Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
BMC Neurosci ; 20(1): 1, 2019 Jan 03.
Article in English | MEDLINE | ID: mdl-30602386

ABSTRACT

BACKGROUND: Peripheral diabetic neuropathy can be painful and its symptoms include hyperalgesia, allodynia and spontaneous pain. Hydrogen sulfide (H2S) is involved in diabetes-induced hyperalgesia and allodynia. However, the molecular target through which H2S induces hyperalgesia in diabetic animals is unclear. The aim of this study was to determine the possible involvement of transient receptor potential (TRP) channels in H2S-induced hyperalgesia in diabetic rats. RESULTS: Streptozotocin (STZ) injection produced hyperglycemia in rats. Intraplantar injection of NaHS (an exogenous donor of H2S, 3-100 µg/paw) induced hyperalgesia, in a time-dependent manner, in formalin-treated diabetic rats. NaHS-induced hyperalgesia was partially prevented by local intraplantar injection of capsazepine (0.3-3 µg/paw), HC-030031 (100-316 µg/paw) and SKF-96365 (10-30 µg/paw) blockers, at 21 days post-STZ injection. At the doses used, these blockers did not modify formalin-induced nociception. Moreover, capsazepine (0.3-30 µg/paw), HC-030031 (100-1000 µg/paw) and SKF-96365 (10-100 µg/paw) reduced formalin-induced nociception in diabetic rats. Contralateral injection of the highest doses used did not modify formalin-induced flinching behavior. Hyperglycemia, at 21 days, also increased protein expression of cystathionine-ß-synthase enzyme (CBS) and TRPC6, but not TRPA1 nor TRPV1, channels in dorsal root ganglia (DRG). Repeated injection of NaHS enhanced CBS and TRPC6 expression, but hydroxylamine (HA) prevented the STZ-induced increase of CBS protein. In addition, daily administration of SKF-96365 diminished TRPC6 protein expression, whereas NaHS partially prevented the decrease of SKF-96365-induced TRPC6 expression. Concordantly, daily intraplantar injection of NaHS enhanced, and HA prevented STZ-induced intraepidermal fiber loss, respectively. CBS was expressed in small- and medium-sized cells of DRG and co-localized with TRPV1, TRPA1 and TRPC6 in IB4-positive neurons. CONCLUSIONS: Our data suggest that H2S leads to hyperalgesia in diabetic rats through activation of TRPV1, TRPA1 and TRPC channels and, subsequent intraepidermal fibers loss. CBS enzyme inhibitors or TRP-channel blockers could be useful for treatment of painful diabetic neuropathy.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Hydrogen Sulfide/metabolism , Hyperalgesia/metabolism , Transient Receptor Potential Channels/metabolism , Acetanilides/pharmacology , Analgesics/pharmacology , Animals , Capsaicin/analogs & derivatives , Capsaicin/pharmacology , Cystathionine beta-Synthase/metabolism , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/pathology , Female , Formaldehyde , Hydroxylamine/pharmacology , Hyperalgesia/drug therapy , Hyperalgesia/pathology , Imidazoles/pharmacology , Nociception/drug effects , Nociception/physiology , Purines/pharmacology , Rats, Wistar , Skin/innervation , Skin/metabolism , Spinal Nerve Roots/drug effects , Spinal Nerve Roots/metabolism , Spinal Nerve Roots/pathology , Sulfites
2.
J Neurosci Res ; 97(4): 520-534, 2019 04.
Article in English | MEDLINE | ID: mdl-30549080

