Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Free Radic Biol Med ; 29(11): 1085-95, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11121715

RESUMO

3-Nitrotyrosine (3NT) is used as a biomarker of nitrative pathology caused by peroxynitrite (PN), myeloperoxidase (MPO)-, and/or eosinophil peroxidase (EPO)-dependent nitrite oxidation. 3NT measurements in biological materials are usually based on either antibody staining, HPLC detection, or GC detection methodologies. In this report, a procedure is described for the measurement of 3NT and tyrosine (TYR) by LC-MS/MS that is simple, direct, and sensitive. Though highly specialized in its use as an assay, LC-MS/MS technology is available in many research centers in academia and industry. The critical assay for 3NT was linear below 100 ng/ml and the limit of detection was below 100 pg/ml. Regarding protein digested samples, we found that MRM was most selective with 133.1 m/z as the daughter ion. In comparison, LC-ECD was 100 times less sensitive. Basal levels of 3NT in extracted digests of rat brain homogenate were easily detected by LC-MS/MS, but were below detection by LC-ECD. The LC-MS/MS assay was used to detect 3NT in rat brain homogenate that was filtered through a 180 micron nylon mesh. Three fractions were collected and examined by phase contrast microscopy. The mass ratio (3NT/TYR) of 3NT in fractions of large vessel enrichment, microvessel enrichment, and vessel depletion was 0.6 ng/mg, 1.2 ng/mg, and 0.2 ng/mg, respectively. Ultimately, we found that the basal 3NT/TYR mass ratio as determined by LC-MS/MS was six times greater in microvessel-enriched brain tissue vs. tissue devoid of microvessels.


Assuntos
Cromatografia Líquida , Espectrometria de Massas , Microcirculação/química , Nitratos/química , Tirosina/análogos & derivados , Tirosina/análise , Animais , Encéfalo/irrigação sanguínea , Microscopia de Contraste de Fase , Ratos , Ratos Wistar , Sensibilidade e Especificidade
2.
Chem Res Toxicol ; 13(4): 294-300, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10775330

RESUMO

Peroxynitrite (PN), a very reactive oxidant formed by the combination of superoxide and nitric oxide, appears to play a role in producing tissue damage in a number of inflammatory conditions. Pharmacological scavenging and decomposition of PN within these areas has therapeutic value in several tissue injury models. Recently, we have been interested in nitroxide free radical-containing compounds as possible scavengers of PN decomposition products. Nitroxides can undergo redox reactions to the corresponding hydroxylamine anion or oxoammonium cation in biological systems as shown by its ability to react with superoxide, leading to the formation of hydrogen peroxide and molecular oxygen. We found that 4-hydroxy-2,2,6,6-tetramethylpiperidine-1-oxyl (Tempol) inhibits PN-mediated nitration of phenolic compounds in the presence of a large molar excess of PN, suggesting a catalytic-like mechanism. In these experiments, Tempol inhibited PN-mediated nitration over the pH range of 6.5-8.5. This inhibition was specific for nitration and had no effect on hydroxylation. After the inhibition of PN-mediated nitration, Tempol was recovered from the reaction mixtures unmodified. In addition, Tempol was effective in protecting PC-12 cells from death induced by SIN-1, a PN-generating compound. The exact mechanism of Tempol's interaction with PN is not clear; however, we propose that an intermediate in this reaction may be a nitrogen dioxide radical-Tempol complex. This complex could react with water to form either nitrite or nitrate, or with a phenol radical to produce nitrophenol or nitrosophenol products and regenerate the nitroxide.


Assuntos
Antioxidantes/farmacologia , Óxidos N-Cíclicos/farmacologia , Nitratos/antagonistas & inibidores , Fenol/metabolismo , Animais , Concentração de Íons de Hidrogênio , Hidroxilação , L-Lactato Desidrogenase/metabolismo , Molsidomina/análogos & derivados , Molsidomina/antagonistas & inibidores , Nitratos/metabolismo , Nitrocompostos/metabolismo , Compostos Nitrosos/metabolismo , Células PC12 , Ratos , Marcadores de Spin
3.
Ann N Y Acad Sci ; 890: 462-8, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10668450

RESUMO

Recent evidence has suggested that the superoxide and nitric oxide-derived reactive oxygen species peroxynitrite (ONOO-) may play a significant role in the acute pathophysiology of brain injury. One pharmacological mechanism by which ONOO(-)-mediated damage might be interrupted is by the administration of scavenging compounds such as the thiol-containing compound penicillamine. In the present study, we examined the ability of either penicillamine (Pen) or the more brain penetrable penicillamine methyl ester (PenME) (0.01, 0.1, 1.0 or 10.0 mg/kg i.v. 5 min post-injury) to improve the early (1 hr) neurological recovery (grip score) of male CF-1 mice after a severe (900 g-cm; 50 g x 18 cm) injury. Pen produced a dose-related improvement in grip score. At 1.0 mg/kg, a +112% improvement was observed compared to vehicle-treated mice, and at 10.0 mg/kg, the increase was +168% (both, p < 0.05). PenME more potently improved the 1-hr grip score, but the magnitude of the optimal effect (+96% at 0.1 mg/kg; p < 0.02) was no greater than that observed with Pen, which largely remains in the cerebral microvasculature. These results are consistent with a role of ONOO- in acute head injury, but suggest that microvascular scavenging may be of primary therapeutic importance during the early post-traumatic period.


