Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
2.
J Biol Chem ; 276(52): 49133-41, 2001 Dec 28.
Article in English | MEDLINE | ID: mdl-11590164

ABSTRACT

Pathological nitric oxide (NO) generation in sepsis, inflammation, and stroke may be therapeutically controlled by inhibiting NO synthases (NOS). Here we targeted the (6R)-5,6,7,8-tetrahydro-l-biopterin (H(4)Bip)-binding site of NOS, which, upon cofactor binding, maximally increases enzyme activity and NO production from substrate l-arginine. The first generation of H(4)Bip-based NOS inhibitors employed a 4-amino pharmacophore of H(4)Bip analogous to antifolates such as methotrexate. We developed a novel series of 4-oxo-pteridine derivatives that were screened for inhibition against neuronal NOS (NOS-I) and a structure-activity relationship was determined. To understand the structural basis for pterin antagonism, selected derivatives were docked into the NOS pterin binding cavity. Using a reduced 4-oxo-pteridine scaffold, derivatives with certain modifications such as electron-rich aromatic phenyl or benzoyl groups at the 5- and 6-positions, were discovered to markedly inhibit NOS-I, possibly due to hydrophobic and electrostatic interactions with Phe(462) and Ser(104), respectively, within the pterin binding pocket. One of the most effective 4-oxo compounds and, for comparisons an active 4-amino derivative, were then co-crystallized with the endothelial NOS (NOS-III) oxygenase domain and this structure solved to confirm the hypothetical binding modes. Collectively, these findings suggest (i) that, unlike the antifolate principle, the 4-amino substituent is not essential for developing pterin-based NOS inhibitors and (ii), provide a steric and electrostatic basis for their rational design.


Subject(s)
Biopterins/analogs & derivatives , Biopterins/chemistry , Biopterins/metabolism , Nitric Oxide Synthase/chemistry , Nitric Oxide Synthase/metabolism , Animals , Antioxidants/chemistry , Antioxidants/metabolism , Binding Sites , Cerebellum/enzymology , Humans , Isoenzymes/chemistry , Isoenzymes/metabolism , Models, Molecular , Molecular Structure , Nitric Oxide Synthase/antagonists & inhibitors , Protein Binding , Protein Structure, Tertiary , Structure-Activity Relationship , Swine
3.
Br J Pharmacol ; 132(2): 489-99, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11159699

ABSTRACT

1. The ability of several phorbol ester protein kinase C (PKC) activators (phorbol 12, 13-dibutyrate, PDB; phorbol 12, 13-diacetate, PDA; and 12-deoxyphorbol 13-acetate, dPA) to down-regulate PKC was studied by assessing their effects on electrical stimulation-induced (S-I) noradrenaline release from rat brain cortical slices and phosphorylation of the PKC neural substrate B-50 in rat cortical synaptosomal membranes. 2. In cortical slices which were incubated for 20 h with vehicle, acute application of PDB, PDA and dPA (0.1 - 3.0 microM) enhanced the S-I noradrenaline release in a concentration-dependent manner to between 200 - 250% of control in each case. In slices incubated with PDB (1 microM for 20 h), subsequent acute application of PDB (0.1 - 3.0 microM) failed to enhance S-I release, indicating PKC down-regulation. However, in tissues incubated with PDA or dPA (3 microM) for 20 h, there was no reduction in the facilitatory effect of their respective phorbol esters or PDB (0.1 - 3.0 microM) when acutely applied, indicating that PKC was not down-regulated. This was confirmed using Western blot analysis which showed that PDB (1 microM for 20 h) but not PDA (3 microM for 20 h) caused a significant reduction in PKCalpha. 3. Incubation with PDB for 20 h, followed by acute application of PDB (3 microM) failed to increase phosphorylation of B-50 in synaptosomal membranes, indicating down-regulation. In contrast, tissues incubated with PDA or dPA for 20 h, acute application of their respective phorbol ester (10 microM) or PDB (3 microM) induced a significant increase in B-50 phosphorylation. 4. Acutely all three phorbol esters elevate noradrenaline release to about the same extent, yet PDA and dPA have lower affinities for PKC compared to PDB, suggesting unique neural effects for these agents. This inability to cause functional down-regulation of PKC extends their unusual neural properties. Their neural potency and lack of down-regulation may be related to their decreased lipophilicity compared to other phorbol esters. 5. We suggest that PKC down-regulation appears to be related to binding affinity, where agents with high affinity, irreversibly insert PKC into artificial membrane lipid and generate Ca(2+)-independent kinase activity which degrades and deplete PKC. We suggest that this mechanism may also underlie the ability of PDB to down-regulate PKC in nerve terminals, in contrast to PDA and dPA.


Subject(s)
Down-Regulation/drug effects , Neurons/enzymology , Norepinephrine/physiology , Phorbol Esters/pharmacology , Protein Kinase C/biosynthesis , Sympathetic Nervous System/enzymology , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , In Vitro Techniques , Male , Neurons/drug effects , Norepinephrine/metabolism , Phorbol Esters/metabolism , Phosphorylation , Rats , Rats, Sprague-Dawley , Sympathetic Nervous System/cytology , Sympathetic Nervous System/drug effects , Synaptosomes/drug effects , Synaptosomes/metabolism
4.
Minerva Ginecol ; 52(4): 111-21, 2000 Apr.
Article in Italian | MEDLINE | ID: mdl-10900941