ABSTRACT

Astrogliosis and microglial reactions are correlated with the formation of scar tissue and synapse loss. 4-hydroxy-tempo (TEMPOL) is a reactive oxygen species scavenger with proven neuroprotective efficacy in experimental models of traumatic injury and cerebral ischemia. TEMPOL has not, however, been applied following ventral root lesions, which are particularly correlated with the degeneration of spinal motoneurons following brachial plexus injuries. Thus, the present study investigated the effects of TEMPOL on motoneurons and adjacent glial reactions, with a particular focus on the preservation of excitatory and inhibitory spinal circuits. Adult female Sprague Dawley rats were subjected to ventral root crush (VRC) at the lumbar intumescence. Animals were divided into the following experimental groups: (a) VRC-saline treatment; (b) VRC-TEMPOL treatment (12 mg/kg, n = 5), and (c) VRC-TEMPOL treatment (250 mg/kg, n = 5). The spinal cord tissue located contralateral to the lesion was used as the control. Fourteen days after lesioning, the rats were euthanized and the spinal cords were removed for motoneuron counting and immunolabeling with glial (GFAP and Iba-1) and synapse markers (synaptophysin, VGLUT-1, and GAD65). Although TEMPOL did not exert neuroprotective effects at the studied concentrations, the modulation of glial reactions was significant at higher doses. Thus, synaptophysin staining was preserved and, in particular, VGLUT-1-positive inputs were maintained, thereby indicating that TEMPOL preserved proprioceptive glutamatergic inputs without exacerbating the rate of motoneuron degeneration. Consequently, its administration with other efficient neuroprotective substances may significantly improve the outcomes following spinal cord lesioning.


Subject(s)
Cyclic N-Oxides/pharmacology , Hydroxylamine/pharmacology , Neuroglia/drug effects , Neuroprotective Agents/pharmacology , Spinal Nerve Roots/injuries , Spinal Nerve Roots/metabolism , Synapses/drug effects , Animals , Antioxidants , Female , Glial Fibrillary Acidic Protein/metabolism , Gliosis , Motor Neurons/pathology , Rats , Rats, Sprague-Dawley , Spinal Cord Lateral Horn/metabolism , Spinal Nerve Roots/pathology , Synaptophysin/metabolism , Vesicular Glutamate Transport Protein 1/metabolism
3.
Neuroscience ; 250: 786-97, 2013 Oct 10.
Article in English | MEDLINE | ID: mdl-23830907

ABSTRACT

Hydrogen sulfide (H2S) is a gasotransmitter endogenously generated from the metabolism of L-cysteine by action of two main enzymes called cystathionine ß-synthase (CBS) and cystathionine γ-lyase (CSE). This gas has been involved in the pain processing and insulin resistance produced during diabetes development. However, there is no evidence about its participation in the peripheral neuropathy induced by this metabolic disorder. Experimental diabetes was induced by streptozotocin (50mg/kg, i.p.) in female Wistar rats. Streptozotocin injection increased formalin-evoked flinching in diabetic rats as compared to non-diabetic rats after 2 weeks. Peripheral administration of NaHS (an exogenous donor of H2S) and L-cysteine (an endogenous donor of H2S) dose-dependently increased flinching behavior in diabetic and non-diabetic rats. Contrariwise, hydroxylamine (HA, a CBS inhibitor) and DL-propargylglycine (PPG, a CSE inhibitor) decreased formalin-induced nociceptive behavior in both experimental groups. In addition, an ineffective dose of HA and PPG partially prevented the L-cysteine-induced hyperalgesia in diabetic and non-diabetic rats. Interestingly, HA and PPG were three order of magnitude more potent in diabetic rats respect to non-diabetic rats, whereas NaHS was ten times more potent in the streptozotocin-diabetic group. Nine to 11 weeks after diabetes induction, tactile allodynia was observed in the streptozotocin-injected rats. On this condition, subcutaneous administration of PPG or HA reduced tactile allodynia in diabetic rats. Paradoxically, H2S levels were decreased in nerve sciatic, dorsal root ganglion and spinal cord, but not paw nor blood plasma, during diabetes-associated peripheral neuropathy development. Collectively, results suggest that H2S synthesized by CBS and CSE participate in formalin-induced nociception in diabetic and non-diabetic rats, as well as; in tactile allodynia in streptozotocin-injected rats. In addition, data seems to indicate that diabetic rats are more sensible to H2S-induced hyperalgesia than normoglycemic rats.


