Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
Add more filters










Publication year range
1.
Med Health Care Philos ; 24(4): 563-572, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34259964

ABSTRACT

Chronic pain patients often miss receiving acknowledgement for the multidimensional struggles they face with their specific conditions. People suffering from chronic pain experience a type of invisibility that is also borne by other chronically ill people and their respective medical conditions. However, chronic pain patients face both passive and active exclusion from social participation in activities like family interactions or workplace inclusion. Although such aspects are discussed in the debates lead by the bio-psycho-social model of pain, there seems to be a lack of a distinct interest in assessing more specifically the social aspects regarding chronic pain. As a result, the social aspects have yet to be taken into a more thorough theoretical consideration of chronic pain and to be practically implemented to help affected patients. By addressing chronic pain patients' struggle for recognition, this paper attempts to shed light on some of these social aspects. We base this attempt on a theoretical framework that combines patients' statements with an adaptation of Axel Honneth's social-philosophical work on recognition. Thus, this paper tries to make a suggestion on how the bio-psycho-social model of pain can live up to its name by helping to address more adequately some of the more neglected aspects in chronic pain patients' suffering than has been possible to date.


Subject(s)
Chronic Pain , Chronic Disease , Humans , Workplace
2.
Pflugers Arch ; 467(6): 1383-94, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25010738

ABSTRACT

GABAergic interneurons are the predominant source of inhibition in the brain that coordinate the level of excitation and synchronization in neuronal circuitries. However, the underlying cellular mechanisms are still not fully understood. Here we report nitric oxide (NO)/NO-GC1 signalling as an important regulatory mechanism of GABAergic and glutamatergic synaptic transmission in the hippocampal CA1 region. Deletion of the NO receptor NO-GC1 induced functional alterations, indicated by a strong reduction of spontaneous and evoked inhibitory postsynaptic currents (IPSCs), which could be compensated by application of the missing second messenger cGMP. Moreover, we found a general impairment in the strength of inhibitory and excitatory synaptic inputs onto CA1 pyramidal neurons deriving from NO-GC1KO mice. Finally, we disclosed one subpopulation of GABAergic interneurons, fast-spiking interneurons, that receive less excitatory synaptic input and consequently respond with less spike output after blockage of the NO/cGMP signalling pathway. On the basis of these and previous findings, we propose NO-GC1 as the major NO receptor which transduces the NO signal into cGMP at presynaptic terminals of different neuronal subtypes in the hippocampal CA1 region. Furthermore, we suggest NO-GC1-mediated cGMP signalling as a mechanism which regulates the strength of synaptic transmission, hence being important in gating information processing between hippocampal CA3 and CA1 region.


Subject(s)
CA1 Region, Hippocampal/metabolism , Excitatory Postsynaptic Potentials , Guanylate Cyclase/metabolism , Inhibitory Postsynaptic Potentials , Interneurons/metabolism , Nitric Oxide/metabolism , Animals , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/physiology , Cyclic GMP/metabolism , Interneurons/physiology , Male , Mice , Mice, Inbred C57BL , gamma-Aminobutyric Acid/metabolism
3.
J Thromb Haemost ; 8(6): 1343-52, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20149081

ABSTRACT

BACKGROUND: The nitric oxide (NO)/cyclic guanosine monophosphate (cGMP) signaling cascade is involved in the precise regulation of platelet responses. NO released from the endothelium is known to activate NO-sensitive guanylyl cyclase (NO-GC) in platelets. By the generation of cGMP and subsequent activation of cGMP-dependent protein kinase (PKG), NO-GC mediates the reduction of the intracellular calcium and inhibits platelet adhesion and aggregation. However, NO has been postulated to influence these platelet functions also via cGMP-independent mechanisms. OBJECTIVE: We studied the effect of NO on platelets lacking NO-sensitive guanylyl cyclase with regards to aggregation, adhesion, calcium mobilization and bleeding time. METHODS AND RESULTS: Here, we show that NO signaling leading to inhibition of agonist-induced platelet aggregation is totally abrogated in platelets from mice deficient in NO-GC (GCKO). Even at millimolar concentrations none of the several different NO donors inhibited collagen-induced aggregation of GCKO platelets. In addition, NO neither affected adenosine 5'-diphosphate (ADP)-induced adhesion nor thrombin-induced calcium release in GCKO platelets. Although the NO-induced cGMP signal transduction was totally abrogated cyclic adenosine monophosphate (cAMP) signaling was still functional; however, cGMP/cAMP crosstalk was disturbed on the level of phosphodiesterase type 3 (PDE3). These in vitro data are completed by a reduced bleeding time indicating the lack of NO effect in vivo. CONCLUSIONS: We conclude that NO-GC is the only NO receptor in murine platelets mediating the inhibition of calcium release, adhesion and aggregation: lack of the enzyme leads to disturbance of primary hemostasis.