ABSTRACT

In the vagina there are several microorganisms. Their survival is fundamental to have a physiological environment in the vagina (ecosystem). The same vaginal environment playg a fundamental role to guarantee tho survival of resident microorganisms. Several factors, like changes of temperature and oxygenation, can interfere on vaginal ecosystem, but the resident vaginal microorganisms are the main factors to stabilize the vaginal ecosystem. The Doderlein's lactobacillus is the predominant vaginal microorganism. It is capable of fermenting the glycogen deriving from the decline of the eutrophic vaginal mmucosa, to lactic acid with release of hydrogen ions. The final result of that metabolism is an acid pH with values between 4-4.5. Vaginal pH undergoes physiologically changes from birth to menopause, according to changes of ovarian steroids occurring during woman's life. Adequate levels of estrogens play a fundamental role in the trophism of vaginal mucosa. In fact, estrogens increase the cellular content of glycogen. Exogenous acbvities on vaginal pH can be exerted by several factors, such as sexual activily, oral contraceptives, systemic diseases, vaginal infections (candidosis, thrichomonias, vaginosis), systemic or local therapies. They incrcase vaginal pH by acting through different mechanisms. The increase of vaginal pH is detrimental for the survival of Doderlein's lactobacillus, but not for the pathogenetic microorganisms whose replication, on the contrary, is favored by the absence of contraction exerted by Doderlein's bacillus. It has been showed that local acidifying substances (lactic acid, lactobacillus and substances recently synthesized like alpha-aminovalerianic acid, policarbophil and carbopol 934) are useful in restoring the biological and chemical characteristics of the vaginal ecosystem.


Subject(s)
Vagina/physiology , Animals , Female , Humans , Hydrogen-Ion Concentration
5.
Life Sci ; 67(1): 31-8, 2000 May 26.
Article in English | MEDLINE | ID: mdl-10896026

ABSTRACT

Activation of protein kinase C (PKC) results in enhanced action-potential evoked release of a variety of transmitters. However, previous studies have suggested that acetylcholine release is poorly modulated by PKC compared to other transmitter types. We investigated the effect of stimulation conditions on PKC modulation of electrical stimulation-induced acetylcholine release in mouse cortex, which were incubated with [3H]choline. The PKC activator phorbol dibutyrate (PDB) enhanced acetylcholine release at low stimulation frequencies (0.1 and 0.5 Hz) and not at 3 or 10 Hz. At 3 Hz stimulation, when release was inhibited by neostigmine, PDB enhanced acetylcholine release, suggesting that at low levels of acetylcholine release, exogenous activation of PKC can elevate acetylcholine release. However, at higher frequencies, PKC may already be endogenously activated since the PKC inhibitor polymyxin B (PXB) inhibited acetylcholine release. The other PKC inhibitors, Ro 318220, Gö 6976, bisindolylmaleimide and calphostin C appeared to have no effect at 3 Hz. It may be that these inhibitors do not effectively block PKC in this functional system. Indeed, polymyxin B completely blocked the facilitatory effect of PDB but Ro 318220 was without effect.


Subject(s)
Acetylcholine/metabolism , Cerebral Cortex/metabolism , Protein Kinase C/physiology , Animals , Cerebral Cortex/drug effects , Electric Stimulation , Enzyme Inhibitors/pharmacology , Male , Mice , Neostigmine/pharmacology , Phorbol 12,13-Dibutyrate/pharmacology , Polymyxin B/pharmacology , Protein Kinase C/antagonists & inhibitors
6.
Biochem J ; 346 Pt 3: 767-76, 2000 Mar 15.
Article in English | MEDLINE | ID: mdl-10698705

ABSTRACT

The underlying mechanisms regulating the activity of the family of homodimeric nitric oxide synthases (NOSs) and, in particular, the requirement for (6R)-5,6,7,8-tetrahydro-L-biopterin (H(4)Bip) are not fully understood. Here we have investigated possible allosteric and stabilizing effects of H(4)Bip on neuronal NOS (NOS-I) during the conversion of substrate, L-arginine, into L-citrulline and nitric oxide. Indeed, in kinetic studies dual allosteric interactions between L-arginine and H(4)Bip activated recombinant human NOS-I to increase L-arginine turnover. Consistent with this was the observation that H(4)Bip, but not the pterin-based NOS inhibitor 2-amino-4,6-dioxo-3,4,5,6,8,8a,9,10-octahydrooxazolo[1, 2-f]-pteridine (PHS-32), caused an L-arginine-dependent increase in the haem Soret band, indicating an increase in substrate binding to recombinant human NOS-I. Conversely, L-arginine was observed to increase in a concentration-dependent manner H(4)Bip binding to pig brain NOS-I. Secondly, we investigated the stabilization of NOS quaternary structure by H(4)Bip in relation to uncoupled catalysis. Under catalytic assay conditions and in the absence of H(4)Bip, dimeric recombinant human NOS-I dissociated into inactive monomers. Monomerization was related to the uncoupling of reductive oxygen activation, because it was inhibited by both superoxide dismutase and the inhibitor N(omega)-nitro-L-arginine. Importantly, H(4)Bip was found to react chemically with superoxide (O(2)(-.)) and enzyme-bound H(4)Bip was consumed under O(2)(-.)-generating conditions in the absence of substrate. These results suggest that H(4)Bip allosterically activates NOS-I and stabilizes quaternary structure by a novel mechanism involving the direct interception of auto-damaging O(2)(-.).