Subject(s)
Diabetes Mellitus, Experimental/complications , Hydrogen Sulfide/pharmacology , Nociception/physiology , Algorithms , Alkynes/pharmacology , Animals , Blood Glucose/metabolism , Cystathionine gamma-Lyase/metabolism , Cysteine/antagonists & inhibitors , Cysteine/pharmacology , Data Interpretation, Statistical , Female , Glycine/analogs & derivatives , Glycine/pharmacology , Hydrogen Sulfide/metabolism , Hydroxylamine/pharmacology , Hyperalgesia/chemically induced , Hyperalgesia/psychology , Neural Pathways/drug effects , Pain/psychology , Pain Measurement , Physical Stimulation , Rats , Rats, Wistar
4.
Fundam Clin Pharmacol ; 22(3): 299-304, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18485148

ABSTRACT

South American (SA) opossum lower esophageal sphincter (LES) circular smooth muscle relaxes by activation of enteric nerves elicited by EFS (electrical field stimulation, 0.5 ms, 48 V, 0.5-8 Hz for 10 s). The identity of the mediator released and the cellular mechanism, however, remain to be fully elucidated. The purpose of this study was to determine the effect of the enzyme soluble guanylate cyclase (cGC) inhibitors, cystamine (100 microM), methylene blue (30 microM), LY 83583 (6-anilino-5,8 quinoledione, 10 microM) and ODQ (H-[1,2,4]oxadiazolo[4,3]quinoxalin-1-one, 1 microM) on the relaxations induced by EFS and by exogenous NO (nitric oxide, 0.5 mM) or NO-donors on SA opossum LES smooth muscle strips. EFS caused frequency-dependent relaxations, which were inhibited by NO-synthase inhibitors and abolished by tetrodotoxin. Cystamine did not affect relaxations caused by EFS and NO or NO-donor. Methylene blue also failed to affect EFS-caused relaxations, although it was capable of inhibiting relaxation induced by NO. LY 83583 inhibited relaxations induced by NO, but did not affect those induced by EFS or by SNAP and HXA. ODQ abolished relaxations caused by EFS at lower frequencies and by HXA (hydroxylamine, 10 microM) and SNAP (S-nitroso-N-acetyl penicillamine, 10 microM). Relaxations at higher frequencies of EFS and induced by SNP (sodium nitroprusside, 30 microM) and NO were only reduced by ODQ. These findings indicate that activation of the cGC can be involved in relaxations induced by EFS at lower frequencies, but other mechanisms can be involved at higher frequencies of EFS and caused by SNP or NO.


Subject(s)
Esophageal Sphincter, Lower/physiology , Guanylate Cyclase/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Aminoquinolines/pharmacology , Animals , Cyclic GMP/metabolism , Cysteamine/pharmacology , Electric Stimulation , Esophageal Sphincter, Lower/innervation , Esophageal Sphincter, Lower/metabolism , Female , Hydrazines/pharmacology , Hydroxylamine/pharmacology , In Vitro Techniques , Male , Methylene Blue/pharmacology , Muscle Relaxation/drug effects , Nitric Oxide/physiology , Nitric Oxide Donors/pharmacology , Nitroprusside/pharmacology , Opossums , Oxadiazoles/pharmacology , Penicillamine/analogs & derivatives , Penicillamine/pharmacology , Soluble Guanylyl Cyclase
5.
Biochimie ; 89(11): 1425-32, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17614193