Subject(s)
Blood Platelets/physiology , Guanylate Cyclase/metabolism , Nitric Oxide/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Animals , Cell Adhesion/physiology , Cyclic GMP/metabolism , Exocytosis , Guanylate Cyclase/genetics , Guanylate Cyclase/physiology , Mice , Mice, Knockout , Phosphorylation , Platelet Aggregation/physiology , Signal Transduction , Soluble Guanylyl Cyclase
4.
Biochem Soc Trans ; 33(Pt 5): 1119-22, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16246060

ABSTRACT

Most of the effects of the signalling molecule nitric oxide (NO) are mediated by the stimulation of the NO-sensitive GC (guanylate cyclase) and the subsequent increase in cGMP formation. The enzyme contains a prosthetic haem group, which mediates NO stimulation. In addition to the physiological activator NO, NO-sensitizers like the substance YC-1 sensitize the enzyme towards NO and may therefore have important pharmacological implications. Two isoforms of NO-sensitive GC have been identified to date that share regulatory properties, but differ in the subcellular localization. The more ubiquitously expressed alpha1beta1 heterodimer and the alpha2beta1 isoform are mainly expressed in brain. In intact cells, NO-induced cGMP signalling not only depends on cGMP formation, but is also critically determined by the activity of the enzymes responsible for cGMP degradation, e.g. PDE5 (phosphodiesterase 5). Recently, direct activation of PDE5 by cGMP was demonstrated, limiting the cGMP increase and thus functioning as a negative feedback. As the cGMP-induced PDE5 activation turned out to be sustained, in the range of hours, it is probably responsible for the NO-induced desensitization observed within NO/cGMP signalling.


Subject(s)
Cyclic GMP/physiology , Guanylate Cyclase/metabolism , Nitric Oxide/physiology , Signal Transduction/physiology , 3',5'-Cyclic-GMP Phosphodiesterases , Binding Sites , Cell Line , Cyclic Nucleotide Phosphodiesterases, Type 5 , Feedback , Guanosine Monophosphate/metabolism , Guanosine Triphosphate/metabolism , Humans , Models, Biological , Phosphoric Diester Hydrolases/metabolism
6.
J Cell Biol ; 155(2): 271-8, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11604422

ABSTRACT

Most of the effects of the signaling molecule nitric oxide (NO) are mediated by cGMP, which is synthesized by soluble guanylyl cyclase and degraded by phosphodiesterases. Here we show that in platelets and aortic tissue, NO led to a biphasic response characterized by a tremendous increase in cGMP (up to 100-fold) in less than 30 s and a rapid decline, reflecting the tightly controlled balance of guanylyl cyclase and phosphodiesterase activities. Inverse to the reported increase in sensitivity caused by NO shortage, concentrating NO attenuated the cGMP response in a concentration-dependent manner. We found that guanylyl cyclase remained fully activated during the entire course of the cGMP response; thus, desensitization was not due to a switched off guanylyl cyclase. However, when intact platelets were incubated with NO and then lysed, enhanced activity of phosphodiesterase type 5 was detected in the cytosol. Furthermore, this increase in cGMP degradation is paralleled by the phosphorylation of phosphodiesterase type 5 at Ser-92. Thus, our data suggest that NO-induced desensitization of the cGMP response is caused by the phosphorylation and subsequent activity increase of phosphodiesterase type 5.