Assuntos
Lesões Encefálicas/tratamento farmacológico , Quelantes/uso terapêutico , Nitratos/antagonistas & inibidores , Penicilamina/análogos & derivados , Penicilamina/uso terapêutico , Animais , Quelantes/farmacologia , Avaliação Pré-Clínica de Medicamentos , Masculino , Camundongos , Nitratos/fisiologia , Penicilamina/farmacologia
4.
Ann Neurol ; 44(5): 763-70, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9818932

RESUMO

Mutations of the SOD1 gene encoding copper/zinc superoxide dismutase (CuZnSOD) cause an inherited form of amyotrophic lateral sclerosis. When expressed in transgenic mice, the same SOD1 mutations cause progressive loss of spinal motor neurons with consequent paralysis and death. In vitro biochemical studies indicate that SOD1 mutations enhance free radical generation by the mutant enzyme. We investigated those findings in vivo by using a novel, brain-permeable spin trap, azulenyl nitrone. Reaction of azulenyl nitrone with a free radical forms a nitroxide adduct that then fragments to yield the corresponding azulenyl aldehyde. Transgenic mice expressing mutant SOD1-G93A show enhanced free radical content in spinal cord but not brain. This correlates with tissue-specific differences in the level of transgene expression. In spinal cord, the increase in free radical content is in direct proportion to the age-dependent increase in mutant human CuZnSOD expression. This increase precedes motor neuron degeneration. The higher level of human CuZnSOD expression seen in spinal cord compared with brain, and consequent difference in free radical generation, provides a basis for understanding the selective vulnerability of the spinal cord in this disease model.


Assuntos
Esclerose Lateral Amiotrófica/genética , Encéfalo/enzimologia , Mutação Puntual , Medula Espinal/enzimologia , Superóxido Dismutase/genética , Envelhecimento/metabolismo , Esclerose Lateral Amiotrófica/enzimologia , Animais , Azulenos , Encéfalo/crescimento & desenvolvimento , Modelos Animais de Doenças , Radicais Livres/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Humanos , Camundongos , Camundongos Transgênicos , Óxidos de Nitrogênio , Sesquiterpenos , Marcadores de Spin , Medula Espinal/crescimento & desenvolvimento , Superóxido Dismutase/metabolismo , Superóxidos/metabolismo
5.
J Inherit Metab Dis ; 21(5): 587-97, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9728338

RESUMO

CuZn superoxide dismutase (CuZn SOD) is one of several antioxidant enzymes that defend the cell against damage by oxygen free radicals. Mutations of the SOD1 gene encoding CuZn SOD are found in patients with familial amyotrophic lateral sclerosis (FALS), a progressive and fatal paralytic disease that is caused by the death of motor neurons in cortex, brainstem and spinal cord. The disease can be reproduced in transgenic mice by expression of mutant human CuZn SOD. Recent studies both in vitro and in vivo suggest that the effect of mutation is to enhance the generation of oxygen radicals by the mutant enzyme. Thus, mutation converts a protective, antioxidant enzyme into a destructive, prooxidant form that catalyses free radical damage to which motor neurons are selectively vulnerable. Recent studies of neuroprotective agents in the FALS model show that inhibition of oxidative mechanisms (copper chelation therapy, dietary antioxidants, and coexpression of bcl-2) delays disease onset but does not extend disease duration. In contrast, inhibition of glutamatergic or apoptotic mechanisms (riluzole, gabapentin, and coexpression of glutamatergic or apoptotic mechanisms (riluzole, gabapentin, and coexpression of an inhibitor of caspase-1) has no effect on disease onset but extends survival by increasing the duration of symptomatic disease. Thus, neuroprotective agents differentially target the processes underlying disease initiation and propagation.


Assuntos
Doença dos Neurônios Motores/enzimologia , Doença dos Neurônios Motores/genética , Superóxido Dismutase/genética , Esclerose Lateral Amiotrófica/enzimologia , Esclerose Lateral Amiotrófica/genética , Animais , Humanos , Camundongos , Camundongos Transgênicos
6.
J Pharmacol Exp Ther ; 286(1): 497-508, 1998 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9655895