ABSTRACT

We show that MDCK I cells express, besides the classical (Na(+)+K(+))ATPase, a Na(+)-stimulated ATPase activity with the following characteristics: (1) K(0.5) for Na(+) 7.5+/-1.5 mM and V(max) 23.12+/-1.1 nmol Pi/mg per min; (2) insensitive to 1 mM ouabain and 30 mM KCl; and (3) inhibited by furosemide and vanadate (IC(50) 42.1+/-8.0 and 4.3+/-0.3 microM, respectively). This enzyme forms a Na(+)-stimulated, furosemide- and hydroxylamine-sensitive ATP-driven acylphosphate phosphorylated intermediate with molecular weight of 100 kDa. Immunoprecipitation of the (Na(+)+K(+))ATPase with monoclonal anti-alpha(1) antibody reduced its activity in the supernatant by 90%; the Na(+)-ATPase activity was completely maintained. In addition, the formation of the Na(+)-stimulated, furosemide- and hydroxylamine-sensitive ATP-driven acylphosphate intermediate occurred at the same magnitude as that observed before immunoprecipitation. These data suggest that Na(+)-ATPase and (Na(+)+K(+))ATPase activities are independent, with Na(+)-ATPase belonging to a different enzyme entity.


Subject(s)
Adenosine Triphosphatases/isolation & purification , Adenosine Triphosphatases/metabolism , Ouabain/pharmacology , Sodium-Potassium-Exchanging ATPase/isolation & purification , Sodium-Potassium-Exchanging ATPase/metabolism , Adenosine Triphosphatases/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Animals , Cell Line , Dogs , Enzyme Inhibitors/pharmacology , Furosemide/pharmacology , Hydrolysis/drug effects , Hydroxylamine/pharmacology , Immunoblotting , Immunoprecipitation , Kinetics , Phosphorylation/drug effects , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Vanadates/pharmacology
6.
J Neural Transm (Vienna) ; 114(6): 863-5, 2007.
Article in English | MEDLINE | ID: mdl-17385063

ABSTRACT

The endogenous compound hydroxylamine relaxes vascular smooth muscle in vitro, apparently through conversion to the vasodilator factor nitric oxide, but its effect on blood pressure has not been characterized. We found that in the anesthetized rat the amine elicits dose-related hypotension when administered by continuous iv infusion. In experiments designed to explore the mechanism of this effect, hydroxylamine was compared with the nitric oxide donor nitroprusside and the direct-acting vasodilator hydralazine, using pretreatments known to modify diverse mechanisms of vasodilation. Hydroxylamine hypotension was enhanced by the SSAO inhibitor isoniazid and the SSAO substrate methylamine, a pattern shared by hydralazine. Responses were blocked by the guanylate cyclase inhibitor methylene blue and were increased by the nitric oxide synthase inhibitor L-NAME, a pattern shared by nitroprusside. It was concluded that hydroxylamine exerts hypotension partly through conversion to nitric oxide and partly by a "hydralazine-like" mechanism involving SSAO inhibition.


Subject(s)
Amine Oxidase (Copper-Containing)/antagonists & inhibitors , Blood Pressure/drug effects , Enzyme Inhibitors/pharmacology , Hydroxylamine/pharmacology , Hypotension/chemically induced , Hypotension/enzymology , Nitric Oxide/metabolism , Amine Oxidase (Copper-Containing)/metabolism , Animals , Blood Pressure/physiology , Dose-Response Relationship, Drug , Guanylate Cyclase/antagonists & inhibitors , Guanylate Cyclase/metabolism , Hydralazine/pharmacology , Hydroxylamine/metabolism , Hypotension/physiopathology , Injections, Intravenous , Isoniazid/pharmacology , Male , Methylamines/metabolism , Methylamines/pharmacology , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Nitroprusside/pharmacology , Rats , Rats, Wistar , Vasodilator Agents/pharmacology
7.
J Biol Chem ; 280(34): 30611-8, 2005 Aug 26.
Article in English | MEDLINE | ID: mdl-15987689

