Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Neurobiol ; 58(10): 5396-5419, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34331199

RESUMO

Neuropathic pain is more prevalent in women. However, females are under-represented in animal experiments, and the mechanisms of sex differences remain inadequately understood. We used the spared nerve injury (SNI) model in rats to characterize sex differences in pain behaviour, unbiased RNA-Seq and proteomics to study the mechanisms. Male and female rats were subjected to SNI- and sham-surgery. Mechanical and cold allodynia were assessed. Ipsilateral lumbar dorsal root ganglia (DRG) and spinal cord (SC) segments were collected for RNA-seq analysis with DESeq2 on Day 7. Cerebrospinal fluid (CSF) samples for proteomic analysis and DRGs and SCs for analysis of IB-4 and CGRP, and IBA1 and GFAP, respectively, were collected on Day 21. Females developed stronger mechanical allodynia. There were no differences between the sexes in CGRP and IB-4 in the DRG or glial cell markers in the SC. No CSF protein showed change following SNI. DRG and SC showed abundant changes in gene expression. Sexually dimorphic responses were found in genes related to T-cells (cd28, ctla4, cd274, cd4, prf1), other immunological responses (dpp4, c5a, cxcr2 and il1b), neuronal transmission (hrh3, thbs4, chrna4 and pdyn), plasticity (atf3, c1qc and reg3b), and others (bhlhe22, mcpt1l, trpv6). We observed significantly stronger mechanical allodynia in females and numerous sexually dimorphic changes in gene expression following SNI in rats. Several genes have previously been linked to NP, while some are novel. Our results suggest gene targets for further studies in the development of new, possibly sex-specific, therapies for NP.


Assuntos
Gânglios Espinais/metabolismo , Hiperalgesia/genética , Hiperalgesia/metabolismo , Neuropatia Ciática/genética , Neuropatia Ciática/metabolismo , Diferenciação Sexual , Medula Espinal/metabolismo , Animais , Peptídeo Relacionado com Gene de Calcitonina/biossíntese , Peptídeo Relacionado com Gene de Calcitonina/genética , Proteínas de Ligação ao Cálcio/biossíntese , Proteínas de Ligação ao Cálcio/genética , Feminino , Expressão Gênica , Proteína Glial Fibrilar Ácida/biossíntese , Proteína Glial Fibrilar Ácida/genética , Masculino , Proteínas dos Microfilamentos/biossíntese , Proteínas dos Microfilamentos/genética , Medição da Dor/métodos , Proteômica/métodos , Ratos , Ratos Sprague-Dawley
2.
Br J Anaesth ; 120(4): 818-826, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29576122

RESUMO

BACKGROUND: Ketamine attenuates morphine tolerance by antagonising N-methyl-d-aspartate receptors. However, a pharmacokinetic interaction between morphine and ketamine has also been suggested. The interaction between oxycodone and ketamine is unclear. We studied the effects of ketamine and norketamine on the attenuation of morphine and oxycodone tolerance focusing on both the pharmacodynamic and pharmacokinetic interactions. METHODS: Morphine 9.6 mg day-1 or oxycodone 3.6 mg day-1 was delivered to Sprague-Dawley rats by subcutaneous pumps. Once tolerance had developed, the rats received subcutaneous injections of ketamine or norketamine. Tail-flick, hot-plate, and rotarod tests were performed. Drug concentrations were measured with high-performance liquid chromatography-tandem mass spectrometry. RESULTS: Anti-nociceptive tolerance to morphine and oxycodone developed similarly by Day 6. Acute ketamine 10 mg kg-1 and norketamine 30 mg kg-1 attenuated morphine tolerance for 120 and 150 min, respectively, whereas in oxycodone-tolerant rats the effect lasted only 60 min. Both ketamine and norketamine increased the brain and serum concentrations of morphine, and inhibited its metabolism to morphine-3-glucuronide, whereas oxycodone concentrations were not changed. Morphine, but not oxycodone, pretreatment increased the brain and serum concentrations of ketamine and norketamine. Ketamine, but not norketamine, significantly impaired the motor coordination. CONCLUSIONS: Ketamine and norketamine attenuated morphine tolerance more effectively than oxycodone tolerance. Ketamine and norketamine increased morphine, but not oxycodone brain concentrations, which may partly explain this difference. Norketamine is effective in attenuating morphine tolerance with minor effects on motor coordination. These results warrant pharmacokinetic studies in patients who are co-treated with ketamine and opioids.


Assuntos
Analgésicos Opioides/farmacologia , Analgésicos/farmacologia , Tolerância a Medicamentos/fisiologia , Ketamina/farmacologia , Morfina/farmacologia , Oxicodona/farmacologia , Animais , Interações Medicamentosas , Ketamina/análogos & derivados , Masculino , Modelos Animais , Ratos , Ratos Sprague-Dawley
3.
Eur J Pain ; 20(2): 297-306, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26031840