Subject(s)
Biopterins/analogs & derivatives , Nitric Oxide Synthase/metabolism , Superoxides/antagonists & inhibitors , Allosteric Regulation , Arginine/pharmacology , Binding Sites , Biopterins/metabolism , Biopterins/pharmacology , Catalysis , Cell Line , Enzyme Activation , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Humans , Indicators and Reagents , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/chemistry , Nitric Oxide Synthase Type I , Protein Structure, Quaternary , Superoxides/metabolism
7.
J Biol Chem ; 275(7): 5179-87, 2000 Feb 18.
Article in English | MEDLINE | ID: mdl-10671564

ABSTRACT

Endothelial nitric-oxide synthase (NOS-III) is defined as being strictly dependent on Ca(2+)/calmodulin (CaM) for activity, although NO release from endothelial cells has been reported to also occur at intracellular free Ca(2+) levels that are substimulatory for the purified enzyme. We demonstrate here that NOS-III, but neither NOS-I nor -II, is rapidly and strongly activated and phosphorylated on both Ser and Thr in the presence of cGMP-dependent protein kinase II (cGK II) and the catalytic subunit of cAMP-dependent protein kinase (cAK) in vitro. Phosphopeptide analysis by mass spectrometry identified Ser(1177), as well as Ser(633) which is situated in a recently defined CaM autoinhibitory domain within the flavin-binding region of human NOS-III. Phosphoamino acid analysis identified a putative phosphorylation site at Thr(495) in the CaM-binding domain. Importantly, both cAK and cGK phosphorylation of NOS-III in vitro caused a highly reproducible partial (10-20%) NOS-III activation which was independent of Ca(2+)/CaM, and as much as a 4-fold increase in V(max) in the presence of Ca(2+)/CaM. cAK stimulation in intact endothelial cells also increased both Ca(2+/)CaM-independent and -dependent activation of NOS-III. These data collectively provide new evidence for cAK and cGK stimulation of both Ca(2+)/CaM-independent and -dependent NOS-III activity, and suggest possible cross-talk between the NO and prostaglandin I(2) pathways and a positive feedback mechanism for NO/cGMP signaling.


Subject(s)
Calcium/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Nitric Oxide Synthase/metabolism , Amino Acid Sequence , Animals , Calmodulin/metabolism , Enzyme Activation , Flavins/metabolism , Humans , Molecular Sequence Data , Nitric Oxide Synthase/chemistry , Nitric Oxide Synthase Type III , Phosphorylation , Protein Conformation , Sequence Homology, Amino Acid , Substrate Specificity
8.
Eur J Pharmacol ; 381(1): 77-84, 1999 Sep 17.
Article in English | MEDLINE | ID: mdl-10528137

ABSTRACT

The present study used structurally distinct phorbol esters to investigate the relationship between their pharmacokinetics of binding to protein kinase C (PKC) in rat brain cortex synaptosomes, their affinity for PKC in synaptosomes and ability to enhance noradrenaline release from rat brain cortex. Affinity binding studies using [3deoxyphorbol 13-tetradecanoate (dPT)=PDB&z. Gt;12-deoxyphorbol 13-acetate (dPA)=phorbol 12,13-diacetate (PDA). In intact synaptosomes PDB, dPA and PDA rapidly displaced bound [3H]PDB whereas PMA and dPT were comparatively slow. However, the displacement rates for all the phorbol esters were equally rapid in synaptosomal membranes or synaptosomes permeabilised with Staphylococcus alpha-toxin. These results suggest that the lipophilic phorbol esters (dPT and PMA) are slower to displace [3H]PDB binding because they are hindered by the plasma membrane. In brain cortex slices it was found that the rate of displacement of [3H]PDB binding was closely correlated with the degree of elevation of transmitter noradrenaline release. Thus kinetic characteristics may determine biological responses and this may be particularly evident in events which occur rapidly or where there is fast counter-regulation.


Subject(s)
Phorbol Esters/metabolism , Synaptosomes/metabolism , Animals , Binding Sites , Binding, Competitive/drug effects , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Kinetics , Male , Norepinephrine/metabolism , Phorbol 12,13-Dibutyrate/metabolism , Phorbol 12,13-Dibutyrate/pharmacokinetics , Phorbol 12,13-Dibutyrate/pharmacology , Phorbol Esters/pharmacokinetics , Phorbol Esters/pharmacology , Protein Binding , Protein Kinase C/metabolism , Rats , Rats, Sprague-Dawley , Synaptic Membranes/metabolism , Synaptosomes/enzymology , Tetradecanoylphorbol Acetate/metabolism , Tetradecanoylphorbol Acetate/pharmacokinetics , Tetradecanoylphorbol Acetate/pharmacology , Time Factors , Tritium
9.
J Med Chem ; 42(20): 4108-21, 1999 Oct 07.
Article in English | MEDLINE | ID: mdl-10514281

ABSTRACT

The family of nitric oxide synthases (NOS) catalyzes the conversion of L-arginine to L-citrulline and nitric oxide (NO), an important cellular messenger molecule which has been implicated in the pathophysiology of septic shock and inflammatory and neurodegenerative disease states. NOS can be maximally activated by the ubiquitous cofactor, (6R)-5,6,7,8-tetrahydrobiopterin (H(4)Bip), and antagonists of H(4)Bip may be of therapeutic importance to inhibit pathologically high NO formation. The 4-amino substituted analogue of H(4)Bip was reported to be a potent NOS inhibitor. Therefore, we developed a series of novel 4-amino pteridine derivatives, anti-pterins, to pharmacologically target the neuronal isoform of nitric oxide synthase (NOS-I). To functionally characterize the pterin/anti-pterin interaction and establish a structure-activity relationship (SAR), we systematically altered the substituents in the 2-, 4-, 5-, 6-, and 7-position of the pteridine nucleus. Varying the substitution pattern in the 2-, 5-, and 7-position resulted in no significant inhibitory effect on enzyme activity. In contrast, bulky substituents in the 6-position, such as phenyl, markedly increased the inhibitory potency of the reduced 4-amino-5,6,7,8-tetrahydropteridines, possibly as a consequence of hydrophobic interactions within NOS-I. However, this was not the case for the aromatic 4-amino pteridines. Interestingly, chemical modification of the 4-amino substituent by dialkyl/diaralkylation together with 6-arylation of the aromatic 2,4-diamino pteridine resulted in potent and efficacious inhibitors of NOS-I, suggesting possible hydrophilic and hydrophobic interactions within NOS-I. This SAR agrees with (a) the recently published crystal structure of the oxygenase domain of the inducible NOS isoform (NOS-II) and (b) the comparative molecular field analysis of selected NOS-I inhibitors, which resulted in a 3D-QSAR model of the pterin binding site interactions. Further optimization should be possible when the full length structure of NOS-I becomes available.