Subject(s)
Blood Platelets/metabolism , Cyclic GMP/biosynthesis , Glutathione/analogs & derivatives , Muscle, Smooth, Vascular/metabolism , Nitric Oxide/physiology , Phosphoric Diester Hydrolases/metabolism , 3',5'-Cyclic-GMP Phosphodiesterases , Animals , Aorta , Blood Platelets/drug effects , Blood Platelets/enzymology , Cell Adhesion Molecules/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 5 , Enzyme Activation , Glutathione/pharmacology , Guanylate Cyclase/metabolism , Humans , In Vitro Techniques , Kinetics , Male , Microfilament Proteins , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/enzymology , Nitric Oxide Donors/pharmacology , Nitro Compounds/pharmacology , Phosphoproteins/metabolism , Phosphorylation , Rats , Rats, Inbred WKY
7.
J Biol Chem ; 276(48): 44647-52, 2001 Nov 30.
Article in English | MEDLINE | ID: mdl-11572861

ABSTRACT

The signaling molecule nitric oxide (NO) exerts most of its effects by the stimulation of the NO-sensitive guanylyl cyclase. Two isoforms of the NO receptor molecule exist: the ubiquitously occurring alpha(1)beta(1) and the alpha(2)beta(1) with a more limited distribution. As the isoforms are functionally indistinguishable, the physiological relevance of these isoforms remained unclear. The neuronal NO synthase has been reported to be associated with PSD-95. Here, we demonstrate the interaction of the so far unnoticed alpha(2)beta(1) isoform with PSD-95 in rat brain as shown by coprecipitation. The interaction is mediated by the alpha(2) C-terminal peptide and the third PDZ domain of PSD-95. As a consequence of the PSD-95 interaction, the so far considered "soluble" alpha(2)beta(1) isoform is recruited to the membrane fraction of synaptosomes, whereas the alpha(1)beta(1) isoform is found in the cytosol. Our results establish the alpha(1)beta(1) as the cytosolic and the alpha(2)beta(1) as the membrane-associated NO-sensitive guanylyl cyclase and suggest the alpha(2)beta(1) isoform as the sensor for the NO formed by the PSD-95-associated neuronal NO synthase.


Subject(s)
Guanylate Cyclase/metabolism , Nerve Tissue Proteins/metabolism , Animals , Blotting, Western , Brain/metabolism , Cyclic GMP/metabolism , Cytosol/metabolism , Dimerization , Disks Large Homolog 4 Protein , Guanylate Kinases , Intracellular Signaling Peptides and Proteins , Membrane Proteins , Nitric Oxide Synthase/metabolism , Peptides/chemistry , Precipitin Tests , Protein Binding , Protein Isoforms , Protein Structure, Tertiary , Rats , Synaptosomes/metabolism , Tumor Suppressor Proteins
8.
Nitric Oxide ; 5(3): 233-42, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11384196

ABSTRACT

In previous studies, a strong synergism between low concentrations of hydrogen peroxide and nitric oxide in the inhibition of agonist-induced platelet aggregation has been established and may be due to enhanced formation of cyclic GMP. In this investigation, hydrogen peroxide and NO had no effect on the activity of pure soluble guanylyl cyclase or its activity in platelet lysates and cytosol. H(2)O(2) was found to increase the phosphorylation of vasodilator-stimulated phosphoprotein (VASP), increasing the amount of the 50-kDa form that results from phosphorylation at serine(157). This occurs both in the presence and in the absence of low concentrations of NO, even at submicromolar concentrations of the peroxide, which alone was not inhibitory to platelets. These actions of H(2)O(2) were inhibited to a large extent by an inhibitor of cyclic AMP-dependent protein kinase, even though H(2)O(2) did not increase cyclic AMP. This inhibitor reversed the inhibition of platelets induced by combinations of NO and H(2)O(2) at low concentrations. The results suggest that the action on VASP may be one site of action of H(2)O(2) but that this event alone does not lead to inhibition of platelets; another unspecified action of NO is required to complete the events required for inhibition.


Subject(s)
Blood Platelets/physiology , Carbazoles , Cell Adhesion Molecules/metabolism , Cyclic GMP/blood , Guanylate Cyclase/blood , Hydrogen Peroxide/pharmacology , Indoles , Nitric Oxide/pharmacology , Phosphoproteins/metabolism , Platelet Aggregation/drug effects , 1-Methyl-3-isobutylxanthine/pharmacology , Alkaloids/pharmacology , Blood Platelets/drug effects , Blood Proteins/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/blood , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinases/blood , Cytosol/enzymology , Drug Synergism , Enzyme Inhibitors/pharmacology , Humans , Hydrazines/pharmacology , In Vitro Techniques , Indazoles/pharmacology , Kinetics , Microfilament Proteins , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Donors/pharmacology , Nitrogen Oxides , Phosphoserine/blood , Platelet Aggregation/physiology , Platelet Aggregation Inhibitors/pharmacology , Subcellular Fractions/metabolism , Thrombin/pharmacology
9.
Mol Pharmacol ; 59(2): 220-4, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11160856