RESUMO

Our studies examined the role of dopamine D4 receptors in the induction of behavioral sensitization to amphetamine (Amp) and accompanying neurochemical and molecular adaptive responses using a highly selective D4 antagonist, PNU-101387G. Behavioral sensitization to an acute challenge of Amp (2 mg/kg, s.c.) was observed in rats pretreated with five daily doses of Amp (2 mg/kg/d, s.c.) followed by 7-day withdrawal. Interestingly, coadministration of PNU-101387G with Amp during pretreatment completely blocked the sensitized response to an acute Amp challenge. The behavioral sensitization and its blockade by the D4 antagonist were observed in the absence of significant differences in cerebellar Amp levels among the various pretreatment groups. Accompanying behavioral sensitization were two postsynaptic neuroadaptive responses: reduction in the ability of Amp to induce c-fos gene expression in the infralimbic/ventral prelimbic cortex and NT/N mRNA in the accumbal shell. However, concurrent blockade of D4 receptors during Amp pretreatment prevented the refractoriness in c-fos and NT/N responsiveness to acute Amp. We observed also a presynaptic neuroplastic response associated with the behavioral sensitization: a significant augmentation in the ability of Amp to increase extracellular dopamine concentrations in the nucleus accumbens shell. As with the behavioral sensitization and associated postsynaptic adaptive responses, concurrent administration of PNU-101387G with Amp during pretreatment blocked the augmentation in Amp-induced dopamine release. Taken together, these data demonstrate that dopamine D4 receptors play an important role in the induction of behavioral sensitization to Amp and accompanying adaptations in pre- and postsynaptic neural systems associated with the mesolimbocortical dopamine projections.


Assuntos
Anfetamina/farmacologia , Comportamento Animal/efeitos dos fármacos , Receptores de Dopamina D2/fisiologia , Adaptação Fisiológica , Anfetamina/farmacocinética , Animais , Cerebelo/metabolismo , Dopamina/análise , Genes fos , Masculino , Atividade Motora/efeitos dos fármacos , Neurotensina/análise , Núcleo Accumbens/química , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Receptores de Dopamina D4
7.
Free Radic Biol Med ; 24(5): 738-44, 1998 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-9586804

RESUMO

We present analytical and neuroprotective data on a unique spin trapping agent derived from a novel chemical class known as an azulenyl nitrone (AZN). Based on Colorimetric properties, AZN was used to assess the formation of free radicals in a bilateral carotid occlusion (BCO) model in gerbils by monitoring the conversion of the nitrone to the aldehyde in affected tissue. In addition, AZN was tested as a neuroprotectant in this model regarding the preservation of CA1 pyramidal cells of the hippocampus following transient ischemia/reperfusion. AZN was electrochemically oxidized to give the aldehyde using an HPLC system with on line electrochemical oxidation. The oxidation potential associated with a 50% loss of AZN occurred at about 600 mV (half-wave potential versus palladium electrode). The major product detected as AZN oxidation occurred in an aqueous methanolic medium was the corresponding azulenyl aldehyde. Oxidation of AZN was inversely related to the formation of the aldehyde. Based on this test, we considered the in vivo conversion of AZN to aldehyde to be a measurement of oxidative stress in tissue. Results show that 0.3% of hippocampal AZN was converted to aldehyde in animals treated as shams. However, in gerbils subjected to a 7-min ischemic insult plus 7-min reperfusion, the conversion rate was about 3 times higher at 1.0%. In this model, surviving CA1 hippocampal neurons were counted from gerbils that were subjected to 7 mins of BCO followed by 5 days of reperfusion. In sham animals, about 89 cells were counted in a selected field of CA1 neurons. With injury, only 27 cells on average survived (70% loss) and were counted from this selected field. Under similar conditions and AZN treatment, 57 cells survived (36% loss). We conclude, therefore, that the demonstrated neuroprotection occurs because AZN neutralizes radicals which contribute to neuronal damage following ischemia/reperfusion.


Assuntos
Arteriopatias Oclusivas/tratamento farmacológico , Doenças das Artérias Carótidas/tratamento farmacológico , Ataque Isquêmico Transitório/prevenção & controle , Fármacos Neuroprotetores/uso terapêutico , Óxidos de Nitrogênio/uso terapêutico , Traumatismo por Reperfusão/prevenção & controle , Animais , Arteriopatias Oclusivas/metabolismo , Doenças das Artérias Carótidas/metabolismo , Contagem de Células/efeitos dos fármacos , Morte Celular , Colorimetria , Eletroquímica , Radicais Livres , Gerbillinae , Ataque Isquêmico Transitório/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/patologia , Fármacos Neuroprotetores/análise , Óxidos de Nitrogênio/análise , Traumatismo por Reperfusão/metabolismo , Detecção de Spin
8.
Amino Acids ; 14(1-3): 189-96, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9871460

RESUMO

Dopa and related catecholamines and their degradation products have been demonstrated to have neurotoxic potential in a number of cellular and in vivo experiments. Several mechanisms have been hypothesized to be involved including generation of prooxidant products that subsequently oxidize membrane lipids and exposed macromolecules. We have utilized a neuronal culture of cerebellar granule cells to study the toxicity of Dopa and the ability of various neuroprotective and antiparkinsonian compounds to offer protection therefrom. This model is apparently based on the ability of Dopa to non-enzymatically induce an oxidative injury to the neuronal cultures. Evidence for this arises from the equal neurotoxic potency of L- and D-Dopa in these cells and the ability of catalase, superoxide dismutase and glutathione to protect the neurons from this toxicity. Further, we found that the neuroprotective antioxidant, PNU-101033 is more effective and potent than vitamin E and deprenyl in this regard. Similarly the D2/D3 agonist, pramipexole is also capable of blocking Dopa toxicity in this model and this effect is independent of dopamine receptor affinity as both enantiomers are equally potent in this assay but disparate in receptor affinity. Also the protection by pramipexole is accompanied by the preservation of reduced glutathione. Thus, this activity seems to be a function of the oxidation potential of pramipexole and it's consequent antioxidant property. Potent antioxidants are effective blockers of Dopa toxicity. If the mechanisms involved in this toxicity have relevance to the progression of Parkinson's pathology in Dopa treated (or untreated) patients, these compounds have the potential to alter the course of the illness.