Subject(s)
Biopterins/analogs & derivatives , Enzyme Inhibitors/chemical synthesis , Neurons/enzymology , Nitric Oxide Synthase/antagonists & inhibitors , Pteridines/chemical synthesis , Animals , Biopterins/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Isoenzymes/antagonists & inhibitors , Pteridines/chemistry , Structure-Activity Relationship , Swine
10.
J Biol Chem ; 274(35): 24921-9, 1999 Aug 27.
Article in English | MEDLINE | ID: mdl-10455167

ABSTRACT

The biosynthesis of nitric oxide (NO) is catalyzed by homodimeric NO synthases (NOS). For unknown reasons, all NOS co-purify with substoichiometric amounts of (6R)-5,6,7,8-tetrahydrobiopterin (H(4)Bip) and require additional H(4)Bip for maximal activity. We examined the effects of H(4)Bip and pterin-derived inhibitors (anti-pterins) on purified neuronal NOS-I quaternary structure and H(4)Bip content. During L-arginine turnover, NOS-I dimers time dependently dissociated into inactive monomers, paralleled by a loss of enzyme-associated pterin. Dimer dissociation was inhibited when saturating levels of H(4)Bip were added during catalysis. Similar results were obtained with pterin-free NOS-I expressed in Escherichia coli. This stabilizing effect of H(4)Bip was mimicked by the anti-pterin 2-amino-4,6-dioxo-3,4,5,6,8,8a,9, 10-octahydro-oxazolo[1,2f]-pteridine (PHS-32), which also displaced NOS-associated H(4)Bip in a competitive manner. Surprisingly, H(4)Bip not only dissociated from NOS during catalysis, but was only partially recovered in the solute (50.0 +/- 16.5% of control at 20 min). NOS-associated H(4)Bip appeared to react with a NOS catalysis product to a derivative distinct from dihydrobiopterin or biopterin. Under identical conditions, reagent H(4)Bip was chemically stable and fully recovered (95.5 +/- 3.4% of control). A similar loss of both reagent and enzyme-bound H(4)Bip and dimer content was observed by NO generated from spermine NONOate. In conclusion, we propose a role for H(4)Bip as a dimer-stabilizing factor of neuronal NOS during catalysis, possibly by interfering with enzyme destabilizing products.


Subject(s)
Biopterins/analogs & derivatives , Nitric Oxide Synthase/chemistry , Arginine/metabolism , Biopterins/metabolism , Biopterins/pharmacology , Dimerization , Enzyme Stability/drug effects , Escherichia coli , Humans , Kinetics , Nitric Oxide/metabolism , Nitric Oxide Synthase Type I , Protein Conformation , Pteridines/pharmacology , Recombinant Proteins/chemistry , Spectrophotometry , Spermine/analogs & derivatives
11.
Br J Pharmacol ; 127(5): 1177-89, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10455264

ABSTRACT

The effects of various phorbol-based protein kinase C (PKC) activators on the electrical stimulation-induced (S-I) release of serotonin and acetylcholine was studied in rat brain cortical slices pre-incubated with [3H]-serotonin or [3H]-choline to investigate possible structure-activity relationships. 4beta-phorbol 12,13-dibutyrate (4betaPDB, 0.1-3.0 microM), enhanced S-I release of serotonin in a concentration-dependent manner whereas the structurally related inactive isomer 4alpha-phorbol 12, 13-dibutyrate (4alphaPDB) and phorbol 13-acetate (PA) were without effect. Another group of phorbol esters containing a common 13-ester substituent (phorbol 12,13-diacetate, PDA; phorbol 12-myristate 13-acetate, PMA; phorbol 12-methylaminobenzoate 13-acetate, PMBA) also enhanced S-I serotonin release with PMA being least potent. The deoxyphorbol monoesters, 12-deoxyphorbol 13-acetate (dPA), 12-deoxyphorbol 13-angelate (dPAng), 12-deoxyphorbol 13-phenylacetate (dPPhen) and 12-deoxyphorbol 13-isobutyrate (dPiB) enhanced S-I serotonin release but 12-deoxyphorbol 13-tetradecanoate (dPT) was without effect. The 20-acetate derivatives of dPPhen and dPAng were less effective in enhancing S-I serotonin release compared to the parent compounds. With acetylcholine release all phorbol esters tested had a far lesser effect when compared to their facilitatory action on serotonin release with only 4betaPDB, PDA, dPA, dPAng and dPiB having significant effects. The effects of the phorbol esters on serotonin release were not correlated with their reported in vitro affinity and isozyme selectivity for PKC. A comparison across three transmitter systems (noradrenaline, dopamine, serotonin) suggests basic similarities in the structural requirements of phorbol esters to enhance transmitter release with short chain substituted mono- and diesters of phorbol being more potent facilitators of release than the long chain esters. Some compounds notably PDA, PMBA, dPPhen, dPPhenA had different potencies across noradrenaline, dopamine and serotonin.