RESUMO

BACKGROUND: Oxycodone is increasingly being used in combination with pregabalin. Pregabalin use is prevalent in opioid-dependent individuals. A high number of deaths caused by the co-use of gabapentinoids and opioids occur. It is not known whether pregabalin affects concentrations of oxycodone or morphine in the central nervous system. METHODS: Effects of pregabalin on acute oxycodone or morphine-induced antinociception, tolerance and sedation were studied using tail-flick, hot plate and rotarod tests in male Sprague-Dawley rats. Concentrations of pregabalin, opioids and their major metabolites in the brain were quantified by mass spectrometry. RESULTS: In the hot plate test, morphine (2.5 mg/kg, s.c.) caused antinociception of 28% maximum possible effect (MPE), whereas pregabalin (50 mg/kg, i.p.) produced 8-10% MPE. Co-administration of pregabalin and morphine resulted in antinociception of 63% MPE. Oxycodone (0.6 mg/kg s.c.) produced antinociception of 18% MPE, which increased to 39% MPE after co-administration with pregabalin. When pregabalin 10 mg/kg was administered before oxycodone (0.6 mg/kg, s.c.) or morphine (2.5 mg/kg), only the effect of oxycodone was potentiated in the tail-flick and the hot plate tests. Brain concentrations of the opioids, their major metabolites and pregabalin were unchanged. Pregabalin co-administration (50 mg/kg, i.p., once daily) did not prevent the development of morphine tolerance. CONCLUSIONS: Pregabalin potentiated antinociceptive and sedative effects of oxycodone and morphine in acute nociception. Co-administration of pregabalin with the opioids did not affect the brain concentrations of oxycodone or morphine. Pregabalin did not prevent morphine tolerance.


Assuntos
Analgésicos Opioides/uso terapêutico , Morfina/uso terapêutico , Nociceptividade/efeitos dos fármacos , Oxicodona/uso terapêutico , Dor/tratamento farmacológico , Pregabalina/uso terapêutico , Analgésicos Opioides/farmacologia , Animais , Interações Medicamentosas , Quimioterapia Combinada , Temperatura Alta , Masculino , Morfina/farmacologia , Oxicodona/farmacologia , Medição da Dor/efeitos dos fármacos , Pregabalina/farmacologia , Ratos , Ratos Sprague-Dawley
4.
Br J Pharmacol ; 172(11): 2799-813, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25297798

RESUMO

BACKGROUND AND PURPOSE: The effects of ketamine in attenuating morphine tolerance have been suggested to result from a pharmacodynamic interaction. We studied whether ketamine might increase brain morphine concentrations in acute coadministration, in morphine tolerance and morphine withdrawal. EXPERIMENTAL APPROACH: Morphine minipumps (6 mg·day(-1) ) induced tolerance during 5 days in Sprague-Dawley rats, after which s.c. ketamine (10 mg·kg(-1) ) was administered. Tail flick, hot plate and rotarod tests were used for behavioural testing. Serum levels and whole tissue brain and liver concentrations of morphine, morphine-3-glucuronide, ketamine and norketamine were measured using HPLC-tandem mass spectrometry. KEY RESULTS: In morphine-naïve rats, ketamine caused no antinociception whereas in morphine-tolerant rats there was significant antinociception (57% maximum possible effect in the tail flick test 90 min after administration) lasting up to 150 min. In the brain of morphine-tolerant ketamine-treated rats, the morphine, ketamine and norketamine concentrations were 2.1-, 1.4- and 3.4-fold, respectively, compared with the rats treated with morphine or ketamine only. In the liver of morphine-tolerant ketamine-treated rats, ketamine concentration was sixfold compared with morphine-naïve rats. After a 2 day morphine withdrawal period, smaller but parallel concentration changes were observed. In acute coadministration, ketamine increased the brain morphine concentration by 20%, but no increase in ketamine concentrations or increased antinociception was observed. CONCLUSIONS AND IMPLICATIONS: The ability of ketamine to induce antinociception in rats made tolerant to morphine may also be due to increased brain concentrations of morphine, ketamine and norketamine. The relevance of these findings needs to be assessed in humans.


Assuntos
Analgésicos Opioides/farmacologia , Comportamento Animal/efeitos dos fármacos , Encéfalo/metabolismo , Tolerância a Medicamentos , Ketamina/farmacologia , Morfina/farmacologia , Analgésicos/metabolismo , Analgésicos/farmacologia , Analgésicos Opioides/metabolismo , Animais , Cromatografia Líquida de Alta Pressão , Quimioterapia Combinada , Injeções Subcutâneas , Ketamina/análogos & derivados , Ketamina/metabolismo , Fígado/metabolismo , Morfina/metabolismo , Derivados da Morfina/metabolismo , Dor/tratamento farmacológico , Medição da Dor/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Espectrometria de Massas em Tandem
5.
Eur J Pain ; 18(3): 386-95, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23900882

RESUMO

BACKGROUND: Spironolactone, a commonly used mineralocorticoid receptor antagonist, has been reported to potentiate the effect of morphine in the rat. The aim of this study was to investigate the effects of spironolactone on morphine antinociception and tissue distribution. METHODS: The effects of spironolactone on acute morphine-induced antinociception, induction of morphine tolerance and established morphine tolerance were studied with tail-flick and hot plate tests in male Sprague-Dawley rats. Serum, brain, and liver morphine and its metabolite concentrations were quantified using high-pressure liquid chromatography-tandem mass spectrometry. Spironolactone was also administered with the peripherally acting, P-glycoprotein (P-gp) substrate loperamide to test whether spironolactone allows loperamide to pass the blood-brain barrier. RESULTS: Spironolactone (50 mg/kg, i.p.) had no antinociceptive effects of its own, but it enhanced the antinociceptive effect of morphine in both thermal tests. Two doses of spironolactone enhanced the maximum possible effect (MPE) from 19.5% to 100% in the hot plate test 90 min after administration of 4 mg/kg morphine. Morphine concentrations in the brain were increased fourfold at 90 min by spironolactone. Spironolactone did not inhibit the formation of morphine-3-glucuronide. Acute spironolactone restored morphine antinociception in morphine-tolerant rats but did not inhibit the development of tolerance. The peripherally restricted opioid, loperamide (10 mg/kg), had no antinociceptive effects when administered alone, but co-administration with spironolactone produced a 40% MPE in the hot plate test. CONCLUSIONS: Spironolactone has no antinociceptive effects in thermal models of pain, but it enhances the antinociceptive effects of morphine mainly by increasing morphine central nervous system concentrations, probably by inhibiting P-gp.