ABSTRACT

The aim of this study was to investigate (a) whether Ca2+/calmodulin-dependent protein kinase II (CaM kinase II) participates in the regulation of plasma membrane Ca2+-ATPase and (b) its possible cross-talk with other kinase-mediated modulatory pathways of the pump. Using isolated innervated membranes of the electrocytes from Electrophorus electricus L., we found that stimulation of endogenous protein kinase A (PKA) strongly phosphorylated membrane-bound CaM kinase II with simultaneous substantial activation of the Ca2+ pump (approximately 2-fold). The addition of cAMP (5-50 pM), forskolin (10 nM), or cholera toxin (10 or 100 nM) stimulated both CaM kinase II phosphorylation and Ca2+-ATPase activity, whereas these activation processes were cancelled by an inhibitor of the PKA alpha-catalytic subunit. When CaM kinase II was blocked by its specific inhibitor KN-93, the Ca2+-ATPase activity decreased to the levels measured in the absence of calmodulin; the unusually high Ca2+ affinity dropped 2-fold; and the PKA-mediated stimulation of Ca2+-ATPase was no longer seen. Hydroxylamine-resistant phosphorylation of the Ca2+-ATPase strongly increased when the PKA pathway was activated, and this phosphorylation was suppressed by inhibition of CaM kinase II. We conclude that CaM kinase II is an intermediate in a complex regulatory network of the electrocyte Ca2+ pump, which also involves calmodulin and PKA.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/physiology , Calcium-Transporting ATPases/metabolism , Calmodulin/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Electrophorus/physiology , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Animals , Benzylamines/pharmacology , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Calmodulin/chemistry , Catalytic Domain , Cell Membrane/enzymology , Cell Membrane/metabolism , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/chemistry , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Electrophorus/metabolism , Hydroxylamine/chemistry , Hydroxylamine/pharmacology , Immunoblotting , Kinetics , Models, Chemical , Phosphorylation , Protein Binding , Sulfonamides/pharmacology , Thapsigargin/pharmacology , Time Factors
8.
J Biol Chem ; 278(16): 13789-94, 2003 Apr 18.
Article in English | MEDLINE | ID: mdl-12571240

ABSTRACT

Entry of enveloped animal viruses into their host cells always depends on a step of membrane fusion triggered by conformational changes in viral envelope glycoproteins. Vesicular stomatitis virus (VSV) infection is mediated by virus spike glycoprotein G, which induces membrane fusion at the acidic environment of the endosomal compartment. VSV-induced membrane fusion occurs at a very narrow pH range, between 6.2 and 5.8, suggesting that His protonation is required for this process. To investigate the role of His in VSV fusion, we chemically modified these residues using diethylpyrocarbonate (DEPC). We found that DEPC treatment inhibited membrane fusion mediated by VSV in a concentration-dependent manner and that the complete inhibition of fusion was fully reversed by incubation of modified virus with hydroxylamine. Fluorescence measurements showed that VSV modification with DEPC abolished pH-induced conformational changes in G protein, suggesting that His protonation drives G protein interaction with the target membrane at acidic pH. Mass spectrometry analysis of tryptic fragments of modified G protein allowed the identification of the putative active His residues. Using synthetic peptides, we showed that the modification of His-148 and His-149 by DEPC, as well as the substitution of these residues by Ala, completely inhibited peptide-induced fusion, suggesting the direct participation of these His in VSV fusion.


Subject(s)
Histidine/chemistry , Membrane Fusion , Phosphorylcholine/analogs & derivatives , Vesicular stomatitis Indiana virus/metabolism , Alanine/chemistry , Amino Acid Sequence , Amino Acids/chemistry , Cell Membrane/metabolism , Dose-Response Relationship, Drug , Glycoproteins/metabolism , Hydrogen-Ion Concentration , Hydroxylamine/pharmacology , Liposomes/metabolism , Molecular Sequence Data , Peptide Biosynthesis , Peptides/chemistry , Phosphorylcholine/chemistry , Protein Conformation , Protons , Spectrometry, Fluorescence , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Time Factors
9.
Free Radic Biol Med ; 31(11): 1323-33, 2001 Dec 01.
Article in English | MEDLINE | ID: mdl-11728803

