of nNOS, suggesting the role of superoxide as an intermediate. Studies in rats show that administration of tetrahydrobiopterin prevents the inhibition and loss of penile nNOS due to guanabenz, indicating that the loss of tetrahydrobiopterin plays a major role in the effects of guanabenz in vivo. Our findings are consistent with the destabilization and enhanced degradation of nNOS found after tetrahydrobiopterin depletion. These studies suggest that drug-mediated destabilization and subsequent enhanced degradation of protein targets will likely be an important toxicological consideration.


Subject(s)
Biopterins/analogs & derivatives , Enzyme Inhibitors/pharmacology , Guanabenz/pharmacology , Nitric Oxide Synthase Type I/antagonists & inhibitors , Nitric Oxide Synthase/antagonists & inhibitors , Penis/drug effects , Animals , Biopterins/chemistry , Biopterins/metabolism , Biopterins/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Stability , Male , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type I/chemistry , Nitric Oxide Synthase Type I/metabolism , Oxidation-Reduction , Penis/enzymology , Rats , Rats, Wistar , Superoxide Dismutase/chemistry , Superoxide Dismutase/metabolism , Time Factors , Ubiquitin/metabolism
3.
Brain Res Mol Brain Res ; 142(1): 19-27, 2005 Dec 07.
Article in English | MEDLINE | ID: mdl-16216381

ABSTRACT

Tetrahydrobiopterin is a necessary cofactor for the synthesis of nitric oxide by the hemeprotein enzyme, NO-synthase (NOS). It is widely thought that inadequate levels of tetrahydrobiopterin lead to tissue injury and organ dysfunction due, in part, to formation of superoxide from pterin-deficient NOS. In the course of studies on the ubiquitylation of neuronal NOS (nNOS), we have found that certain substrate analogs, such as N(G)-nitro-L-arginine, stabilize the dimeric form of nNOS and protect the enzyme from ubiquitylation. Since tetrahydrobiopterin is known to bind near heme and confers stability to the active dimeric structure of nNOS, we wondered if the loss of tetrahydrobiopterin could be an endogenous signal for nNOS ubiquitylation and degradation. We show here in HEK293 cells stably transfected with nNOS that depletion of tetrahydrobiopterin by treatment with 2,4-diamino-6-hydroxypyrimidine leads to destabilization of the dimeric form and enhances ubiquitylation of nNOS. Sepiapterin, a precursor to tetrahydrobiopterin in the salvage pathway, completely reverses the effect of 2,4-diamino-6-hydroxypyrimidine on nNOS ubiquitylation. Consistent with that found in cells, the in vitro ubiquitylation of nNOS by reticulocyte proteins decreases when tetrahydrobiopterin is present. Thus, inadequate amounts of tetrahydrobiopterin may lead to a sustained decrease in the steady state level of nNOS that is not readily reversed.


Subject(s)
Biopterins/analogs & derivatives , Nitric Oxide Synthase Type I/metabolism , Ubiquitins/metabolism , Biopterins/metabolism , Biopterins/pharmacology , Blotting, Western/methods , Cell Line , Chromatography, High Pressure Liquid/methods , Dimerization , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Heme/metabolism , Humans , Hypoxanthines/pharmacology , Immunoprecipitation/methods , Leupeptins/pharmacology , Nitric Oxide Synthase Type I/chemistry , Pterins/pharmacology , Time Factors
4.
Drug Metab Dispos ; 33(1): 131-8, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15470159