Assuntos
Analgésicos/uso terapêutico , Antagonistas de Receptores de Mineralocorticoides/uso terapêutico , Morfina/uso terapêutico , Dor/tratamento farmacológico , Espironolactona/uso terapêutico , Analgésicos/farmacocinética , Animais , Interações Medicamentosas , Masculino , Morfina/farmacocinética , Medição da Dor , Ratos , Ratos Sprague-Dawley , Distribuição Tecidual
6.
J Neural Transm (Vienna) ; 112(9): 1213-21, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15614425

RESUMO

Levodopa treatment has been shown to increase plasma homocysteine levels in Parkinson's disease (PD) patients and this may lead to an increased risk for coronary arterial diseases. Levodopa is metabolised via O-methylation by catechol-O-methyltransferase (COMT) using S-adenosyl-L-methionine (SAM) as the methyl donor, this leading to the subsequent formation of homocysteine. In this study, the effects of the COMT inhibitor, entacapone, on levodopa-induced hyperhomocysteinaemia were studied in rats. Using a single dose acute treatment paradigm, entacapone (10 or 30 mg/kg) prevented the levodopa (30 or 100 mg/kg) induced rise in plasma homocysteine levels in a dose-dependent manner. Five-day sub-chronic treatment with levodopa (3 x 100 mg/kg per day) resulted in a marked rise in plasma homocysteine levels when measured 2 hours post-treatment on Day 5. These levels fell but remained greater than baseline at 8 hours post-treatment on Day 5. Consistent with findings in the acute treatment test paradigm, the co-administration of entacapone (30 mg/kg) significantly (p<0.001) reduced levodopa-induced hyperhomocysteinaemia for up to 2 hours post-treatment on Day 5 of the sub-chronic study. These results suggest that entacapone may reduce levodopa-induced hyperhomocysteinaemia in PD patients.


Assuntos
Antiparkinsonianos/farmacologia , Inibidores de Catecol O-Metiltransferase , Catecóis/farmacologia , Homocisteína/sangue , Hiper-Homocisteinemia/tratamento farmacológico , Levodopa/farmacologia , Animais , Relação Dose-Resposta a Droga , Interações Medicamentosas , Inibidores Enzimáticos/farmacologia , Hiper-Homocisteinemia/induzido quimicamente , Masculino , Nitrilas , Ratos , Ratos Wistar
7.
Kidney Int ; 58(6): 2462-72, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11115079

RESUMO

BACKGROUND: Cyclosporine A (CsA)-induced hypertension and nephrotoxicity are aggravated by high sodium intake. Accumulating evidence suggests that potassium and magnesium supplementation could protect against the detrimental effects of dietary salt. In the present study, we tested the hypothesis of whether concurrent supplementation with potassium and magnesium could protect against the development of CsA-induced hypertension and nephrotoxicity more effectively than supplementation with one mineral alone. METHODS: Eight-week-old spontaneously hypertensive rats (SHRs) were divided into four groups (N = 10 in each group): (1) CsA group (5 mg/kg subcutaneously) receiving high-sodium diet (Na 2.6%, K 0.8%, Mg 0.2% wt/wt); (2) CsA group receiving a high-sodium, high-potassium diet (Na 2.6%, K 2.4%, Mg 0.2%); (3) CsA group receiving high-sodium, high-magnesium diet (Na 2.6%, K 0.8%, Mg 0.6%); and (4) CsA group receiving high-sodium, high-potassium, high-magnesium diet (Na 2.6%, K 2.4%, Mg 0.6%). RESULTS: CsA induced severe hypertension and deteriorated renal functions in SHRs on high-sodium diet. Histologically, the kidneys showed severe thickening of the media of the afferent artery with fibrinoid necrosis. Potassium supplementation lowered blood pressure (198 +/- 5 vs. 212 +/- 2 mm Hg, P < 0.05) and partially prevented the development of proteinuria (-25%, P < 0.05). Magnesium supplementation decreased blood pressure to the same extent but improved renal functions more effectively than potassium. The greatest protection against CsA toxicity was achieved when dietary potassium and magnesium supplementations were combined. Urinary N-acetyl-beta-D-glucosaminidase (NAG) excretion, a marker for renal proximal tubular damage, increased progressively in CsA-treated SHRs on the high-sodium diet. Neither potassium nor magnesium influenced urinary NAG excretion. We also estimated the activity of the renal dopaminergic system by measuring 24-hour urinary dopamine excretion rates. CsA suppressed the renal dopaminergic system during high-sodium diet. Magnesium supplementation, alone and in combination with potassium, protected against the development of renal dopaminergic deficiency in CsA-treated SHRs on high-sodium diet. Magnesium supplementation increased plasma-free ionized magnesium (iMg) and bone magnesium by 50 and 16%, respectively. CONCLUSIONS: Our findings indicate that both potassium and magnesium supplementations showed beneficial effects against CsA-induced hypertension and nephrotoxicity. The protective effect of magnesium clearly exceeded that of potassium. The greatest protection against CsA toxicity was achieved when potassium and magnesium were combined. We also provide evidence that the development of CsA-induced glomerular, tubular, and vascular lesions are associated with renal dopaminergic deficiency.