ABSTRACT

YC-1 is a direct activator of soluble guanylyl cyclase (sGC) and sensitizes the enzyme for activation by nitric oxide (NO) and CO. Because the potentiating effect of YC-1 on NO-induced cGMP formation in platelets and smooth muscle cells has been shown to be substantially higher than observed with the purified enzyme, the synergism between heme ligands and YC-1 is apparently more pronounced in intact cells than in cell-free systems. Here, we investigated the mechanisms underlying the synergistic activation of sGC by YC-1 and NO in endothelial cells. Stimulation of the cells with YC-1 enhanced cGMP accumulation up to approximately 100-fold. The maximal effect of YC-1 was more pronounced than that of the NO donor DEA/NO (approximately 20-fold increase in cGMP accumulation) and markedly diminished in the presence of L-N(G)-nitroarginine, EGTA, or oxyhemoglobin. Because YC-1 did not activate endothelial NO synthase, the pronounced effect of YC-1 on cGMP accumulation was apparently caused by a synergistic activation of sGC by YC-1 and basal NO. The effect of YC-1 was further enhanced by addition of DEA/NO, resulting in a approximately 160-fold stimulation of cGMP accumulation. Thus, YC-1 increased the NO-induced accumulation of cGMP in intact cells by approximately 8-fold. Addition of endothelial cell homogenate increased the stimulatory effect of YC-1 on NO-activated purified sGC from 1.2- to 3.7-fold. This effect was not observed with heat-denatured homogenates, suggesting that a heat-labile factor present in endothelial cells potentiates the effect of YC-1 on NO-activated sGC.


Subject(s)
Endothelium, Vascular/drug effects , Enzyme Activators/pharmacology , Guanylate Cyclase/metabolism , Indazoles/pharmacology , Nitric Oxide/pharmacology , Animals , Cells, Cultured , Cyclic GMP/metabolism , Drug Synergism , Endothelium, Vascular/enzymology , Enzyme Activation , Enzyme Inhibitors/pharmacology , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase/metabolism , Swine
10.
Brain Res ; 891(1-2): 206-12, 2001 Feb 09.
Article in English | MEDLINE | ID: mdl-11164824

ABSTRACT

The localisation of particulate and soluble guanylyl cyclase was studied in hippocampal astrocytes. Counting the colocalisation of cGMP immunoreactivity with the astrocytic marker glial fibrillary acidic protein after stimulation of brain slices with sodium nitroprusside (0.1 mM) or atrial natriuretic peptide (100 nM), we were able to show that at least 67% of the hippocampal astrocytes contained both guanylyl cyclase isoforms. In addition, it was shown that a large number of atrial natriuretic peptide, brain-derived natriuretic peptide or sodium nitroprusside responsive cells contain the beta1-subunit of the soluble guanylyl cyclase. The results show that, in at least a subset of hippocampal astrocytes, soluble and particulate guanylyl cyclases are simultaneously present in the same cells.


Subject(s)
Astrocytes/enzymology , Guanylate Cyclase/metabolism , Hippocampus/enzymology , 1-Methyl-3-isobutylxanthine/pharmacology , Animals , Astrocytes/cytology , Astrocytes/drug effects , Atrial Natriuretic Factor/metabolism , Atrial Natriuretic Factor/pharmacology , Cyclic GMP/biosynthesis , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/cytology , Hippocampus/drug effects , Isoenzymes/metabolism , Male , Nitroprusside/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Rats , Rats, Inbred Lew , Solubility , Vasodilator Agents/pharmacology
11.
Brain Res ; 885(1): 6-13, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11121524