Assuntos
Cerebelo/efeitos dos fármacos , Grânulos Citoplasmáticos/efeitos dos fármacos , Levodopa/efeitos adversos , Fármacos Neuroprotetores/farmacologia , Animais , Benzotiazóis , Células Cultivadas , Cerebelo/metabolismo , Grânulos Citoplasmáticos/metabolismo , Glutationa/metabolismo , Glutationa/farmacologia , Levodopa/antagonistas & inibidores , Pramipexol , Pirimidinas/farmacologia , Pirróis/farmacologia , Ratos , Ratos Sprague-Dawley , Selegilina/farmacologia , Tiazóis/farmacologia , Vitamina E/farmacologia
9.
Age (Omaha) ; 21(2): 85-9, 1998 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23604357

RESUMO

Cu,Zn superoxide dismutase (Cu,Zn SOD) is one of several anti-oxidant enzymes which defend the cell against damage by oxygen free radicals. Mutations of the SOD1 gene encoding Cu,Zn SOD are found familial amyotrophic lateral sclerosis, a progressive and fatal paralytic disease which is caused by the death of motor neurons in cortex, brainstem and spinal cord. The disease can be reproduced in transgenic mice by expression of mutant human Cu,Zn SOD. Recent studies both in vitro and in vivo suggest that the effect of mutation is to enhance the generation of oxygen radicals by the mutant enzyme. Thus, mutation converts a protective, antioxidant enzyme into a destructive pro-oxidant form which catalyzes free radical damage to which motor neurons are uniquely vulnerable.

10.
J Pharmacol Exp Ther ; 281(2): 895-904, 1997 May.
Artigo em Inglês | MEDLINE | ID: mdl-9152399

RESUMO

A novel group of antioxidant compounds, the pyrrolopyrimidines, has been discovered recently. Many of these possess significantly improved oral bioavailability (56-70% in rats), increased efficacy and potency in protecting cultured neurons against iron-induced lipid peroxidative injury and as much as a 5-fold increase in brain uptake compared with the 21-aminosteroid antioxidant compound, tirilazad mesylate (U-74006F), described earlier. They appear to quench lipid peroxidation reactions by electron-donating and/or radical-trapping mechanisms. Several compounds in the series, such as U-101033E and U-104067F, demonstrate greater ability than tirilazad to protect the hippocampal CA1 region in the gerbil transient (5-min) forebrain ischemia model. Delaying treatment until 4 hr after the ischemic insult still results in significant CA1 neuronal protection. U-101033E is still effective in salvaging a portion of the CA1 neuronal population when the ischemic duration is extended to 10 min. In addition, U-101033E has been found to be protective in the context of focal cerebral ischemia, reducing infarct size in the mouse permanent middle cerebral artery occlusion model, in contrast to tirilazad which is minimally effective. These results suggest that antioxidant compounds with improved brain parenchymal penetration are better able to limit certain types of ischemic brain damage than those which are localized in the cerebral microvasculature. However, the activity of U-101033E in improving early post-traumatic recovery in mice subjected to severe concussive head injury is similar to that of tirilazad. Last, the oral bioavailability of many pyrrolopyrimidines suggests that they may be useful for certain chronic neurodegenerative disorders in which lipid peroxidation plays a role.


Assuntos
Antioxidantes/farmacologia , Lesões Encefálicas/prevenção & controle , Isquemia Encefálica/prevenção & controle , Fármacos Neuroprotetores/farmacologia , Pirimidinas/farmacologia , Animais , Antioxidantes/farmacocinética , Disponibilidade Biológica , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Lesões Encefálicas/metabolismo , Isquemia Encefálica/metabolismo , Feminino , Sequestradores de Radicais Livres , Gerbillinae , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Camundongos , Fármacos Neuroprotetores/farmacocinética , Pirimidinas/farmacocinética , Ratos , Ratos Sprague-Dawley
11.
Free Radic Biol Med ; 22(1-2): 223-8, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-8958148