Subject(s)
Acetylcholine/metabolism , Cerebral Cortex/drug effects , Phorbol Esters/pharmacology , Protein Kinase C/physiology , Serotonin/metabolism , Animals , Cerebral Cortex/metabolism , Choline/metabolism , Dose-Response Relationship, Drug , Phorbol 12,13-Dibutyrate/pharmacology , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
12.
Mol Pharmacol ; 56(2): 243-53, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10419542

ABSTRACT

Soluble guanylyl cyclase (sGC) is an important effector for nitric oxide (NO). It acts by increasing intracellular cyclic GMP (cGMP) levels to mediate numerous biological functions. Recently, 1H-[1,2, 4]oxadiazolo[4,3,-a]quinoxalin-1-one (ODQ) was identified as a novel and selective inhibitor of this enzyme. Therefore, ODQ may represent an important pharmacological tool for differentiating cGMP-mediated from cGMP-independent effects of NO. In the present study, we examined the inhibitory action of ODQ both functionally and biochemically. In phenylephrine-preconstricted, endothelium-intact, isolated aortic rings from the rat, ODQ, in a concentration-dependent manner, increased contractile tone and inhibited relaxations to authentic NO with maximal effects at 3 microM. Pretreatment of vascular rings with ODQ induced a parallel, 2-log-order shift to the right of the concentration-response curves (CRCs) to histamine, ATP, NO, the NO-donors S-nitrosoglutathione, S-nitroso-N-acetyl-D,L-penicillamine, and spermine NONOate [N-[4-[1-(3-amino propyl)-2-hydroxy-2-nitroso hydrazino]butyl]-1, 3-propane diamine], and the direct sGC-stimulant [3-(5'-hydroxymethyl-2'furyl)-1-benzyl indazole] YC-1 but did not affect relaxations induced by papaverine and atriopeptin II. Moreover, the rightward shift of the CRCs to Angeli's salt, peroxynitrite, and linsidomine was similar to that of NO. These results suggested that ODQ is specific for sGC. Furthermore, they indicate that NO can cause vasorelaxation independent of cGMP. Three interesting exceptions were observed to the otherwise rather uniform inhibitory effect of ODQ: the responses to acetylcholine, glycerol trinitrate, and sodium nitroprusside. The latter two agents are known to require metabolic activation, possibly by cytochrome P-450-type proteins. The 3- to 5-log-order rightward shift of their CRCs suggests that, in addition to sGC, ODQ may interfere with heme proteins involved in the bioactivation of these NO donors and the mechanism of vasorelaxation mediated by acetylcholine. In support of this notion, ODQ inhibited hepatic microsomal NO production from both glycerol trinitrate and sodium nitroprusside as well as NO synthase activity in aortic homogenates. The latter effect seemed to require biotransformation of ODQ. Collectively, these data reveal that ODQ interferes with various heme protein-dependent processes in vascular and hepatic tissue and lacks specificity for sGC.


Subject(s)
Cytochrome P-450 Enzyme Inhibitors , Enzyme Inhibitors/pharmacology , Guanylate Cyclase/antagonists & inhibitors , Nitric Oxide Donors/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Oxadiazoles/pharmacology , Quinoxalines/pharmacology , Animals , Aorta/metabolism , Biotransformation , Cytochrome P-450 Enzyme System/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Hemeproteins/antagonists & inhibitors , Indazoles/pharmacology , Male , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Nitric Oxide/physiology , Nitric Oxide Synthase/metabolism , Nitroprusside/pharmacology , Rats , Rats, Wistar , Vasodilation/drug effects
13.
Biochem J ; 340 ( Pt 3): 745-52, 1999 Jun 15.
Article in English | MEDLINE | ID: mdl-10359660

ABSTRACT

Nitric oxide (NO) synthases (NOSs), which catalyse the oxidation of L-arginine to L-citrulline and an oxide of nitrogen, possibly NO or nitroxyl (NO-), are subject to autoinhibition by a mechanism that has yet to be fully elucidated. In the present study we investigated the actions of NO and other NOS-derived products as possible autoregulators of enzyme activity. With the use of purified NOS-I, L-arginine turnover was found to operate initially at Vmax (0-15 min, phase I) although, despite the presence of excess substrate and cofactors, prolonged catalysis (15-90 min, phase II) was associated with a rapid decline in L-arginine turnover. Taken together, these observations suggested that one or more NOS products inactivate NOS. Indeed, exogenously applied reactive nitrogen oxide species (RNSs) decreased Vmax during phase I, although with different potencies (NO->NO> ONOO-) and efficacies (NO>NO-=ONOO-). The NO scavengers oxyhaemoglobin (HbO2; 100 microM) and 1H-imidazol-1 - yloxy - 2 - (4-carboxyphenyl) - 4,5 - dihydro - 4,4,5,5 - tetramethyl - 3 -oxide (CPTIO; 10 microM) and the ONOO- scavenger GSH (7 mM) had no effect on NOS activity during phase I, except for an endogenous autoinhibitory influence of NO and ONOO-. However, superoxide dismutase (SOD; 300 units/ml), which is thought either to increase the half-life of NO or to convert NO- to NO, lowered Vmax in an NO-dependent manner because this effect was selectively antagonized by HbO2 (100 microM). This latter observation demonstrated the requirement of SOD to reveal endogenous NO-mediated autoinhibition. Importantly, during phase II of catalysis, NOS became uncoupled and began to form H2O2 because catalase, which metabolizes H2O2, increased enzyme activity. Consistent with this, exogenous H2O2 also inhibited NOS activity during phase I. Thus during catalysis NOS is subject to complex autoinhibition by both enzyme-derived RNS and H2O2, differentially affecting enzyme activity.