ABSTRACT

Guanylyl cyclases (GC) catalyze the formation of the intracellular signal molecule cyclic GMP from GTP. For some years it has been known that the heme-containing soluble guanylyl cyclase (sGC) is stimulated by NO and NO-containing compounds. The sGC enzyme consists of two subunits (alpha(1) and beta(1)). In the present study, the alpha(1) and beta(1)-subunits were identified in the guinea pig cochlea at the electron microscopic level using a post-embedding immuno-labeling procedure. Ultrathin sections of LR White embedded specimens were incubated with various concentrations of two rabbit polyclonal antibodies to the alpha(1)- and beta(1)-subunit, respectively. The immunoreactivity was visualized by a gold-labeled secondary antibody in an energy-filtering transmission electron microscope (EFTEM). Marked immunoreactivity for both antibodies was found in the inner and outer hair cells, with numerous gold particles at the border of the cuticular plates, associated with the cell nuclei or attached to electron-dense parts of the cytoplasm. In the pillar cells and apical Deiters cells, soluble guanylyl cyclase immunoreactivity was located at the rim of the cuticular plates and between the microtubuli bundles. Together with the recently identified nitric oxide synthase isoforms [Eur. Arch. Otorhinolaryngol. 254 (1997) 396; Eur. Arch. Otorhinolaryngol. 255 (1998) 483], the soluble guanylyl cyclase may be involved in signalling processes in the organ of Corti.


Subject(s)
Guanylate Cyclase/analysis , Hair Cells, Auditory, Inner/enzymology , Hair Cells, Auditory, Outer/enzymology , Animals , Antibodies , Guanylate Cyclase/immunology , Guinea Pigs , Hair Cells, Auditory, Inner/ultrastructure , Hair Cells, Auditory, Outer/ultrastructure , Microscopy, Immunoelectron , Nitric Oxide Synthase/analysis , Signal Transduction/physiology , Solubility , Tissue Embedding
12.
Cardiovasc Res ; 47(3): 602-8, 2000 Aug 18.
Article in English | MEDLINE | ID: mdl-10963733

ABSTRACT

OBJECTIVE: L-Ascorbic acid has been described to exert multiple beneficial effects in cardiovascular disorders associated with impaired nitric oxide (NO)/cGMP signalling. The aim of the present study was to investigate the effect of vitamin C on the most prominent physiological target of endogenous and exogenous NO, i.e. soluble guanylyl cyclase (sGC). METHODS: To address this issue we used a highly purified enzyme preparation from bovine lung (from the slaughterhouse). Enzymic activity was measured by a standard assay based on the conversion of [alpha-32P]GTP to [32P]cGMP and the subsequent quantification of the radiolabelled product. NO was quantified using a commercially available Clark-type electrode. RESULTS: Stimulation of sGC by the NO donor 2, 2-diethyl-1-nitroso-oxyhydrazine was inhibited by ascorbate with an IC(50) of approximately 2 microM. Maximal enzyme inhibition ( approximately 70%) was observed at 0.1-1 mM vitamin C. Stimulation of sGC by the NO-independent activator protoporphyrin-IX was also inhibited with similar potency. The effect of ascorbate on sGC was largely antagonised by reduced glutathione (1 mM) and the specific iron chelator diethylenetriaminepentaacetic acid (0.1 mM). Electrochemical experiments revealed that NO is potently scavenged by vitamin C. Consumption of NO by ascorbate was prevented by reduced glutathione (1 mM), diethylenetriaminepentaacetic acid (0.1 mM) and superoxide dismutase (500 units/ml) whereas up to 5000 units/ml superoxide dismutase failed to restore sGC activity. CONCLUSIONS: Our results suggest that physiological concentrations of L-ascorbic acid diminish cGMP accumulation via both scavenging of NO and direct inhibition of sGC.


Subject(s)
Antioxidants/pharmacology , Ascorbic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Guanylate Cyclase/antagonists & inhibitors , Animals , Cattle , Dose-Response Relationship, Drug , Electrochemistry , Hydrazines/pharmacology , Lung/enzymology , Nitric Oxide/analysis , Nitric Oxide Donors/pharmacology , Nitroso Compounds/pharmacology , Oxidation-Reduction
13.
Br J Pharmacol ; 130(2): 205-8, 2000 May.
Article in English | MEDLINE | ID: mdl-10807655