Assuntos
Ciclosporina/toxicidade , Hipertensão Renal/induzido quimicamente , Hipertensão Renal/tratamento farmacológico , Imunossupressores/toxicidade , Magnésio/farmacologia , Potássio na Dieta/farmacologia , Acetilglucosaminidase/urina , Animais , Pressão Sanguínea , Osso e Ossos/química , Colesterol/sangue , Ciclosporina/sangue , Ciclosporina/farmacocinética , Dopamina/fisiologia , Frequência Cardíaca , Hipertensão Renal/patologia , Hipertrofia Ventricular Esquerda/induzido quimicamente , Hipertrofia Ventricular Esquerda/tratamento farmacológico , Hipertrofia Ventricular Esquerda/patologia , Imunossupressores/sangue , Imunossupressores/farmacocinética , Falência Renal Crônica/induzido quimicamente , Falência Renal Crônica/tratamento farmacológico , Falência Renal Crônica/patologia , Túbulos Renais Proximais/química , Túbulos Renais Proximais/patologia , Magnésio/análise , Masculino , Miocárdio/química , Norepinefrina/urina , Proteinúria/induzido quimicamente , Proteinúria/tratamento farmacológico , Proteinúria/patologia , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Sódio na Dieta/farmacologia , Distribuição Tecidual
8.
Ann N Y Acad Sci ; 899: 238-54, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10863543

RESUMO

A fluorescent assay of brain lipid peroxidation was used for screening new antioxidants for the prevention of neurodegeneration caused by free radicals. Incubation of rat brain homogenates led to a temperature-dependent increase in production of fluorescent adducts of peroxidized polyunsaturated fatty acids; it was inhibited completely by lowering the incubation temperature to 4 degrees C. This tissue disruption-induced brain lipid peroxidation at 37 degrees C was blocked by deferoxamine (IC50 = 0.3 microM) and EDTA; it was augmented by adding submicromolar iron and hemoglobin. Ferrous ion's pro-oxidative activities were five times more potent than ferric ion. Micromolar manganese completely inhibited lipid peroxidation, confirming earlier unexpected in vivo reports. Trolox and vitamin C suppressed brain lipid peroxidation with IC50 values of 20 and 500 microM, respectively. U-78517F was approximately 20 times more potent than Trolox. 17 beta-Estradiol, hydralazine, S-nitrosoglutathione and 3-hydroxybenzylhydrazine were as potent as Trolox. Melatonin, glutathione, alpha-lipoic acid and l-deprenyl were about 20 times less potent than Trolox. Surprisingly, N-tert-butyl-alpha-phenylnitrone was a weak antioxidant. Furthermore, this procedure can also detect pro-oxidative side effects of vitamin C, oxidized glutathione, penicillamine and Angeli's salt. The present results obtained from this selective fluorescent assay are consistent with earlier reports that iron complexes promote while manganese inhibits brain lipid peroxidation caused by cell disruption. S-Nitrosoglutathione, melatonin, 17 beta-estradiol, and manganese have been successfully tested in cell/animal models for their potential neuroprotective effects. In conclusion, monitoring fluorescent adducts of peroxidizing polyunsaturated fatty acids in brain homogenates is a simple, quantitative method for studying iron-dependent brain lipid peroxidation and for screening of potential neuroprotective antioxidants in both in vitro and in vivo preparations.


Assuntos
Antioxidantes/farmacologia , Encéfalo/efeitos dos fármacos , Glutationa/análogos & derivados , Ferro/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Manganês/farmacologia , Compostos Nitrosos/farmacologia , Animais , Encéfalo/metabolismo , Quelantes/farmacologia , Glutationa/farmacologia , Masculino , Metais/farmacologia , Ratos , Ratos Sprague-Dawley , S-Nitrosoglutationa , Temperatura
9.
Neurotoxicology ; 20(2-3): 455-66, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10385904

RESUMO

Our group recently observed that manganese prevents oxidative brain injury in the iron-induced parkinsonian animal model. It has also been suggested that manganese retards while copper promotes the development of atherosclerosis. In this report, we provide further evidence to support a controversial notion that manganese is an atypical antioxidant. Among transition metals, Cu2+ and Fe2+ (0.1 to 125 microM), but not Mn2+, converted hydrogen peroxide to reactive hydroxyl radicals via the Fenton reaction at pH 7.4. Iron's pro-oxidative rate is relatively slow, but it is accelerated further by ascorbate (50 microM) in 37 degrees C Dulbecco's phosphate buffered saline. Moreover, Mn2+ (0-80 microM) concentration dependently retarded diene conjugation of human low density lipoproteins stimulated by 5 microM Cu2+. This new result is consistent with our recent finding that Mn2+ (0 to 20 microM) does not initiate brain lipid peroxidation while it inhibits iron-induced peroxidation of polyunsaturated fatty acids. These unexpected manganese results are somewhat at odds with a prominent theory that manganese is a prooxidative transition metal. Furthermore, iron and copper induced free radical generation and lipid peroxidation are suppressed by lowering the incubation temperature; this suggests that hypothermia may decrease the oxidative stress and damage in vivo. In conclusion, normal dietary intake of manganese may protect cells and neurons from oxidant stress through the inhibition of propagation of lipid peroxidation caused by hydroxyl radicals generated by pro-oxidative transition metals such as iron and copper. Potential therapeutical uses of manganese, manganese SOD mimetics and hypothermia for protecting brain neurons and vascular endothelial cells against oxidative stress and damage have been successfully demonstrated in both animal models and clinical trials.