RESUMO

We developed a cerebellar granule cell model of peroxynitrite toxicity and showed that certain sulfhydryl-containing compounds (e.g., penicillamine) present as concurrent treatments could inhibit this toxicity. In the present study, 21-aminosteroid and pyrrolopyrimidine lazaroids were tested for cytoprotection in this peroxynitrite toxicity model. In addition, we tested for added protection using a peroxynitrite scavenger concurrent treatment combined with a lazaroid post-treatment. The toxicity assay utilized cells that were previously exposed to 100 microM L-buthionine (S,R)-sulfoximine (BSO), an inhibitor of gamma-glutamyl-cysteine synthetase, for 24 h. This sublethal concentration of BSO shifted the peroxynitrite (1-1000 microM) toxicity curve to the left by more than one-half of a log unit. The half-maximal toxicity concentration (TC50) of peroxynitrite in cells treated with BSO was 50 microM. The 21-aminosteroids, U-74006F and U-74500A, and the pyrrolopyrimidines, U-91736B and U-101033E, were tested as post-treatments. U-74006F and U-74500A had EC50 values of approximately 100 microM (concentrations which blocked 50% of the toxicity). U-91736B and U-101033E had EC50 values of 1 microM and showed 100% protection at 3-10 microM. Treatment with either 100 microM U-74006F or 1 microM U-101033E resulted in a right-hand shift (protection) in the peroxynitrite toxicity curve. Further, combination treatment of lazaroids with 1 mM penicillamine resulted in additive protection compared to either treatment alone.


Assuntos
Antioxidantes/farmacologia , Sequestradores de Radicais Livres/farmacologia , Fármacos Neuroprotetores/farmacologia , Pregnatrienos/farmacologia , Pirimidinas/farmacologia , Pirrolidinonas/farmacologia , Animais , Células Cultivadas , Cerebelo/citologia , Cerebelo/efeitos dos fármacos , Modelos Biológicos , Nitratos/toxicidade , Pirróis/farmacologia , Pirrolidinas/farmacologia , Ratos , Ratos Sprague-Dawley
12.
Brain Res ; 742(1-2): 80-8, 1996 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-9117424

RESUMO

We have examined the neuroprotective efficacy of the selective dopamine (DA) D2/D3 receptor agonist pramipexole in two models of nigrostriatal (NS) degeneration. The first involves the delayed (28-day) postischemic retrograde NS degeneration that takes place in gerbils following a 10-min episode of bilateral carotid arterial occlusion-induced forebrain ischemia. In vehicle (40% hydroxypropyl cyclodextrin)-treated male gerbils, there was a 40-45% loss of NS cell bodies in the pars compacta and pars reticulata (TH immunohistochemistry and Cresyl violet histochemistry) by 28 days after ischemia/reperfusion. Daily postischemic oral dosing (1 mg/kg p.o., b.i.d., beginning at 1 h after insult) decreased the 28-day postischemic loss of NS DA neurons by 36% (P < 0.01 vs. vehicle-treated). The effect was specific for dopamine neurons since no significant salvage of hippocampal CA1 neurons was observed. In a second model, pramipexole's effects were examined on methamphetamine-induced (10 mg/kg, i.p. X 4, each 2 h apart) NS degeneration in male Swiss-Webster mice. In vehicle-treated mice, there was a 40% loss of NS neurons by day 5. In contrast, pramipexole dosing (1 mg/kg, p.o., 1 h after the last methamphetamine dose, plus daily) attenuated the NS degeneration from 40% to only 8% (P < 0.00001 vs. vehicle). We postulated that pramipexole acts in both of these models to reduce the elevated DA turnover and the associated elevation in hydroxyl radical production secondary to increased MAO activity that could be responsible for oxidative damage to the NS neurons. Indeed, in the gerbil ischemia model, we documented by HPLC-ECD a 135% postreperfusion increase in DA turnover (DOPAC + HVA/DA) at 5 min after reperfusion. Pramipexole at the 1 mg/kg, p.o., dose level was able to significantly reduce the increased DA turnover, but by only 16%. Thus, it is conceivable that other mechanisms may also contribute to pramipexole's dopaminergic neuroprotection. Based on a preliminary examination of pramipexole's oxidation potential, it appears that the compound may possess significant intrinsic antioxidant properties that might contribute to its neuroprotective effects.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Corpo Estriado/efeitos dos fármacos , Agonistas de Dopamina/farmacologia , Degeneração Neural/efeitos dos fármacos , Substância Negra/efeitos dos fármacos , Tiazóis/farmacologia , Animais , Benzotiazóis , Relação Dose-Resposta a Droga , Gerbillinae , Masculino , Metanfetamina/farmacologia , Pramipexol
13.
Brain Res ; 739(1-2): 210-4, 1996 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-8955941

RESUMO

Although alpha-methyl-L-p-tyrosine (alpha-MPT), an inhibitor of catecholamine synthesis, has been used to study catecholamine turnover in diabetic animals, effects of diabetes on metabolism of the drug have not been investigated. In this study, administration of a standard dose of alpha-MPT (250 mg/kg initially and 125 mg/kg at 2 h intervals) resulted in lower plasma and tissue levels of alpha-MPT and its metabolites in streptozocin-diabetic rats than in controls. Two to six hours after the initial dose of alpha-MPT, concentrations of alpha-MPT were 2-8-fold lower in the hypothalamus, medulla/pons, and plasma of diabetic animals than in controls. Brain and plasma levels of the alpha-MPT metabolite, alpha-methyl DOPA (alpha-MD) were 2-10-fold lower in tissues of diabetic animals. Levels of the alpha-MPT metabolite alpha-methyl norepinephrine (alpha-MNE), measured only in the hypothalamus, were 4-fold lower in diabetic rats than in controls. There were no differences in the ratio of free/conjugated alpha-MPT in plasma. Treatment of diabetic rats with insulin restored alpha-MPT and alpha-MD to control levels. These findings indicate that i.p. administration of alpha-MPT does not result in equivalent levels of the drug in diabetic and control rats and suggest caution in the use of alpha-MPT to compare catecholamine turnover in diabetic and healthy animals.