ABSTRACT

We used YC-1 as a pharmacological tool to investigate the short-term blood pressure effects of NO-independent activation of sGC in normotensive and hypertensive rats. Four groups of normotensive Wistar-Kyoto rats were treated by i.v. injection with vehicle (V), YC-1 (YC-1), sodium nitroprusside (SNP), or YC-1 and SNP (YC-1+SNP). Hypertension was induced in four additional groups of WKY rats by 3 weeks of oral treatment with L-NAME. These animals were investigated with the same protocol as the normotensive animals: L-NAME/V, L-NAME/YC-1, L-NAME/SNP, L-NAME/YC-1+SNP. YC-1 lowered mean arterial blood pressure (MAP) in normotensive and hypertensive animals similarly to SNP alone (P<0.05, respectively). The combination of YC-1 with SNP caused a strong decrease of MAP in both the hypertensive and normotensive animals (P<0.05, respectively). SNP with YC-1 also induced a pronounced cyclic GMP increase in the aorta. This study shows for the first time the blood pressure lowering potential of bimodal targeting of the NO-sGC-system.


Subject(s)
Blood Pressure/drug effects , Guanylate Cyclase/metabolism , Hypertension/enzymology , Indazoles/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Animals , Drug Interactions , Enzyme Activation , Enzyme Inhibitors/pharmacology , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Rats , Rats, Inbred WKY
14.
Biochem J ; 347(Pt 2): 475-84, 2000 Apr 15.
Article in English | MEDLINE | ID: mdl-10749677

ABSTRACT

Nitric oxide synthase (NOS) catalysis results in formation of NO or superoxide (O(2)(-.)) depending on the presence or absence of the cofactor tetrahydrobiopterin (BH4). In the absence of O(2)(-.) scavengers, net NO formation cannot be detected even at saturating BH4 concentrations, which is thought to be due to O(2)(-.) production by BH4 autoxidation. Because the N-5-methylated analogue of BH4 (5-Me-BH4) sustains NOS catalysis and is autoxidation-resistant, net NO formation by the neuronal isoform of NOS (nNOS) can be observed at saturating 5-Me-BH4 concentrations. Here we compare the effects of 5-Me-BH4 on L-citrulline formation, NADPH oxidation, H(2)O(2) production and soluble guanylate cyclase (sGC) stimulation. All activities were stimulated biphasically (EC(50) approx. 0.2 microM and more than 1 mM), with an intermediate inhibitory phase at the same pterin concentration as that required for net NO generation and sGC stimulation (4 microM). Concomitantly with inhibition, the NADP(+)/L-citrulline stoichiometry decreased from 2.0 to 1.6. Inhibition occurred only at high enzyme concentrations (IC(50) approx. 10 nM nNOS) and was antagonized by oxyhaemoglobin and by BH4. We ascribe the first stimulatory phase to high-affinity binding of 5-Me-BH4. The inhibitory phase is due to low-affinity binding, resulting in fully coupled catalysis, complete inhibition of O(2)(-.) production and net NO formation. At high enzyme concentrations and thus high NO levels, this causes autoinhibition. NO scavenging by 5-Me-BH4 at concentrations above 1 mM, resulting in the antagonization of inhibition of NOS, explains the second stimulatory phase. In agreement with these assignments 5-Me-BH4 was found to stimulate formation of a haem-NO complex during NOS catalysis. The observation of inhibition with 5-Me-BH4 but not with BH4 implies that, unless O(2)(-.) scavengers are present, a physiological role for NO-induced autoinhibition is unlikely.


Subject(s)
Biopterins/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide/pharmacology , Pteridines/metabolism , Animals , Arginine/metabolism , Biopterins/analogs & derivatives , Biopterins/pharmacology , Catalase/metabolism , Catalysis , Cattle , Citrulline/metabolism , Dimerization , Free Radical Scavengers/metabolism , Guanylate Cyclase/metabolism , Heme/metabolism , Hydrogen Peroxide/metabolism , NADP/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type I , Oxidants/metabolism , Pteridines/pharmacology , Rats , Superoxide Dismutase/metabolism , Superoxides/metabolism
15.
Methods ; 19(4): 485-93, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10581148

ABSTRACT

Soluble guanylyl cyclase acts as the receptor for the signaling molecule nitric oxide. The enzyme consists of two different subunits. Each subunit shows the cyclase catalytic domain, which is also conserved in the membrane-bound guanylyl cyclases and the adenylyl cyclases. The N-terminal regions of the subunits are responsible for binding of the prosthetic heme group of the enzyme, which is required for the stimulatory effect of nitric oxide (NO). The five-coordinated ferrous heme displays a histidine as the axial ligand; activation of soluble guanylyl cyclase by NO is initiated by binding of NO to the heme iron and proceeds via breaking of the histidine-to-iron bond. Recently, a novel pharmacological and possibly physiological principle of guanylyl cyclase sensitization was demonstrated. The substance YC-1 has been shown to activate the enzyme independent of NO, to potentiate the effect of submaximally effective NO concentrations, and to turn carbon monoxide into an effective activator of soluble guanylyl cyclase.