Subject(s)
Nitric Oxide Synthase/antagonists & inhibitors , Nitrogen/metabolism , Reactive Oxygen Species/metabolism , Animals , Arginine/metabolism , Cerebellum/enzymology , Citrulline/metabolism , Feedback , Free Radical Scavengers/metabolism , Glutathione/metabolism , Hydrogen Peroxide/metabolism , Inhibitory Concentration 50 , Kinetics , Methemoglobin/metabolism , Methionine/metabolism , Nitrates/metabolism , Nitric Oxide/metabolism , Nitric Oxide Donors/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type I , Superoxide Dismutase/metabolism , Swine
14.
J Biol Chem ; 273(50): 33142-9, 1998 Dec 11.
Article in English | MEDLINE | ID: mdl-9837881

ABSTRACT

Nitric oxide synthases (NOS) are homodimeric enzymes that NADPH-dependently convert L-arginine to nitric oxide and L-citrulline. Interestingly, all NOS also require (6R)-5,6,7, 8-tetrahydro-L-biopterin (H4Bip) for maximal activity although the mechanism is not fully understood. Basal NOS activity, i.e. that in the absence of exogenous H4Bip, has been attributed to enzyme-associated H4Bip. To elucidate further H4Bip function in purified NOS, we developed two types of pterin-based NOS inhibitors, termed anti-pterins. In contrast to type II anti-pterins, type I anti-pterins specifically displaced enzyme-associated H4Bip and inhibited H4Bip-stimulated NOS activity in a fully competitive manner but, surprisingly, had no effect on basal NOS activity. Moreover, for a number of different NOS preparations basal activity (percent of Vmax) was frequently higher than the percentage of pterin saturation and was not affected by preincubation of enzyme with H4Bip. Thus, basal NOS activity appeared to be independent of enzyme-associated H4Bip. The lack of intrinsic 4a-pterincarbinolamine dehydratase activity argued against classical H4Bip redox cycling in NOS. Rather, H4Bip was required for both maximal activity and stability of NOS by binding to the oxygenase/dimerization domain and preventing monomerization and inactivation during L-arginine turnover. Since anti-pterins were also effective in intact cells, they may become useful in modulating states of pathologically high nitric oxide formation.


Subject(s)
Biopterins/analogs & derivatives , Nitric Oxide Synthase/metabolism , Animals , Biopterins/antagonists & inhibitors , Biopterins/metabolism , Catalysis , Cerebellum/enzymology , Humans , Kinetics , Recombinant Proteins/metabolism , Swine
15.
Clin Exp Pharmacol Physiol ; 24(8): 619-23, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9269538

ABSTRACT

1. Protein kinase C (PKC) is an important second messenger-activated enzyme. In noradrenergic nerves it appears to be tonically activated by diacylglycerol (DAG) to facilitate transmitter release and the steps in this involve activation of phospholipase C, generation of DAG and activation of PKC. It is suggested that the subsequent facilitation of transmitter release is due to the phosphorylation of proteins involved in the release process distal to Ca2+ entry, presumably those involved in vesicle dynamics. 2. There are differences between central noradrenergic neurons and sympathetic nerves. In central neurons PKC appears to be tonically active and its inhibition results in a decrease in noradrenaline release under most, if not all, conditions. 3. In sympathetic nerves PKC inhibitors only decrease transmitter release during high-frequency stimulation and not during low-frequency stimulation. At high frequency there is a gradual increase in the effect of PKC inhibitors on transmitter release during the first 15 s of a stimulation train. It is suggested that this is due to a progressive rise in intracellular Ca2+ and a consequent activation of PKC. 4. Activation of PKC by phorbol esters produces a large enhancement in action potential-evoked noradrenaline release in both the central nervous system and in peripheral tissues. The structural requirements of the phorbol esters for maximal effect suggest that the phorbol esters must access the interior of the nerve terminal to activate PKC and the neural membrane acts as a barrier for highly lipophilic phorbol esters, thereby reducing their activity. Activation of PKC represents one of the most powerful ways to enhance transmitter release and may have therapeutic potential.


Subject(s)
Neurotransmitter Agents/metabolism , Protein Kinase C/physiology , Animals , Norepinephrine/metabolism , Signal Transduction/physiology
16.
Br J Pharmacol ; 121(6): 1196-202, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9249257

ABSTRACT

1. It has been proposed that protein kinase C (PKC) in sympathetic nerves is activated during action-potential evoked release of noradrenaline and helps maintain transmitter output. We studied this phenomenon further in rat atria radiolabelled with [3H]-noradrenaline. 2. Noradrenaline release was elevated by continuous electrical stimulation of the atria for 10 min at either 5 or 10 Hz. Two inhibitors of PKC, polymyxin B (21 microM) and Ro 318220 (3 microM), markedly inhibited the release of noradrenaline but only at the higher stimulation frequency. 3. Further experiments were conducted with 10 Hz stimulation but for shorter train durations. In this case polymyxin B inhibited noradrenaline release during a 10 or 15 s train of impulses but not during a 5 s train. This suggests that PKC effects are induced during the stimulation train by some process. 4. The diacylglycerol kinase inhibitor R59949 (10 microM), which prevents the breakdown of diacylglycerol, enhanced noradrenaline release elicited by stimulation at 10 Hz for 10 or 15 s. This effect was not seen if polymyxin B was present and suggests that diacylglycerol is the endogenous activator of PKC. 5. The source of the diacylglycerol may be through phospholipase C pathways, since the phospholipase C inhibitor U73122 (3 microM) inhibited noradrenaline release at 10 Hz for 10 s and the effect was not seen if polymyxin B was also present. 6. It is unlikely that phospholipase D is the source of diacylglycerol. Although the phospholipase D inhibitor wortmannin (1 microM) inhibited noradrenaline release, this effect was still observed in the presence of polymyxin B. Furthermore ethanol, which inhibits diacylglycerol formation by phospholipase D, had no effect on noradrenaline release. 7. We therefore suggest that during a train of high frequency pulses phospholipase C is activated and this results in the production of diacylglycerol which in turn activates PKC. This enables the neurones to maintain transmitter release at a high level.