ABSTRACT

Smoking causes a dysfunction in endothelial nitric-oxide synthase (eNOS), which is ameliorated, in part, by administration of tetrahydrobiopterin (BH(4)). The exact mechanism by which the nitric oxide deficit occurs is unknown. We have previously shown that aqueous extracts of chemicals in cigarettes (CE) cause the suicide inactivation of neuronal NO synthase (nNOS) by interacting at the substrate-binding site. In the current study, we have found that CE directly inactivates eNOS by a process that is not affected by the natural substrate l-arginine and is distinct from the mechanism of inactivation of nNOS. We discovered that CE causes a time-, concentration-, and NADPH-dependent inactivation of eNOS in an in vitro system containing the purified enzyme, indicating a metabolic component to the inactivation. The CE-treated eNOS but not nNOS was nearly fully reactivated upon incubation with excess BH(4), suggesting that BH(4) depletion is a potential mechanism of inactivation. Moreover, in the presence of CE, eNOS catalyzed the oxidation of BH(4) to dihydrobiopterin and biopterin by a process attenuated by high concentrations of superoxide dismutase but not catalase. We speculate that a redox active component in CE, perhaps a quinone compound, causes oxidative uncoupling of eNOS to form superoxide, which in turn oxidizes BH(4). The discovery of a direct inactivation of eNOS by a compound(s) present in tobacco provides a basis not only for further study of the mechanisms responsible for the biological effects of tobacco but also a search for a potentially novel inactivator of eNOS.


Subject(s)
Biopterins/analogs & derivatives , Biopterins/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Tars/pharmacology , Animals , Dose-Response Relationship, Drug , Enzyme Inhibitors/isolation & purification , Enzyme Inhibitors/pharmacology , Insecta , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type III , Rats , Smoking/metabolism , Tars/isolation & purification , Time Factors
5.
J Biol Chem ; 279(51): 52970-7, 2004 Dec 17.
Article in English | MEDLINE | ID: mdl-15466472

ABSTRACT

It is established that neuronal nitric-oxide synthase (nNOS) is ubiquitylated and proteasomally degraded. The proteasomal degradation of nNOS is enhanced by suicide inactivation of nNOS or by the inhibition of hsp90, which is a chaperone found in a native complex with nNOS. In the current study, we have examined whether CHIP, a chaperone-dependent E3 ubiquitin-protein isopeptide ligase that is known to ubiquitylate other hsp90-chaperoned proteins, could act as an ubiquitin ligase for nNOS. We found with the use of HEK293T or COS-7 cells and transient transfection methods that CHIP overexpression causes a decrease in immunodetectable levels of nNOS. The extent of the loss of nNOS is dependent on the amount of CHIP cDNA used for transfection. Lactacystin (10 microM), a selective proteasome inhibitor, attenuates the loss of nNOS in part by causing the nNOS to be found in a detergent-insoluble form. Immunoprecipitation of the nNOS and subsequent Western blotting with an anti-ubiquitin IgG shows an increase in nNOS-ubiquitin conjugates because of CHIP. Moreover, incubation of nNOS with a purified system containing an E1 ubiquitin-activating enzyme, an E2 ubiquitin carrier protein conjugating enzyme (UbcH5a), CHIP, glutathione S-transferase-tagged ubiquitin, and an ATP-generating system leads to the ubiquitylation of nNOS. The addition of purified hsp70 and hsp40 to this in vitro system greatly enhances the amount of nNOS-ubiquitin conjugates, suggesting that CHIP is an E3 ligase for nNOS whose action is facilitated by (and possibly requires) its interaction with nNOS-bound hsp70.


Subject(s)
Acetylcysteine/analogs & derivatives , HSP90 Heat-Shock Proteins/metabolism , Nitric Oxide Synthase/metabolism , Ubiquitin-Protein Ligases/physiology , Ubiquitin/metabolism , Acetylcysteine/metabolism , Adenosine Triphosphate/metabolism , Animals , Blotting, Western , COS Cells , Cell Line , Cysteine Proteinase Inhibitors/pharmacology , DNA, Complementary/metabolism , Detergents/pharmacology , Dose-Response Relationship, Drug , Glutathione Transferase/metabolism , HSP70 Heat-Shock Proteins/metabolism , Humans , Immunoglobulin G/chemistry , Immunoprecipitation , Lactones/pharmacology , Macrolides , Nitric Oxide Synthase Type I , Palmitic Acids/metabolism , Proteasome Inhibitors , Protein Structure, Tertiary , Rabbits , Rats , Time Factors , Transfection
6.
Mol Pharmacol ; 66(4): 964-9, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15235101