Assuntos
Antioxidantes/farmacologia , LDL-Colesterol/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Manganês/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Animais , Cobre/farmacologia , Relação Dose-Resposta a Droga , Fluorescência , Humanos , Hipotermia/metabolismo , Técnicas In Vitro , Ferro/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Ratos
10.
Free Radic Res ; 31(6): 631-40, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10630686

RESUMO

In the present in vitro and in vivo study we investigated the pro-oxidant effects of hemoglobin, as well as the antioxidant effects of its metabolites, in the brain. Incubation of rat brain homogenates with hemoglobin (0-10 microM) but not hemin induced lipid peroxidation up to 24 h (EC50 = 1.2 microM). Hemoglobin's effects were similar to ferrous ion (EC50 = 1.7 microM) and were blocked by the chelating agent deferoxamine (IC50 0.5 microM) and a nitric oxide-releasing compound S-nitrosoglutathione (IC50 = 40 microM). However, metabolites of hemoglobin - biliverdin and bilirubin - inhibited brain lipid peroxidation induced by cell disruption and hemoglobin (biliverdin IC50 = 12-30 and bilirubin IC50 = 75-170 microM). Biliverdin's antioxidative effects in spontaneous and iron-evoked lipid peroxidation were further augmented by manganese (2 microM) since manganese is an antioxidative transition metal and conjugates with bile pigments. Intrastriatal infusion of hemoglobin (0-24 nmol) produced slight, but significant 20-22% decreases in striatal dopamine levels. Whereas, intrastriatal infusion of ferrous citrate (0-24 nmol) dose-dependently induced a greater 66% depletion of striatal dopamine which was preceded by an acute increase of lipid peroxidation. In conclusion, contrary to the in vitro results hemoglobin is far less neurotoxic than ferrous ions in the brain. It is speculated that hemoglobin may be partially detoxified by heme oxygenase and biliverdin reductase to its antioxidative metabolites in the brain. However, in head trauma and stroke, massive bleeding could significantly produce iron-mediated oxidative stress and neurodegeneration which could be minimized by endogenous antioxidants such as biliverdin, bilirubin, manganese and S-nitrosoglutathione.


Assuntos
Pigmentos Biliares/metabolismo , Encéfalo/metabolismo , Glutationa/análogos & derivados , Hemoglobinas/metabolismo , Ferro/metabolismo , Manganês/metabolismo , Compostos Nitrosos/metabolismo , Estresse Oxidativo/fisiologia , Animais , Antioxidantes/metabolismo , Pigmentos Biliares/farmacologia , Bilirrubina/metabolismo , Bilirrubina/farmacologia , Biliverdina/metabolismo , Biliverdina/farmacologia , Encéfalo/citologia , Encéfalo/efeitos dos fármacos , Ácido Cítrico , Desferroxamina/farmacologia , Compostos Ferrosos/farmacologia , Glutationa/metabolismo , Glutationa/farmacologia , Hemoglobinas/farmacologia , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Manganês/farmacologia , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Compostos Nitrosos/farmacologia , Oxidantes/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , S-Nitrosoglutationa , Extratos de Tecidos/metabolismo
11.
Free Radic Res ; 31(6): 641-50, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10630687

RESUMO

Recent results demonstrated that S-nitrosoglutathione (GSNO) and nitric oxide (*NO) protect brain dopamine neurons from hydroxyl radical (*OH)-induced oxidative stress in vivo because they are potent antioxidants. GSNO and *NO terminate oxidant stress in the brain by (i) inhibiting iron-stimulated hydroxyl radicals formation or the Fenton reaction, (ii) terminating lipid peroxidation, (iii) augmenting the antioxidative potency of glutathione (GSH), (iv) mediating neuroprotective action of brain-derived neurotrophin (BDNF), and (v) inhibiting cysteinyl proteases. In fact, GSNO--S-nitrosylated GSH--is approximately 100 times more potent than the classical antioxidant GSH. In addition, S-nitrosylation of cysteine residues by GSNO inactivates caspase-3 and HIV-1 protease, and prevents apoptosis and neurotoxicity. GSNO-induced antiplatelet aggregation is also mediated by S-nitrosylation of clotting factor XIII. Thus the elucidation of chemical reactions involved in this GSNO pathway (GSH GS* + *NO-->[GSNO]-->GSSG + *NO-->GSH) is necessary for understanding the biology of *NO, especially its beneficial antioxidative and neuroprotective effects in the CNS. GSNO is most likely generated in the endothelial and astroglial cells during oxidative stress because these cells contain mM GSH and nitric oxide synthase. Furthermore, the transfer of GSH and *NO to neurons via this GSNO pathway may facilitate cell to neuron communications, including not only the activation of guanylyl cyclase, but also the nitrosylation of iron complexes, iron containing enzymes, and cysteinyl proteases. GSNO annihilates free radicals and promotes neuroprotection via its c-GMP-independent nitrosylation actions. This putative pathway of GSNO/GSH/*NO may provide new molecular insights for the redox cycling of GSH and GSSG in the CNS.