Assuntos
Catecolaminas/biossíntese , Diabetes Mellitus Experimental/metabolismo , Inibidores Enzimáticos/metabolismo , Metiltirosinas/metabolismo , Tirosina 3-Mono-Oxigenase/antagonistas & inibidores , Animais , Encéfalo/metabolismo , Diabetes Mellitus Experimental/sangue , Inibidores Enzimáticos/sangue , Masculino , Metiltirosinas/sangue , Ratos , Ratos Sprague-Dawley , Valores de Referência , alfa-Metiltirosina
14.
Res Commun Mol Pathol Pharmacol ; 91(3): 357-71, 1996 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8829775

RESUMO

Following CNS trauma or ischemia, peroxynitrite may be a toxic intermediate which forms in vivo when nitric oxide condenses with superoxide. Alone, peroxynitrite appears to directly react with aromatic and sulfhydryl nucleophiles. However, at physiological pH, peroxynitrite is protonated and, in that form, will rapidly (within seconds) decompose to species with hydroxyl radical and nitrogen dioxide characteristics. These reactive species are shown to initiate lipid peroxidation, hydroxylate aromatic residues, and nitrate aromatic residues. This reactivity may contribute to differential toxicity in vivo and in vitro. Tirilazad mesylate (TZ) is a lipid-soluble antioxidant shown to inhibit iron-dependent lipid peroxidation. It is an effective therapy in a variety of CNS injury and ischemia models and is currently undergoing human clinical evaluation in stroke, head injury, and spinal injury. This study was designed to investigate the cytoprotective properties of TZ in a cerebellar granule cell model of peroxynitrite toxicity. Cytoprotective efficacy of TZ was based on viability measurements, blockade of lipid hydroperoxide generation, and blockade of nitrotyrosine formation. Cell viability was determined by [3H]-aminoisobutyric acid (3H-AIB) uptake, and lipid hydroperoxide and nitrotyrosine content were determined by HPLC assays. Tirilazad mesylate was found to have similar cytoprotective effects (approximately 50% protection at 100 microM) when applied before or after exposure of cells to peroxynitrite. In contrast, post-treatment with superoxide dismutase (50 units/ml) or allopurinol (100 microM) failed to produce any cytoprotection. Furthermore, we discovered that TZ inhibited the peroxynitrite-induced increase of phosphatidylethanolamine hydroperoxide (PEOOH), but did not affect the peroxynitrite-induced formation of nitrotyrosine formation. This suggests that the ability of TZ to afford cytoprotection in this peroxynitrite toxicity model is due to the inhibition of membrane-localized lipid peroxidation, and not to the inhibition of nitration of tyrosine residues.


Assuntos
Sequestradores de Radicais Livres/farmacologia , Nitratos/antagonistas & inibidores , Nitratos/toxicidade , Pregnatrienos/farmacologia , Ácidos Aminoisobutíricos/metabolismo , Animais , Biomarcadores , Células Cultivadas , Cerebelo/citologia , Cerebelo/efeitos dos fármacos , Cerebelo/ultraestrutura , Emulsões , Peróxidos Lipídicos/metabolismo , Ratos , Ratos Sprague-Dawley , Tirosina/análogos & derivados , Tirosina/metabolismo
15.
Res Commun Mol Pathol Pharmacol ; 87(3): 359-66, 1995 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-7620829

RESUMO

Peroxynitrite may be a physiologically relevant endogenous neurotoxin that forms following CNS trauma when excessive levels of NO and .O2 accumulate. Recently, peroxynitrite was found to inactivate the polyclonal antibody to cAMP. A feasibility study was performed to evaluate the use of capillary electrophoresis as an effective tool regarding the structural transformation of antibody following exposure to peroxynitrite with or without co-incubation with a peroxynitrite scavenger. A polyclonal antibody to cAMP and a monoclonal antibody to plasminogen activation inhibitor-1 were exposed to peroxynitrite with or without penicillamine coincubation. Samples were analyzed by an Applied Biosystems analytical capillary electrophoresis system, model 270A. Initial examination of the peroxynitrite scavenger penicillamine and its reaction with peroxynitrite showed a penicillamine migration peak at about 9.1 min and a presumed s-nitro adduct of penicillamine that migrated at 10.9 min. Exposure of either antibody to peroxynitrite resulted in structural transformation of protein based on changes in migration patterns. In addition, co-incubation with penicillamine prevented this transformation and preserved the pre-peroxynitrite migration patterns of antibodies. In cases of antibody reaction, s-nitro adduct formation could be simultaneously monitored. We found capillary electrophoresis to be ideally suited to this type of analysis. With capillary electrophoresis, we were able to simultaneously monitor the effects of peroxynitrite on large proteins and a small scavenger molecule. As a result, a complete record of the reaction was obtained within a single 15-min analysis period.