ABSTRACT

It is established that neuronal NO synthase (nNOS) is ubiquitinated and proteasomally degraded. The metabolism-based inactivation of nNOS and the inhibition of heat shock protein 90 (hsp90)-based chaperones, which are known to regulate nNOS, both lead to enhanced proteasomal degradation of nNOS. The mechanism of this selective proteolytic degradation, or in essence how the nNOS becomes labilized and recognized for ubiquitination and subsequent degradation, has not been determined. In the current study, we used a crude preparation of reticulocyte proteins, which contains ubiquitin-conjugating enzymes and the proteasome, to determine how nNOS is labilized. We found that the inactive monomeric heme-deficient nNOS (apo-nNOS) is rapidly degraded in vitro, consistent with the finding that both metabolism-based inactivation and inhibition of hsp90-based chaperones cause the formation of apo-nNOS and enhance its degradation in vivo. In the current study, we discovered that destabilization of the dimeric nNOS, as determined by measuring the SDS-resistant dimer, is sufficient to trigger ubiquitin-proteasomal degradation. Treatment of nNOS with NG-nitro-L-arginine or 7-nitroindazole led to stabilization of the dimeric nNOS and decreased proteasomal degradation of the enzyme, consistent with that observed in cells. Thus, it seems that the dimeric structure is a major determinant of nNOS stability and proteolysis.


Subject(s)
Nitric Oxide Synthase/metabolism , Ubiquitin/metabolism , Animals , Biodegradation, Environmental , Dimerization , Enzyme Stability , Heme/metabolism , Nitric Oxide Synthase Type I , Peptide Hydrolases/metabolism , Proteasome Endopeptidase Complex/metabolism , Rabbits
7.
J Biol Chem ; 279(29): 30252-8, 2004 Jul 16.
Article in English | MEDLINE | ID: mdl-15155759

ABSTRACT

Like other nitric-oxide synthase (NOS) enzymes, neuronal NOS (nNOS) turnover and activity are regulated by the ubiquitous protein chaperone hsp90. We have shown previously that nNOS expressed in Sf9 cells where endogenous heme levels are low is activated from the apo- to the holo-enzyme by addition of exogenous heme to the culture medium, and this activation is inhibited by radicicol, a specific inhibitor of hsp90 (Billecke, S. S., Bender, A. T., Kanelakis, K. C., Murphy, P. J. M., Lowe, E. R., Kamada, Y., Pratt, W. B., and Osawa, Y. (2002) J. Biol. Chem. 278, 15465-15468). In this work, we examine heme binding by apo-nNOS to form the active enzyme in a cell-free system. We show that cytosol from Sf9 cells facilitates heme-dependent apo-nNOS activation by promoting functional heme insertion into the enzyme. Sf9 cytosol also converts the glucocorticoid receptor (GR) to a state where the hydrophobic ligand binding cleft is open to access by steroid. Both cell-free heme activation of purified nNOS and activation of steroid binding activity of the immunopurified GR are inhibited by radicicol treatment of Sf9 cells prior to cytosol preparation, and addition of purified hsp90 to cytosol partially overcomes this inhibition. Although there is an hsp90-dependent machinery in Sf9 cytosol that facilitates heme binding by apo-nNOS, it is clearly different from the machinery that facilitates steroid binding by the GR. hsp90 regulation of apo-nNOS heme activation is very dynamic and requires higher concentrations of radicicol for its inhibition, whereas GR steroid binding is determined by assembly of stable GR.hsp90 heterocomplexes that are formed by a purified five-chaperone machinery that does not activate apo-nNOS.