ABSTRACT

Catalase-1 (Cat-1), one of the two monofunctional catalases of Neurospora crassa, increases during asexual spore formation to constitute 0.6% of total protein in conidia. Cat-1 was purified 170-fold with a yield of 48% from conidiating cultures. Like most monofunctional catalases, Cat-1 is a homotetramer, resistant to inactivation by solvents, fully active over a pH range of 4-12, and inactivated by 3-amino-1,2,4-triazole. Unlike most monofunctional catalases, Cat-1 consists of 88 kDa monomers that are glycosylated with alpha-glucose and/or alpha-mannose, is unusually stable, and is not inactivated or inhibited by hydrogen peroxide. Cat-1 was more resistant than other catalases to heat inactivation and to high concentrations of salt and denaturants. Cat-1 exhibited unusual kinetics: at molar concentrations of hydrogen peroxide the apparent V was 10 times higher than at millimolar concentrations. Inactivation of Cat-1 activity with azide and hydroxylamine was according to first order kinetics, while cyanide at micromolar concentrations was a reversible competitive inhibitor.


Subject(s)
Catalase/genetics , Catalase/metabolism , Neurospora crassa/enzymology , Azides/pharmacology , Catalase/chemistry , Catalysis , Dimerization , Electrophoresis, Polyacrylamide Gel , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Enzyme Stability , Glucose/metabolism , Glycosylation , Hot Temperature , Hydrogen Peroxide/pharmacology , Hydrogen-Ion Concentration , Hydroxylamine/pharmacology , Kinetics , Macromolecular Substances , Mannose/metabolism , Protein Denaturation
10.
Life Sci ; 68(6): 709-17, 2000 Dec 29.
Article in English | MEDLINE | ID: mdl-11205885

ABSTRACT

Activation of inhibitory nonadrenergic noncholinergic (NANC) nerves in the rat duodenum cause relaxations, which are reduced by nitric oxide synthase (NOS) inhibitors indicating that this response involves a nitrergic neurotransmission. The precise nature of the nitrergic neurotransmitter is still controversial since nitric oxide (NO) scavengers and superoxide generators, even in the presence of superoxide dismutase inhibitors, failed to inhibit nitrergic neurotransmission mediated relaxations. In order to understand the role of NOS in nitrergic neurotransmission and considering that N-OH-arginine (OH-L-Arg), L-citrulline, NO, S-nitrosoglutathione (GSNO) and hydroxylamine (NH2OH) can be formed in cells during the N(G)-oxidation of L-arginine catalyzed by NOS we explored whether any of these products could exhibit biological properties comparable to those of the nitrergic neurotransmitter. After establishing which of them was able to relax the rat duodenum, the pharmacological profile of such effect was determined employing oxyhemoglobin (OxyHb), pyrogallol (PYR), hydroquinone (HQ), hydroxocobalamin (HC) or carboxy-PTIO (C-PTIO) and compared with that of nerve mediated relaxations. NO, GSNO and NH2OH, but not OH-L-ARG and L-citrulline, caused concentration-dependent relaxations that were not affected by tetrodotoxin or L-NOARG. OxyHb almost abolished NO-induced relaxations but decreased only marginally the magnitude of nerve-, NH2OH- and SNG-induced relaxations. PYR, HQ and C-PTIO reduced significantly GSNO- and NO- induced relaxations but did not affect those induced by NH2OH or nerve activation. In contrast, HC abolished NO-induced relaxations while it did not affect those induced by GSNO, NH2OH and nerve activation. The catalase inhibitor 1,2,4 aminotriazole failed to affect nerve and NH2OH induced relaxations. These findings indicate that among the products that can be formed during NOS catalyzed L-arginine N(G)-oxidation, only NH2OH caused relaxations that exhibited a pharmacological profile similar to those induced by the nitrergic neurotransmitter. Furthermore, if NH2OH is the actual neurotransmitter it appears to be acting either directly or by a catalase independent release of NO.