Assuntos
Anticorpos/efeitos dos fármacos , Nitratos/farmacologia , Anticorpos/análise , Anticorpos/química , AMP Cíclico/imunologia , Interações Medicamentosas , Eletroforese/métodos , Estudos de Viabilidade , Penicilamina/farmacologia , Inibidor 1 de Ativador de Plasminogênio/imunologia , Fatores de Tempo
16.
Res Commun Chem Pathol Pharmacol ; 83(3): 243-54, 1994 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8008974

RESUMO

Nitric oxide (NO) is made by NO synthase during the conversion of arginine to citrulline. Researchers have found that they can block the actions of excitotoxins by inhibiting NO synthase. Released from excitable cells during trauma, NO may react with superoxide to form peroxynitrite. Once formed, peroxynitrite and its products can then react with proteins, lipids and nucleic acids resulting in cell injury and death. The present study was undertaken to investigate analogs of cysteine as scavengers of peroxynitrite. Peroxynitrite scavengers were assayed by Attoflo, an automated radioimmunoassay. Briefly, peroxynitrite, in a dose-dependent manner (0.1 to 10 mM), inhibited the binding of I125 cAMP to a polyclonal antibody used in the assay of cAMP. Drugs were tested for blockade of the inhibition (90%) caused by peroxynitrite at 10 mM. Cysteine blocked the inhibition of ligand/antibody binding in a dose-dependent manner (EC50 = 3 mM). Cysteine, cysteine esters, penicillamine, penicillamine esters and cysteamine were the most effective peroxynitrite scavengers. Analogs of cysteine may thereby protect cells from nitric oxide toxicity.


Assuntos
Cisteína/farmacologia , Sequestradores de Radicais Livres , Nitratos/metabolismo , Animais , Reações Antígeno-Anticorpo/efeitos dos fármacos , Encéfalo/metabolismo , AMP Cíclico/metabolismo , Cisteína/análogos & derivados , Relação Dose-Resposta a Droga , Imunoglobulina G/metabolismo , Nitratos/farmacologia , Óxido Nítrico/metabolismo , Radioimunoensaio , Relação Estrutura-Atividade
17.
Mol Chem Neuropathol ; 20(2): 147-62, 1993 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8297419

RESUMO

Oxygen free radicals have been implicated as a causal factor in posttraumatic neuronal cell loss following cerebral ischemia and head injury. The conversion of salicylate to dihydroxybenzoic acid (DHBA) in vivo was employed to study the formation of hydroxyl radical (.OH) following central nervous system (CNS) injury. Bilateral carotid occlusion (BCO) in gerbils and concussive head trauma in mice were selected as models of brain injury. The lipid peroxidation inhibitor, tirilazad mesylate (U-74006F), was tested for its ability to attenuate hydroxyl radical formation in these models. In addition, U-74006F was studied as a scavenger of hydroxyl radical in an in vitro assay based on the Fenton reaction. For in vivo experimentation, hydroxyl radical formation was expressed as the ratio of DHBA to salicylate (DHBA/SAL) measured in brain. In the BCO model, hydroxyl radical formation increased in whole brain with 10 min of occlusion followed by 1 min of reperfusion. DHBA/SAL was also found to increase in the mouse head injury model at 1 h postinjury. In both models, U-74006F (1 or 10 mg/kg) blocked the increase in DHBA/SAL following injury. In vitro, reaction of U-74006F with hydroxyl radical gave a product with a mol wt that was 16 greater than U-74006F, indicative of hydroxyl radical scavenging. We speculate that U-74006F may function by blocking oxyradical-dependent cell damage, and thereby maintaining free iron (which catalyzes hydroxyl radical formation) concentrations at normal levels.


Assuntos
Lesões Encefálicas/metabolismo , Isquemia Encefálica/metabolismo , Gentisatos , Radical Hidroxila/metabolismo , Peróxidos Lipídicos/antagonistas & inibidores , Pregnatrienos/farmacologia , Salicilatos/metabolismo , Animais , Sequestradores de Radicais Livres , Hidroxibenzoatos/metabolismo , Hidroxilação , Masculino , Camundongos , Ácido Salicílico
18.
Brain Res ; 606(1): 50-5, 1993 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-8384924

RESUMO

U-54494A, 3,4-dichloro-N-methyl-N-[2-(1-pyrrolidinyl)cyclohexyl]benzamide, has been shown to be a potent and long-acting anticonvulsant without analgesic or sedative effects on intact animals. The persistence of anticonvulsant activity after a decline in its concentration in the brain implies the conversion of the parent drug into active metabolites. In this study, two major metabolites of U-54494A, U-83892E [cis-N-(2-aminocyclohexyl)-3,4-dichlorobenzamide] and U-83894A [cis-N-(2-methylaminocyclohexyl)-3,4-dichlorobenzamide], were identified. The synthetic metabolites displayed anticonvulsant activity against electric shock in experimental animals and blocked voltage-gated sodium channel in N1E-115 neuroblastoma cells in voltage- and use-dependent manner by interacting with the inactivated channels as well as with the channels in the resting state (like the parent compound). These observations may provide one explanation for the long duration of the anticonvulsant activity of the parent compound U-54494A and further underscore the importance of voltage-dependent sodium channels in neuronal excitability, especially during seizures.