Subject(s)
HSP90 Heat-Shock Proteins/physiology , Heme/chemistry , Nitric Oxide Synthase/chemistry , Animals , Blotting, Western , Cell Line , Cell-Free System , Culture Media , Cytochrome P-450 Enzyme System/metabolism , Cytosol/metabolism , Dose-Response Relationship, Drug , Enzyme Activation , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Insecta , Lactones/pharmacology , Ligands , Macrolides , Molecular Chaperones/metabolism , Nitric Oxide Synthase Type I , Protein Binding , Protein Structure, Tertiary , Rabbits , Receptors, Glucocorticoid/metabolism , Subcellular Fractions , Time Factors
8.
Drug Metab Dispos ; 31(7): 932-7, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12814971

ABSTRACT

It has been shown that administration of cigarette smoke to rats leads to loss of neuronal nitric-oxide synthase (nNOS) activity and nNOS protein in penile tissue. The exact mechanism for this loss of activity and protein is not known. In the current study, we investigated whether extracts prepared from cigarette smoke or from the cigarette itself could directly inhibit nNOS activity. We discovered that the cigarette smoke extract and the cigarette extract cause a time-, concentration-, and calmodulin-dependent inactivation of nNOS in an in vitro system containing the purified enzyme. L-Arginine, but not D-arginine, protects nNOS from this time-dependent inactivation, suggesting an active site directed event. The kinetics of inactivation are consistent with the metabolism-based or suicide inactivation of nNOS. Based on studies with other metabolism-based inactivators, this cigarette-mediated inactivation may render nNOS more susceptible to proteasomal degradation and thereby may explain the loss of nNOS protein in vivo. The component(s) responsible for nNOS inactivation is not volatile, is not retained by a 3,000 molecular weight cut-off membrane, binds to activated charcoal, and is highly water-soluble under both acidic and basic conditions. The discovery of a direct inactivation of nNOS by an organic, cationic compound(s) present in tobacco and tobacco smoke provides a basis for further study of not only the mechanisms responsible for the biological effects of tobacco but also a search for a potentially novel inactivator of nNOS.


Subject(s)
Arginine/analogs & derivatives , Nicotiana/chemistry , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Smoke/analysis , Arginine/metabolism , Calmodulin/metabolism , Enzyme Inhibitors/metabolism , NADP/metabolism , Nitric Oxide Synthase/drug effects , Nitric Oxide Synthase Type I , Nitric Oxide Synthase Type III , Solubility , Water/analysis , Water/pharmacology
9.
J Pharmacol Exp Ther ; 304(2): 493-7, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12538799

ABSTRACT

Nitric oxide synthase (NOS) is a highly regulated enzyme that produces nitric oxide, a critical messenger in many physiological processes. In this perspective, we explore the role of proteolytic degradation of NOS, in particular the inducible and neuronal isoforms of NOS, as a mechanism of regulation of the enzyme. The ubiquitin-proteasome and calpain pathways are the major proteolytic systems identified to date that are responsible for this regulated degradation. The degradation of NOS is affected by diverse agents, including glucocorticoids, caveolin, neurotoxic compounds, and certain NOS inhibitors. Some irreversible inactivators of NOS enhance the proteolytic degradation of the enzyme, and this property may be of great importance in understanding the biological effects of these inhibitors, some of which are being developed for clinical use. Analogies with the regulated degradation of liver microsomal cytochromes P450, which are related to NOS, provide a framework for understanding these processes. Finally, a new perspective on the regulation of NOS by hsp90-based chaperones is presented that involves facilitated heme insertion into the enzyme.


Subject(s)
Endopeptidases/metabolism , Enzyme Inhibitors/pharmacology , HSP90 Heat-Shock Proteins/physiology , Nitric Oxide Synthase/metabolism , Animals , Humans , Hydrolysis , Molecular Chaperones/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...