Subject(s)
Guanylate Cyclase/chemistry , Guanylate Cyclase/physiology , Animals , Biochemistry/methods , Cysteine/genetics , DNA Mutational Analysis , Enzyme Activation , Guanylate Cyclase/isolation & purification , Heme , Histidine/genetics , Isoenzymes/chemistry , Isoenzymes/isolation & purification , Solubility
16.
Biochemistry ; 38(46): 15253-7, 1999 Nov 16.
Article in English | MEDLINE | ID: mdl-10563809

ABSTRACT

Guanylyl cyclases (GCs) and adenylyl cyclases (ACs) play key roles in various signaling cascades and are structurally closely related. The crystal structure of a soluble AC revealed one binding site each for the substrate ATP and the activator forskolin. Recently, YC-1, a novel activator of the heterodimeric soluble GC (sGC), has been identified which acts like forskolin on AC. Here, we investigated the respective substrate and potential activator domains of sGC using point-mutated subunits. Whereas substitution of the conserved Cys-541 of the beta(1) subunit with serine led to an almost complete loss of activity, mutation of the respective homologue (Cys-596) in the alpha(1) subunit yielded an enzyme with an increased catalytic rate and higher sensitivity toward NO. This phenotype exhibits characteristics similar to those of the YC-1-treated wild-type enzyme. Conceivably, this domain which corresponds to the forskolin site of the ACs may comprise the binding site for YC-1.


Subject(s)
Enzyme Activators/pharmacology , Guanylate Cyclase/genetics , Guanylate Cyclase/metabolism , Indazoles/pharmacology , Point Mutation , Animals , Binding Sites/genetics , Catalytic Domain/genetics , Cattle , Conserved Sequence/genetics , Cyclic GMP/metabolism , Dimerization , Enzyme Activators/metabolism , Guanosine Triphosphate/metabolism , Indazoles/metabolism , Magnesium/metabolism , Manganese/metabolism , Mutagenesis, Site-Directed , Nitric Oxide/metabolism , Nitric Oxide/physiology , Solubility
17.
Biochemistry ; 38(33): 10699-706, 1999 Aug 17.
Article in English | MEDLINE | ID: mdl-10451364

ABSTRACT

Previous work has proved that the enzyme-soluble guanylate cyclase, GC, is activated several 100-fold by the combination of carbon monoxide plus a benzylindazole derivative called YC-1. That is about the same as activation by nitric oxide, which has a well-established role both in vivo and in vitro. This report addresses several spectroscopic, equilibrium, and kinetic effects wrought by YC-1 on carboxyl guanylate cyclase, including the following: a shift in the Soret absorption band by 4 nm to shorter wavelength; an increase in CO affinity by an order of magnitude; a dramatic change in the kinetics of CO association. After photolytic dissociation of CO, the majority, but not all, of bimolecular ligand recombination occurs with a time constant about 1000-fold faster than in the absence of YC-1, while a smaller fraction recombines almost, but not quite, the same as usual. This is reminiscent of the kinetics of NO association with GC, which also shows two prominent phases. The results just listed pertain in the presence of GTP/cGMP, which would be present during enzyme catalysis. Qualitatively similar, but smaller, effects occur in the absence of GTP/cGMP. Measurements are reported to characterize other changes in buffer conditions. The results are consistent with a mechanistic model that attributes a crucial role to the proximal bond that connects the heme iron to a histidine side chain in GC but also requires protein control of the distal environment.