Assuntos
Comunicação Celular/fisiologia , Glutationa/análogos & derivados , Glutationa/metabolismo , Neurônios/metabolismo , Óxido Nítrico/metabolismo , Compostos Nitrosos/metabolismo , Animais , Antioxidantes/metabolismo , Astrócitos/citologia , Astrócitos/metabolismo , Encéfalo/metabolismo , Caspases/metabolismo , Endotélio/citologia , Endotélio/metabolismo , Ativação Enzimática , Glutationa/química , Protease de HIV/metabolismo , Inibidores da Protease de HIV/metabolismo , Humanos , Peroxidação de Lipídeos , Camundongos , Neurônios/citologia , Óxido Nítrico/química , Compostos Nitrosos/química , Oxirredução , S-Nitrosoglutationa
12.
Neuroscience ; 85(4): 1101-11, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9681949

RESUMO

It has been suggested that transition metals such as iron and manganese produce oxidative injury to the dopaminergic nigrostriatal system. which may play a critical role in the pathogenesis of Parkinson's disease. Intranigral infusion of ferrous citrate (0 to 8.4 nmol, i.n.) acutely increased lipid peroxidation in the substantia nigra and dopamine turnover in the caudate nucleus. Subsequently, it caused a severe depletion of dopamine levels in the rat caudate nucleus. In contrast to iron's pro-oxidant effect, manganese (up to 30 nmol, i.n.) causes neither lipid peroxidation nor nigral injury/dopamine depletion. Manganese (1.05 to 4.2 nmol, i.n.) dose-dependently protected nigral neurons from iron-induced oxidative injury and dopamine depletion. Manganese also suppressed acute increase in dopamine turnover and contralateral turning behaviour induced by iron. In brain homogenates manganese (0 to 10 microM) concentration-dependently inhibited propagation of lipid peroxidation caused by iron (0 to 5 microM). Without the contribution of manganese-superoxide dismutase manganese was still effective in sodium azide and/or heat-pretreated brain homogenates. Surprisingly, iron but not manganese, catalysed the Fenton reaction or the conversion of hydrogen peroxide to hydroxyl radicals. The results indicate that iron and manganese are two transition metals mediating opposite effects in the nigrostriatal system, as pro-oxidant and antioxidant, respectively. In conclusion, intranigral infusion of iron, but not manganese, provides an animal model for studying the pathophysiological role of oxidant and oxidative stress in nigrostriatal degeneration and Parkinsonism. The present results further suggest that the atypical antioxidative properties of manganese may protect substantia nigra compacta neurons from iron-induced oxidative stress.


Assuntos
Ferro , Manganês/farmacologia , Neostriado/citologia , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Doença de Parkinson Secundária/metabolismo , Substância Negra/citologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Núcleo Caudado/citologia , Núcleo Caudado/efeitos dos fármacos , Dopamina/metabolismo , Dopamina/fisiologia , Radical Hidroxila/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Mesencéfalo/citologia , Mesencéfalo/efeitos dos fármacos , Neostriado/efeitos dos fármacos , Doença de Parkinson Secundária/induzido quimicamente , Doença de Parkinson Secundária/fisiopatologia , Ratos , Ratos Sprague-Dawley , Comportamento Estereotipado/efeitos dos fármacos , Substância Negra/efeitos dos fármacos , Superóxido Dismutase/metabolismo
13.
Free Radic Biol Med ; 24(7-8): 1065-73, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9626559

RESUMO

Sodium nitroprusside (disodium nitroferricyanide) has been suggested to cause cytotoxicity through either the release of cyanide and/or nitric oxide. The present study investigated a possible mechanism that after a brief release of nitric oxide, iron moiety of breakdown products of sodium nitroprusside could cause a long lasting oxidative stress, such as hydroxyl radical generation, lipid peroxidation and cytotoxicity. Intranigral administration of sodium nitroprusside (0-16.8 nmol) to rats induced an acute increase in lipid peroxidation in the substantia nigra and a chronic dopamine depletion in the caudate nucleus. Photodegraded (nitric oxide-exhausted) sodium nitroprusside, however, still produced lipid peroxidation and neurotoxicity in the midbrain. Moreover, non-iron containing nitric oxide-donor compounds, such as S-nitroso-N-acetylpenicillamine, did not cause oxidative brain injury in vivo suggesting that nitric oxide may not mediate neurotoxicity induced by sodium nitroprusside. Additional in vitro studies demonstrated that both freshly prepared (nitric oxide donor) and photodegraded (nitric oxide-exhausted) sodium nitroprusside generated hydroxyl radicals in the presence of ascorbate and also increased lipid peroxidation in brain homogenates. These pro-oxidative effects of sodium nitroprusside were blocked by nitric oxide, S-nitroso-N-acetylpenicillamine, oxyhemoglobin, and deferoxamine (iron chelator). The present results suggest that iron moiety, rather than nitric oxide, may mediate the pro-oxidative properties of sodium nitroprusside. With this new information in mind, the misuse of sodium nitroprusside as a selective nitric oxide donor in both basic and clinical uses should be urgently addressed.


Assuntos
Lesões Encefálicas/induzido quimicamente , Lesões Encefálicas/metabolismo , Radical Hidroxila/metabolismo , Nitroprussiato/toxicidade , Animais , Ácido Ascórbico/farmacologia , Lesões Encefálicas/patologia , Desferroxamina/farmacologia , Peróxido de Hidrogênio/farmacologia , Técnicas In Vitro , Ferro/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Degeneração Neural/induzido quimicamente , Óxido Nítrico/metabolismo , Óxido Nítrico/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Oxiemoglobinas/farmacologia , Penicilamina/análogos & derivados , Penicilamina/farmacologia , Ratos , Ratos Sprague-Dawley
14.
FASEB J ; 12(2): 165-73, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9472981