Assuntos
Anticonvulsivantes/farmacologia , Benzamidas/farmacologia , Cicloexilaminas/farmacologia , Pirrolidinas/metabolismo , Canais de Sódio/efeitos dos fármacos , Animais , Eletrofisiologia , Homeostase , Masculino , Ratos , Ratos Sprague-Dawley , Canais de Sódio/fisiologia
19.
Res Commun Chem Pathol Pharmacol ; 79(2): 195-208, 1993 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-8451541

RESUMO

We examined the effects of 6-hydroxydopamine (6-OHDA) treatment on the human neuroblastoma cell line SK-N-SH-SY5Y (SY5Y) and the rat pheochromocytoma cell line, PC12. Structural and metabolic integrity was tested by measuring the ability of cells to transport the non-metabolizable amino acid analogue [3H]-alpha-aminoisobutyric acid (AIB). We determined that treatment with 6-OHDA at concentrations of 49 microM and 62 microM inhibited 50% of the AIB uptake in SY5Y and PC12 cells, respectively. Inhibition of AIB uptake was prevented by the addition of catalase, but was not influenced by the addition of 1 mM dopamine. This indicated that cell damage resulted from the generation of H2O2 and was independent of the catecholamine uptake system. Effects directly on the catecholamine uptake system were observed by measuring the uptake of 3H-dopamine. In contrast to the effects on amino acid uptake, dopamine uptake was significantly inhibited by 6-OHDA treatment, and this inhibition was not prevented by the addition of catalase. The results indicate a Ki of 430 microM for inhibition of dopamine uptake by 6-OHDA treatment of PC12 cells. The results are consistent with a competitive irreversible inhibition of the dopamine uptake sites by 6-OHDA or one of its metabolites. Thus, the lack of a catecholamine uptake-dependent cellular toxicity appears to result from the direct inactivation of catecholamine uptake sites. Similarly, the inhibition of dopamine uptake in vivo by 6-OHDA may be explained, at least in part, by direct inactivation of dopamine uptake sites rather than exclusively by intracellular transport and action of 6-OHDA.


Assuntos
Antagonistas de Dopamina , Dopamina/metabolismo , Oxidopamina/farmacologia , Neoplasias das Glândulas Suprarrenais/metabolismo , Ácidos Aminoisobutíricos/antagonistas & inibidores , Ácidos Aminoisobutíricos/metabolismo , Animais , Ligação Competitiva , Transporte Biológico , Catalase/farmacologia , Humanos , Neuroblastoma/metabolismo , Feocromocitoma/metabolismo , Ratos , Células Tumorais Cultivadas
20.
J Neurosci Res ; 34(1): 107-12, 1993 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8380874

RESUMO

The salicylate trapping method was used to investigate the changes in hydroxyl radical (.OH) levels in the selectively vulnerable hippocampus compared to the cerebral cortex of gerbils subjected to a 10 min period of near complete forebrain ischemia. Salicylate-derived 2,5-dihydroxybenzoic acid (2,5-DHBA) was measured in sham-operated animals and at 1, 5, and 15 min of reperfusion. A basal level of 2,5-DHBA was also seen in non-ischemic gerbil brain, both in the hippocampus and cortex. The hippocampal basal level was 160% higher than in the cortex (P < .01). Treatment with the cytochrome P450 inhibitor SKF-525A (50 mg/kg s.c. 30 min before measurement) did not affect this basal level in either hippocampus or cortex, which argues against a contribution of metabolic salicylate hydroxylation as its source. In contrast, pretreatment with the arachidonic acid cyclo-oxygenase inhibitor ibuprofen (20 mg/kg s.c.) decreased (-68.8%) the level of salicylate hydroxylation in the hippocampus, but not the cortex. In animals subjected to 10 min of forebrain ischemia, a selective increase in 2,5-DHBA was observed in the hippocampus at 1 min of reperfusion which subsided by 5 min. No increase in salicylate hydroxylation was apparent in the cortex within the same time frame. The increase in .OH in the hippocampus at 1 min of reperfusion was accompanied by a significant decrease (-15.7%; P < .03) in the hippocampal levels of vitamin E. No loss of vitamin E was observed in the cortex at the same time.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Isquemia Encefálica/metabolismo , Córtex Cerebral/metabolismo , Gentisatos , Hipocampo/metabolismo , Hidróxidos/metabolismo , Peróxidos Lipídicos/metabolismo , Animais , Encéfalo/metabolismo , Inibidores das Enzimas do Citocromo P-450 , Relação Dose-Resposta a Droga , Radicais Livres , Gerbillinae , Hidroxibenzoatos/metabolismo , Radical Hidroxila , Ibuprofeno/farmacologia , Masculino , Proadifeno/metabolismo , Reperfusão , Salicilatos/farmacologia , Ácido Salicílico , Vitamina E/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...