Subject(s)
Heart Atria/metabolism , Norepinephrine/metabolism , Protein Kinase C/metabolism , Signal Transduction , Type C Phospholipases/metabolism , Androstadienes/pharmacology , Animals , Atrial Function , Diacylglycerol Kinase , Electric Stimulation , Enzyme Activation , Enzyme Inhibitors/pharmacology , Estrenes/pharmacology , Heart Atria/enzymology , Male , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Piperidines/pharmacology , Pyrrolidinones/pharmacology , Quinazolines/pharmacology , Quinazolinones , Rats , Rats, Sprague-Dawley , Type C Phospholipases/antagonists & inhibitors , Wortmannin
17.
Br J Pharmacol ; 119(8): 1605-13, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8982508

ABSTRACT

1. The biosynthesis of noradrenaline following sympathetic nerve activation was investigated in rat atria. In particular the time course of noradrenaline synthesis changes, the relationship of changes in synthesis to transmitter release and the possible roles of second messengers and protein kinases were examined. 2. Rat atria incubated with the precursor [3H]-tyrosine synthesized [3H]-noradrenaline. Synthesis was enhanced following pulsatile electrical field stimulation (3 Hz for 5 min) with the bulk of the increase occurring in the first 45 min after the commencement of electrical stimulation. In separate experiments rat atria were pre-incubated with [3H]-noradrenaline and the radioactive outflow in response to electrical field stimulation (3 Hz for 5 min) was taken as an index of noradrenaline release. 3. Stimulation-induced (S-I) noradrenaline synthesis was significantly correlated to S-I noradrenaline release for a variety of procedures which modulate noradrenaline release by mechanisms altering Ca2+ entry into the neurone (r2 = 0.99): those which decreased release: tetrodotoxin (0.3 microM), Ca(2+)-free medium, lowering the frequency of nerve activation to 1 Hz, and those which increased release, tetraethylammonium (0.3 mM), phentolamine (1 microM) and the combination of phentolamine (1 microM) and adenosine (10 microM). On the strength of this relationship we suggest that Ca2+ entry is a determining factor in S-I synthesis changes rather than the amount of noradrenaline released. Indeed the reduction in noradrenaline release with the calmodulin-dependent protein (CAM) kinase II inhibitor KN-62 (10 microM) which acts subsequent to Ca2+ entry, did not affect S-I synthesis. 4. The cell permeable cyclic AMP analogue, 8-bromoadenosine 3',5'-monophosphate (BrcAMP, 90 and 270 microM), dose-dependently increased basal [3H]-noradrenaline synthesis in unstimulated rat atria. This effect was antagonized by the selective protein kinase A (PKA) antagonist, Rp-8-chloroadenosine 3',5'-cyclic monophosphorothioate (RClcAMPS, 300 microM), suggesting that PKA activation enhances basal noradrenaline biosynthesis in sympathetic nerve terminals. 5. The protein kinase inhibitors, KN-62 (CAM kinase II, 10 microM), RClcAMPS (PKA, 300 microM), polymyxin B (protein kinase C (PKC), 21 microM) and staurosporine (PKC, PKA and CAM kinase II, (0.1 microM) did not affect S-I synthesis, although KN-62, polymyxin B and staurosporine decreased S-I release. We conclude that S-I synthesis is triggered by Ca2+ entering the neurone but that the signalling pathway does not involve classical protein kinases and appears distinct from the steps involved in transmitter release.


Subject(s)
Calcium/metabolism , Heart/drug effects , Heart/innervation , Myocardium/metabolism , Norepinephrine/biosynthesis , Sympathetic Nervous System/physiology , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Electric Stimulation , Enzyme Inhibitors/pharmacology , Male , Myocardium/enzymology , Norepinephrine/metabolism , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Rats , Rats, Sprague-Dawley , Second Messenger Systems/drug effects , Second Messenger Systems/physiology , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/metabolism , Thionucleotides/pharmacology
18.
Br J Pharmacol ; 119(1): 115-25, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8872364

ABSTRACT

1. The effects of various protein kinase C (PKC) activators on the stimulation-induced (S-I) release of noradrenaline and dopamine was studied in rat cortical slices pre-incubated with [3H]-noradrenaline or [3H]-dopamine. The aim was to investigate a possible structure-activity relationship for these agents on transmitter release. 2. 4 beta-Phorbol 12,13-dibutyrate (4 beta PDB, 0.1-3.0 microM), enhanced S-I noradrenaline and dopamine release in a concentration-dependent manner whereas the structurally related inactive isomer 4 alpha-phorbol 12, 13-dibutyrate (4 alpha PDB, 0.1-3.0 microM) and phorbol 13-acetate (PA, 0.1-3.0microM) were without effect on noradrednaline release. Another group of phorbol 12, 13-diesters containing a common 13-ester substituent (phorbol 12, 13-diacetate, PDA, 0.1-3.0 microM; phorbol 12-myristate 13-acetate, PMA, 0.1-3.0 microM; phorbol 12-methylaminobenzoate 13-acetate, PMBA, 0.03-3.0 microM) also enhanced S-I noradrenaline and dopamine release in a concentration-dependent manner with PMA being the least potent. 3. The 12-deoxyphorbol 13-substituted monoesters, 12-deoxyphorbol 13-acetate (dPA, 0.1-3.0 microM), 12-deoxyphorbol 13-angelate (dPAng, 0.1-3.0 microM), 12-deoxyphorbol 13-isobutyrate (dPiB, 0.03-3.0 microM) and 12-deoxyphorbol 13-phenylacetate (dPPhen, 0.1-3.0 microM) enhanced S-I noradrenaline and dopamine release in a concentration-dependent manner. In contrast, 12-deoxyphorbol 13-tetradecanoate (dPT, 0.1-3.0 microM) was without effect. 4. The involvement of PKC in mediating the effects of the various phorbol esters was further investigated. PKC was down-regulated by 20 h exposure of the cortical slices to 4 beta-phorbol 12,13-dibutyrate (1 microM). In this case the facilitatory effect of 4 beta PDB and dPA was abolished whilst that of dPAng was significantly attenuated. This indicates that these agents were acting selectively at PKC. In support of this the PKC inhibitors, polymyxin B (21 microM) and bisindolylmaleimide I (3 microM), attenuated the facilitatory effect of 4 beta PDB and dPAng although that of dPA was not significantly altered. 5. The effects of these agents on transmitter release were not correlated with their in vitro affinity and isozyme selectivity for PKC. Short chain substituted mono- and diesters of phorbol were more potent enhancers of action-potential evoked noradrenaline and dopamine release than the long chain esters. Interestingly, these former agents are the least potent or non effective (e.g. dPA, PDA) tumour promoters. We suggest that the reason for the poor effects of lipophilic long chain phorbol esters (PMA, dPT) on transmitter release is that they are sequestered in the plasmalemma and do not access the cell cytoplasm where the PKC may be located.