RESUMO

The proposed anti- and pro-oxidant effects of nitric oxide (NO) derivatives, such as S-nitrosoglutathione (GSNO) and peroxynitrite, were investigated in the rat nigrostriatal dopaminergic system. Intranigral infusion of freshly prepared GSNO (0-16.8 nmol, i.n.) prevented iron-induced (4.2 nmol, i.n.) oxidative stress and nigral injury, reflected by a decrease in striatal dopamine levels. This neuroprotective effect of GSNO was verified by ex vivo imaging of brain dopamine uptake sites using 125I-labeled RTI-55. In addition, in vitro data indicate that GSNO concentration-dependently inhibited iron-evoked hydroxyl radical generation and brain lipid peroxidation. In this iron-induced oxidant stress model, GSNO was approximately 100-fold more potent than the antioxidant glutathione (GSH). Light-exposed, NO-exhausted GSNO produced neither antioxidative nor neuroprotective effects, which indicates that NO may mediate at least part of GSNO's effects. Moreover, GSNO completely (and GSH only partially) inhibited the weak pro-oxidant effect of peroxynitrite, which produced little injury to nigral neurons in vivo. This study provides relevant in vivo evidence suggesting that nanomol GSNO can protect brain dopamine neurons from iron-induced oxidative stress and degeneration. In conclusion, S-nitrosylation of GSH by NO and oxygen may be part of the antioxidative cellular defense system.


Assuntos
Dopamina/metabolismo , Glutationa/análogos & derivados , Peroxidação de Lipídeos/efeitos dos fármacos , Neurônios/fisiologia , Fármacos Neuroprotetores/farmacologia , Compostos Nitrosos/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Substância Negra/fisiologia , Animais , Autorradiografia , Ácido Cítrico , Cocaína/análogos & derivados , Cocaína/farmacocinética , Compostos Ferrosos/toxicidade , Glutationa/administração & dosagem , Glutationa/farmacologia , Infusões Parenterais , Radioisótopos do Iodo , Masculino , Neurônios/efeitos dos fármacos , Neurônios/patologia , Fármacos Neuroprotetores/administração & dosagem , Nitratos/toxicidade , Compostos Nitrosos/administração & dosagem , Oxidantes/toxicidade , Ratos , Ratos Sprague-Dawley , S-Nitrosoglutationa , Substância Negra/efeitos dos fármacos , Substância Negra/patologia
16.
Synapse ; 23(1): 58-60, 1996 May.
Artigo em Inglês | MEDLINE | ID: mdl-8723136

RESUMO

Intranigral infusion of ferrous citrate (4.2 nmol) induced an acute lipid peroxidation in the substantia nigra and a chronic dopamine depletion in the striatum of rat nigrostriatal system. Coinfusion of 8.4 nmol nitric oxide donors such as S-nitrosoglutathione (GSNO) and S-nitroso-N-acetylpenicillamine (SNAP) or nitric oxide (approximately 2 nmol) protected nigrostriatal neurons against iron-induced lipid peroxidation and associated oxidative injury. However, sodium nitroprusside (SNP, 8.4 nmol) augmented dopamine depletion caused by ferrous citrate because SNP is a ferricyanide complex. The present in vivo results indicate that nitric oxide and S-nitrosothiols are antioxidants which can protect brain dopamine neurons against oxidant stress/damage.


Assuntos
Corpo Estriado/efeitos dos fármacos , Dopamina/metabolismo , Mercaptoetanol , Óxido Nítrico/farmacologia , Nitroprussiato/farmacologia , Compostos Nitrosos/farmacologia , Estresse Oxidativo/efeitos dos fármacos , S-Nitrosotióis , Substância Negra/efeitos dos fármacos , Animais , Ferro/farmacologia , Masculino , Ratos , Ratos Sprague-Dawley
17.
Pharmacol Toxicol ; 78(5): 354-60, 1996 May.
Artigo em Inglês | MEDLINE | ID: mdl-8737973

RESUMO

We studied effects of anxiogenic and anxiolytic compounds on the electric self-stimulation of the medial fore-brain bundle in male rats to find out if there is a link between reward and anxiety-related behaviours. The cholecystokinin agonist, caerulein (25-100 micrograms/kg) and the 5-HT agonist 1-(3-chlorophenyl)piperazine (0.2-1 mg/kg) dose-dependently inhibited the electric self-stimulation. The 5-HT2A antagonist, ketanserin, at 2.5 mg/kg, increased the self-stimulation at high currents but not at threshold current. The 5-HT3 antagonist ondansetron (10 and 100 micrograms/kg). The alpha 1-adrenergic antagonist, prazosin (0.125 and 0.5 mg/kg), the beta-adrenergic antagonist, propranolol (5 and 10 mg/kg) and the alpha 2-adreno-receptor antagonist, atipamezole (4 mg/kg), did not affect the self-stimulation. Nor did the benzodiazepine agonist, diazepam (5-15 mg/kg), a benzodiazepine receptor antagonist flumazenil (at 10 and 25 mg/kg) or the inverse agonist of benzodiazepine receptors, N-methyl-beta-carboline-3-carboxamide (10 and 20 mg/kg), cause any substantial changes of the self-stimulation. We conclude that only two anxiolytic drugs (caerulein and 1-(3-chlorophenyl)piperazine) suppress the electric self-stimulation. These findings indicate that anxiogenicity as such is not able to weaken the hypothalamic electric self-stimulation. Anxiety and reward are apparently mediated through separate neural pathways.