Subject(s)
Carbon Monoxide/chemistry , Carbon Monoxide/metabolism , Guanylate Cyclase/chemistry , Guanylate Cyclase/metabolism , Indazoles/chemistry , Indazoles/metabolism , Animals , Cattle , Guanosine Triphosphate/chemistry , Guanosine Triphosphate/metabolism , Kinetics , Ligands , Models, Chemical , Nitric Oxide/chemistry , Nitric Oxide/metabolism , Photolysis , Platelet Aggregation Inhibitors/chemistry , Platelet Aggregation Inhibitors/metabolism , Solubility , Spectrophotometry, Ultraviolet
18.
J Biol Chem ; 274(29): 20083-91, 1999 Jul 16.
Article in English | MEDLINE | ID: mdl-10400618

ABSTRACT

Analysis of purified soybean and rabbit reticulocyte 15-lipoxygenase (15-LOX) and PA317 cells transfected with human 15-LOX revealed a rapid rate of linoleate-dependent nitric oxide (.NO) uptake that coincided with reversible inhibition of product ((13S)-hydroperoxyoctadecadienoic acid, or (13S)-HPODE) formation. No reaction of .NO (up to 2 microM) with either native (Ered) or ferric LOXs (0.2 microM) metal centers to form nitrosyl complexes occurred at these .NO concentrations. During HPODE-dependent activation of 15-LOX, there was consumption of 2 mol of .NO/mol of 15-LOX. Stopped flow fluorescence spectroscopy showed that.NO (2.2 microM) did not alter the rate or extent of (13S)-HPODE-induced tryptophan fluorescence quenching associated with 15-LOX activation. Additionally, .NO does not inhibit the anaerobic peroxidase activity of 15-LOX, inferring that the inhibitory actions of .NO are due to reaction with the enzyme-bound lipid peroxyl radical, rather than impairment of (13S)-HPODE-dependent enzyme activation. From this, a mechanism of 15-LOX inhibition by .NO is proposed whereby reaction of .NO with EredLOO. generates Ered and LOONO, which hydrolyzes to (13S)-HPODE and nitrite (NO2-). Reactivation of Ered, considerably slower than dioxygenase activity, is then required to complete the catalytic cycle and leads to a net inhibition of rates of (13S)-HPODE formation. This reaction of .NO with 15-LOX inhibited. NO-dependent activation of soluble guanylate cyclase and consequent cGMP production. Since accelerated .NO production, enhanced 15-LOX gene expression, and 15-LOX product formation occurs in diverse inflammatory conditions, these observations indicate that reactions of .NO with lipoxygenase peroxyl radical intermediates will result in modulation of both .NO bioavailability and rates of production of lipid signaling mediators.


Subject(s)
Arachidonate 15-Lipoxygenase/metabolism , Guanylate Cyclase/metabolism , Nitric Oxide/metabolism , Animals , Arachidonate 15-Lipoxygenase/genetics , Catalysis , Enzyme Activation , Humans , Kinetics , Linoleic Acid/metabolism , Oxidation-Reduction , Rabbits , Glycine max/enzymology , Transfection
20.
Biochem Biophys Res Commun ; 254(1): 188-91, 1999 Jan 08.
Article in English | MEDLINE | ID: mdl-9920755

ABSTRACT

Recently it has been reported that in the presence of YC-1, a benzyl indazole derivative, carbon monoxide activates soluble guanylate cyclase, GC, to about the same extent as its best known activator, nitric oxide. Kinetic studies utilizing flash photolysis of GC complexed with CO in the presence and absence of YC-1 show, in contrast to another recent report of a mixing experiment, that YC-1 has a profound effect on bimolecular association kinetics and a smaller, but significant, effect on ligand affinity. Most prominent is the appearance of a major, new phase in the bimolecular recombination kinetics in the presence of 200 microM YC-1: This major fraction rebinds CO approximately 1000-fold more rapidly than in the absence of YC-1. Another portion, considerably less than half, exhibits kinetics that are almost exactly the same as in the absence of YC-1. It is now clear that both YC-1 and CO have a strong synergistic effect on enzyme activity and also a dramatic effect on ligand binding behavior. It is, therefore, a reasonable inference that ligand binding at the heme iron atom is intimately connected with enzyme activation, a hypothesis that would have been difficult to maintain if the earlier report, that YC-1 has no effect on CO binding, were correct. Possible reasons for the discrepancy between the two measurements are suggested.


Subject(s)
Carbon Monoxide/chemistry , Guanylate Cyclase/chemistry , Indazoles/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Animals , Carbon Monoxide/metabolism , Enzyme Activation/drug effects , Guanylate Cyclase/metabolism , Humans , Kinetics , Ligands , Photolysis
SELECTION OF CITATIONS
SEARCH DETAIL
...