Subject(s)
Cerebral Cortex/metabolism , Dopamine/metabolism , Norepinephrine/metabolism , Phorbol Esters/pharmacology , Animals , In Vitro Techniques , Phorbol 12,13-Dibutyrate/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Tetradecanoylphorbol Acetate/pharmacology , Tritium
19.
Br J Pharmacol ; 116(6): 2757-63, 1995 Nov.
Article in English | MEDLINE | ID: mdl-8591001

ABSTRACT

1. The role of protein kinase C in the modulation of noradrenaline release was investigated in mouse cortical slices which were pre-incubated with [3H]-noradrenaline. The aim was to investigate the hypothesis that protein kinase C is activated during high levels of transmitter release to maintain transmitter output. 2. The protein kinase C activators, phorbol myristate acetate (0.01-0.3 microM) and to a greater extent 4 beta-phorbol 12,13-dibutyrate (0.01-0.3 microM) significantly enhanced stimulation-induced noradrenaline release whereas 4 alpha-phorbol 12,13-dibutyrate (0.1 microM) which does not activate protein kinase C was without effect. The effect of the protein kinase C activator, phorbol myristate acetate, on noradrenaline release was attenuated by the protein kinase C inhibitor, polymyxin B (21 microM) which by itself inhibited stimulation-induced noradrenaline release. 3. Protein kinase C was down-regulated by 10 h exposure of the cortical slices to 4 beta-phorbol 12,13-dibutyrate (1 microM). In this case the facilitatory effect of 4 beta-phorbol 12,13-dibutyrate (0.1 microM) on noradrenaline release was abolished as was the inhibitory effect produced by polymyxin B. This indicates that polymyxin B was acting selectively at protein kinase C. 4. The inhibitory effect of polymyxin B on noradrenaline release, when expressed as a percentage of the appropriate frequency control, was constant at 1, 5 and 10 Hz. Furthermore, the ratio of release at 5 Hz to that at 10 Hz was not altered by protein kinase C down-regulation, indicating that there is no additional effect of protein kinase C at higher stimulation frequencies. 5. When transmitter release was elevated by blocking alpha 2-adrenoceptor auto-inhibition with idazoxan (0.1 microM) or K+ channels with tetraethylammonium (300 microM), the elevation in transmitter release was significantly attenuated by protein kinase C down-regulation, suggesting an involvement of protein kinase C. 6. We conclude that protein kinase C is involved in the modulation of noradrenaline release over a wide range of stimulation frequencies, in addition to a role when noradrenaline release is elevated by presynaptic mechanisms.


Subject(s)
Cerebral Cortex/enzymology , Cerebral Cortex/metabolism , Norepinephrine/metabolism , Protein Kinase C/physiology , Adrenergic alpha-Antagonists/pharmacology , Animals , Cerebral Cortex/drug effects , Dioxanes/pharmacology , Down-Regulation/drug effects , Electric Stimulation , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Idazoxan , Imidazoles/pharmacology , Male , Mice , Phorbol 12,13-Dibutyrate/pharmacology , Polymyxin B/pharmacology , Potassium Channels/drug effects , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Tetraethylammonium , Tetraethylammonium Compounds/pharmacology , Time Factors , Tritium
20.
Clin Exp Pharmacol Physiol ; 22(6-7): 469-71, 1995.
Article in English | MEDLINE | ID: mdl-8582106

ABSTRACT

1. This study was designed to investigate the effects of isotonic saline loading on renal tubular and neurogenic dopamine (DA) in conscious rabbits. 2. Isotonic saline loading did not affect mean arterial pressure, heart rate or renal blood flow but markedly increased urine volume, sodium excretion and DA excretion. 3. Renal DA spillover was not affected by venous emptying, while renal noradrenaline (NA) spillover tended to decrease during saline loading. The ratio of % renal DA spillover to % renal NA spillover increased to 2.3 +/- 0.6 (P < 0.05) 3 h after saline loading. 4. Isotonic saline loading increased renal tubular DA production but had little effect on neurogenic DA release.


Subject(s)
Dopamine/metabolism , Kidney Tubules/metabolism , Sodium Chloride/pharmacology , Analysis of Variance , Animals , Blood Pressure/drug effects , Diuresis/drug effects , Dopamine/urine , Heart Rate/drug effects , Isotonic Solutions , Kidney Tubules/drug effects , Male , Rabbits , Renal Circulation/drug effects , Sodium/urine
SELECTION OF CITATIONS
SEARCH DETAIL
...