Assuntos
Ansiedade/fisiopatologia , Nível de Alerta/efeitos dos fármacos , Estimulantes do Sistema Nervoso Central/farmacologia , Hipotálamo/efeitos dos fármacos , Autoestimulação/efeitos dos fármacos , Anfetamina/farmacologia , Animais , Antipsicóticos/farmacologia , Ceruletídeo/farmacologia , Masculino , Pimozida/farmacologia , Ratos , Ratos Wistar
18.
Pharmacol Toxicol ; 78(3): 129-35, 1996 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8882344

RESUMO

Rats were made tolerant to morphine by a 5-day regimen with increasing doses. The time course of changes in serum anterior pituitary hormone levels, brain endo- and exopeptidase activity, levels of brain biogenic amines and body weight were studied during abrupt morphine withdrawal. Cold stimulated secretion of thyrotropin and the secretion of growth hormone were both decreased whereas that of prolactin was increased. In the hypothalamus both prolyl endopeptidase and dipeptidyl peptidase IV activities were concomitantly increased. The hypothalamic 5 hydroxyindole acetic acid levels were also increased. Changes in hormone secretion, peptidase activity and monoamine turnover had returned to baseline levels by 92 hr. Our results indicate that morphine withdrawal and the associated stress produce alterations in anterior pituitary thyrotropin and growth hormone secretion. Concomitant increases in hypothalamic prolyl endopeptidase and dipeptidyl peptidase activities may contribute to these changes.


Assuntos
Encéfalo/enzimologia , Encéfalo/metabolismo , Catecolaminas/metabolismo , Endopeptidases/metabolismo , Morfina/toxicidade , Adeno-Hipófise/efeitos dos fármacos , Adeno-Hipófise/metabolismo , Hormônios Adeno-Hipofisários/metabolismo , Síndrome de Abstinência a Substâncias/patologia , Animais , Ativação Enzimática/efeitos dos fármacos , Hipotálamo/enzimologia , Hipotálamo/metabolismo , Masculino , Dependência de Morfina/patologia , Ratos , Ratos Wistar , Síndrome de Abstinência a Substâncias/enzimologia , Síndrome de Abstinência a Substâncias/metabolismo
19.
Pharmacol Toxicol ; 78(3): 136-42, 1996 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8882345

RESUMO

The role of alpha 2-adrenergic receptors (adrenoceptors) in the secretion of growth hormone, prolactin and thyrotropin was studied using highly selective agonists and antagonists of the alpha 2-adrenoceptor. The interplay between opiates and alpha 2-adrenergic drugs in the acute secretion of growth hormone and prolactin, as well as the possible cross-tolerance between morphine (mu-opioid receptor agonist) and dexmedetomidine (alpha 2-adrenoceptor agonist) in growth hormone secretion were also evaluated. Dexmedetomidine dose-dependently increased plasma growth hormone and prolactin levels and decreased thyrotropin levels. The enhanced secretion of both growth hormone and prolactin was antagonized by atipamezole (an alpha 2-adrenoceptor antagonist) but not by prazosin (an alpha 1-adrenoceptor antagonist). Morphine (5 mg/kg)-induced stimulation of growth hormone secretion was antagonized by both naloxone (mu-opioid antagonist) and atipamezole. Naloxone, but not atipamezole, antagonized the morphine-induced increase in prolactin secretion. Dexmedetomidine increased growth hormone secretion in the saline pretreated rats, but did not do so in the morphine-tolerant rats. The stimulation of alpha 2-adrenoceptor enhances secretion of both growth hormone and prolactin. The adrenergic regulation of thyrotropin secretion still remains unclear. Evidently, adrenergic mechanisms are involved in the morphine-induced stimulation of growth hormone secretion, but not in the morphine-induced stimulation of prolactin secretion. In addition, there is a clear cross-tolerance between dexmedetomidine and morphine in growth hormone secretion of the rat.


Assuntos
Agonistas alfa-Adrenérgicos/farmacologia , Hormônio do Crescimento/metabolismo , Imidazóis/farmacologia , Hormônios Adeno-Hipofisários/metabolismo , Receptores Opioides mu/agonistas , Animais , Tolerância a Medicamentos , Hormônio do Crescimento/efeitos dos fármacos , Masculino , Medetomidina , Morfina/farmacologia , Prolactina/efeitos dos fármacos , Prolactina/metabolismo , Ratos , Ratos Wistar , Tireotropina/efeitos dos fármacos , Tireotropina/metabolismo
20.
Free Radic Biol Med ; 21(3): 391-4, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-8855451

RESUMO

The pro-oxidant effects of hydroxyl radical (.OH, ferrous ammonium sulfate/Fe2+) or nitric oxide (NO., S-nitroso-N-acetylpenicillamine/SNAP) generating compounds were studied in rat brain homogenate preparations. Submicromolar concentrations of Fe2+, but not SNAP (up to 100 microM), increased the formation of fluorescent products of malondialdehyde in cortical homogenates. In fact, iron-catalyzed brain lipid peroxidation was inhibited by SNAP (100 microM), but not by light-exposed SNAP or its degradation product penicillamine (100 microM). This study provides relevant evidence to suggest that submicromolar concentrations of Fe2+ can potentiate lipid peroxidation in disrupted brain tissue. NO. released from SNAP did not stimulate, but rather inhibited brain lipid peroxidation. These results support the hypothesis that NO., as opposed to .OH radicals, is not a pro-oxidant but rather an antioxidant.


Assuntos
Encéfalo/metabolismo , Radical Hidroxila/metabolismo , Peroxidação de Lipídeos , Óxido Nítrico/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Compostos Ferrosos/farmacologia , Radical Hidroxila/farmacologia , Masculino , Malondialdeído/metabolismo , Óxido Nítrico/farmacologia , Penicilamina/análogos & derivados , Penicilamina/farmacologia , Ratos , Ratos Sprague-Dawley , S-Nitroso-N-Acetilpenicilamina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...