Subject(s)
Duodenum/drug effects , Glutathione/analogs & derivatives , Glutathione/pharmacology , Hydroxylamine/pharmacology , Muscle, Smooth/drug effects , Nitric Oxide/physiology , Nitroso Compounds/pharmacology , Animals , Autonomic Nervous System/drug effects , Duodenum/innervation , Free Radical Scavengers/pharmacology , In Vitro Techniques , Male , Muscle, Smooth/innervation , Rats , Rats, Wistar , S-Nitrosoglutathione , Superoxide Dismutase/antagonists & inhibitors , Superoxide Dismutase/metabolism , Superoxides/metabolism
11.
Braz J Med Biol Res ; 32(11): 1317-27, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10559832

ABSTRACT

During the past two decades, nitric oxide signaling has been one of the most rapidly growing areas in biology. This simple free radical gas can regulate an ever growing list of biological processes. In most instances nitric oxide mediates its biological effects by activating guanylyl cyclase and increasing cyclic GMP synthesis. However, the identification of effects of nitric oxide that are independent of cyclic GMP is also growing at a rapid rate. The effects of nitric oxide can mediate important physiological regulatory events in cell regulation, cell-cell communication and signaling. Nitric oxide can function as an intracellular messenger, neurotransmitter and hormone. However, as with any messenger molecule, there can be too much or too little of the substance and pathological events ensue. Methods to regulate either nitric oxide formation, metabolism or function have been used therapeutically for more than a century as with nitroglycerin therapy. Current and future research should permit the development of an expanded therapeutic armamentarium for the physician to manage effectively a number of important disorders. These expectations have undoubtedly fueled the vast research interests in this simple molecule.


Subject(s)
Cyclic GMP/biosynthesis , Enzyme Activation , Free Radical Scavengers , Guanylate Cyclase/biosynthesis , Hydroxylamine , Animals , Azides/metabolism , Azides/pharmacology , Enzyme Activation/drug effects , Enzyme Activators/pharmacology , Guanylate Cyclase/drug effects , Humans , Hydroxylamine/pharmacology , Isoenzymes/metabolism , Nitric Oxide/physiology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Vasodilator Agents/pharmacology
12.
Biosci Rep ; 19(6): 525-33, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10841269

ABSTRACT

Ca2+ and inorganic phosphate-induced mitochondrial swelling and membrane protein thiol oxidation, which are associated with mitochondrial permeability transition, are inhibited by progressively decreasing the incubation medium pH between 7.2 and 6.0. Nevertheless, the detection of mitochondrial H2O2 production under these conditions is increased. Permeability transition induced by phenylarsine oxide, which promotes membrane protein thiol cross-linkage in a process independent of Ca2+ or reactive oxygen species, is also strongly inhibited in acidic incubation media. In addition, we observed that the decreased protein thiol reactivity with phenylarsine oxide or phenylarsine oxide-induced swelling at pH 6.0 is reversed by diethyl pyrocarbonate, in a hydroxylamine-sensitive manner. These results provide evidence that the inhibition of mitrochondrial permeability transition observed at lower incubation medium pH is mediated by a decrease in membrane protein thiol reactivity, related to the protonation of protein histidyl residues.


Subject(s)
Intracellular Membranes/metabolism , Membrane Proteins/metabolism , Mitochondria, Liver/metabolism , Sulfhydryl Compounds/metabolism , Animals , Antimycin A/pharmacology , Arsenicals/pharmacology , Calcimycin/pharmacology , Calcium/metabolism , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/pharmacology , Diethyl Pyrocarbonate/pharmacology , Hydrogen-Ion Concentration , Hydroxylamine/pharmacology , Intracellular Membranes/drug effects , Ionophores/pharmacology , Mitochondria, Liver/drug effects , Oxidation-Reduction , Permeability , Rats , Rats, Wistar , Serum Albumin, Bovine/chemistry , Serum Albumin, Bovine/drug effects , Serum Albumin, Bovine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL