Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Pflugers Arch ; 471(4): 583-593, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30613864

RESUMO

Disturbed homeostasis of nitric oxide (NO) is one of the causes of myocardial ischemia/reperfusion (I/R) injury during open-heart surgery. This study was designed to explore mechanisms of action of dinitrosyl iron complexes with reduced glutathione ({(GS-)2Fe+(NO+)2}+, DNIC-GS) added to crystalloid cardioplegia or reperfusion solution in isolated working rat hearts. Hearts of male Wistar rats were subjected to cardioplegic arrest by St. Thomas' Hospital cardioplegic solution (STH) and normothermic global ischemia followed by reperfusion. DNIC-GS were used with STH or during early reperfusion. Lactate dehydrogenase (LDH) activity in the coronary effluent and myocardial contents of adenine nucleotides, phosphocreatine, and lactate were determined spectrophotometrically. Reactive oxygen species (ROS) formation in the coronary effluent and myocardial DNIC content was assessed by EPR technique. Cardioplegia or reperfusion with DNIC-GS significantly improved recovery of coronary flow and cardiac function compared with control. Carboxy-[2-(4-carboxyphenyl)-4,4,5,5-tetramethyl-imidozoline-1-oxy-3-oxide] (C-PTIO), a selective NO scavenger, reduced/abolished protective action of DNIC-GS. Enhanced recovery of cardiac function with DNIC-GS reduced LDH release in the coronary effluent, augmented recovery of myocardial energy state, and decreased formation of ROS-generating systems at reperfusion. Beneficial effects of DNIC-GS were related to the transfer of [Fe(NO)2] cores to thiol groups of myocardial proteins to form intracellular DNIC pools. The study concluded that DNIC-GS is a promising adjunct agent for metabolic and antioxidant protection of the heart during cardioplegic arrest and reperfusion.


Assuntos
Glutationa/farmacologia , Coração/efeitos dos fármacos , Ferro/farmacologia , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Óxidos de Nitrogênio/farmacologia , Animais , Antioxidantes/metabolismo , Parada Cardíaca Induzida/métodos , Soluções Isotônicas/metabolismo , Masculino , Traumatismo por Reperfusão Miocárdica/metabolismo , Miocárdio/metabolismo , Óxido Nítrico/metabolismo , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo
2.
Br J Pharmacol ; 172(12): 2933-45, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25521429

RESUMO

BACKGROUND AND PURPOSE: Mitochondria-derived oxidative stress is believed to be crucially involved in cardiac ischaemia reperfusion (I/R) injury, although currently no therapies exist that specifically target mitochondrial reactive oxygen species (ROS) production. The present study was designed to evaluate the potential effects of the structural analogues of apelin-12, an adipocyte-derived peptide, on mitochondrial ROS generation, cardiomyocyte apoptosis, and metabolic and functional recovery to myocardial I/R injury. EXPERIMENTAL APPROACH: In cultured H9C2 cardiomyoblasts and adult cardiomyocytes, oxidative stress was induced by hypoxia reoxygenation. Isolated rat hearts were subjected to 35 min of global ischaemia and 30 min of reperfusion. Apelin-12, apelin-13 and structural apelin-12 analogues, AI and AII, were infused during 5 min prior to ischaemia. KEY RESULTS: In cardiac cells, mitochondrial ROS production was inhibited by the structural analogues of apelin, AI and AII, in comparison with the natural peptides, apelin-12 and apelin-13. Treatment of cardiomyocytes with AI and AII decreased cell apoptosis concentration-dependently. In a rat model of I/R injury, pre-ischaemic infusion of AI and AII markedly reduced ROS formation in the myocardial effluent and attenuated cell membrane damage. Prevention of oxidative damage by AI and AII was associated with the improvement of functional and metabolic recovery after I/R in the heart. CONCLUSIONS AND IMPLICATIONS: These data provide the evidence for the potential of the structural apelin analogues in selective reduction of mitochondrial ROS generation and myocardial apoptosis and form the basis for a promising therapeutic strategy in the treatment of oxidative stress-related heart disease.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Animais , Apoptose/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Membrana Celular/patologia , Relação Dose-Resposta a Droga , Peptídeos e Proteínas de Sinalização Intercelular/administração & dosagem , Peptídeos e Proteínas de Sinalização Intercelular/química , Masculino , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Ratos , Ratos Wistar
3.
Eur J Pharmacol ; 662(1-3): 40-6, 2011 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21550336

RESUMO

It has been found that dinitrosyl iron complexes with glutathione (DNIC-GS) injected into the blood flow of rats at a dose of 0.05 µmoles/kg prior to hemorrhage significantly improve cardiac function under conditions of hemorrhagic shock manifested in increased stroke volume, left ventricular work and cardiac output to a level exceeding control values 1.5-fold. Enhanced myocardial contractile activity leads to a situation where mean arterial pressure does not decrease further despite the significant decrease of total peripheral resistance. The decrease of total peripheral vascular resistance of the vascular system under vasodilating effects of DNIC-GS used as nitric oxide donors improves microcirculation in experimental rats judging from increased rates of blood flow and low degree of erythrocyte aggregation. Pretreatment of rats with the complexes significantly increases survival (by 21%) under conditions of hemorrhagic shock. It is suggested that beneficial effects of DNIC-GS on systemic circulation parameters under conditions of hemorrhagic shock are determined by their antioxidant activity and the ability to induce S-nitrosylation of proteins.


Assuntos
Glutationa/química , Ferro/química , Ferro/farmacologia , Óxidos de Nitrogênio/química , Óxidos de Nitrogênio/farmacologia , Choque Hemorrágico/tratamento farmacológico , Cloreto de Sódio/farmacologia , Animais , Circulação Sanguínea/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Relação Dose-Resposta a Droga , Feminino , Ferro/uso terapêutico , Óxidos de Nitrogênio/uso terapêutico , Ratos , Ratos Wistar , Choque Hemorrágico/fisiopatologia , Análise de Sobrevida
4.
Methods Mol Biol ; 704: 135-49, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21161635

RESUMO

It has been established that microdialysis ensured by the passage of aqueous solutions of Fe(3+) complexes with N-methyl-D: -glucamine dithiocarbamate (MGDMGD ) through fine dialysis fibers permeable for compounds with molecular weights below 5 kDa. These fibers can be implanted into heart, liver, and kidney tissues, enabling effective binding of Fe(3+)-MGD complexes to nitric oxide generated in interstitial fluids of narcotized rats in vivo. Subsequent treatment of dialyzate samples (60 µL) with sodium dithionite favors conversion of newly formed diamagnetic NO-Fe(3+)-MGD complexes into electron paramagnetic resonance-detectable NO-Fe(2+)-MGD complexes. The basal levels of NO determined from the concentrations of the complexes in the respective tissues are similar (1 µÐœ). The microdialysis data suggest that treatment of rats with a water-soluble analogue of nitroglycerine or a dinitrosyl iron complex with thiosulfate induces a long-lasting (>1 h) increase in the steady-state level of NO in animal tissues. This novel technology can be used for comparative analyses of production rates of NO and reactive oxygen species when using iron-dithiocarbamate complexes and spin traps for reactive oxygen species, respectively.


Assuntos
Espectroscopia de Ressonância de Spin Eletrônica/métodos , Óxido Nítrico/análise , Animais , Compostos Ferrosos/química , Coração , Rim/química , Rim/metabolismo , Fígado/química , Fígado/metabolismo , Masculino , Óxido Nítrico/biossíntese , Especificidade de Órgãos , Ratos , Ratos Wistar
5.
J Control Release ; 142(1): 89-100, 2010 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-19815037

RESUMO

A synthetic amphiphilic block copolymer, Pluronic, is a potent chemosensitizer of multidrug resistant (MDR) cancers that has shown promise in clinical trials. It has unique activities in MDR cells, which include a decrease in ATP pools and inhibition of P-glycoprotein (Pgp) resulting in increased drug accumulation in cells. This work demonstrates that Pluronic rapidly (15min) translocates into MDR cells and co-localizes with the mitochondria. It inhibits complex I and complex IV of the mitochondria respiratory chain, decreases oxygen consumption and causes ATP depletion in MDR cells. These effects are selective and pronounced for MDR cells compared to non-MDR counterparts and demonstrated for both drug-selected and Pgp-transfected cell models. Furthermore, inhibition of Pgp functional activity also abolishes the effects of Pluronic on intracellular ATP levels in MDR cells suggesting that Pgp contributes to increased responsiveness of molecular "targets" of Pluronic in the mitochondria of MDR cells. The Pluronic-caused impairment of respiration in mitochondria of MDR cells is accompanied with a decrease in mitochondria membrane potential, production of ROS, and release of cytochrome c. Altogether these effects eventually enhance drug-induced apoptosis and contribute to potent chemosensitization of MDR tumors by Pluronic.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Carcinoma/tratamento farmacológico , Resistência a Múltiplos Medicamentos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Poloxâmero/farmacologia , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/antagonistas & inibidores , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Trifosfato de Adenosina/metabolismo , Linhagem Celular Tumoral , Citocromos c/metabolismo , Humanos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Óxidos de Nitrogênio/metabolismo , Consumo de Oxigênio/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo
6.
Nitric Oxide ; 19(4): 338-44, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18664386

RESUMO

It was found that microdialysis, i.e., passage of aqueous solutions of iron-N-methyl-D-glucamine dithiocarbamate complexes through dialysis fibers implanted into heart, kidney and liver tissues of narcotized rats, was accompanied by effective binding of the complexes to nitric oxide from interstitial fluid. The walls of dialysis fibers used in this study were permeable for compounds with molecular weight not exceeding 5 kDa. The dialyzate samples collected every 20 min and containing diamagnetic nitrosyl Fe3+-MGD adducts were reduced to the paramagnetic state with sodium dithionite; their concentration was measured by the EPR method. The basic level of the adducts, which represented mononitrosyl iron complexes with MGD (MNIC-MGD), in the dialyzate samples of all tested organs were similar (1 microM). Treatment of animals with the water-soluble nitroglycerine analog Isoket or a low-molecular dinitrosyl iron thiosulfate complex as a NO donor increased the concentration of MNIC-MGD with going out into a plateau. The novel approach allows determination of nitric oxide levels in tissue interstitial fluid from concentration of MNIC-MGD formed during microdialysis.


Assuntos
Líquido Extracelular/metabolismo , Óxido Nítrico/metabolismo , Sorbitol/análogos & derivados , Detecção de Spin/métodos , Tiocarbamatos/farmacologia , Ar , Animais , Espectroscopia de Ressonância de Spin Eletrônica/métodos , Ferro/farmacologia , Dinitrato de Isossorbida/farmacologia , Masculino , Microdiálise , Doadores de Óxido Nítrico/metabolismo , Doadores de Óxido Nítrico/farmacologia , Óxidos de Nitrogênio/farmacologia , Ratos , Ratos Wistar , Sorbitol/farmacologia , Marcadores de Spin , Distribuição Tecidual
7.
Methods Enzymol ; 436: 445-61, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18237648

RESUMO

Prooxidant and antioxidant properties of nitric oxide (NO) during oxidative stress are mostly dependent on its interaction with reactive oxygen species, Fe ions, and hemoproteins. One form of NO storage and transportation in cells and tissues is dinitrosyl iron complexes (DNIC), which can bind with both low-molecular-weight thiols and proteins, including hemoglobin. It was shown that dinitrosyl iron complexes bound with hemoglobin (Hb-DNIC) were formed in rabbit erythrocytes after bringing low-molecular-weight DNIC with thiosulfate into blood. It was ascertained that Hb-DNIC intercepted free radicals reacting with hemoglobin SH-groups and prevented oxidative modification of this protein caused by hydrogen peroxide. Destruction of Hb-DNIC can take place in the presence of both hydrogen peroxide and tert-butyl hydroperoxide. Hb-DNIC can also be destroyed at the enzymatic generation of superoxide-anion radical in the xanthine-xanthine oxidase system. If aeration in this system was absent, formation of the nitrosyl R-form of hemoglobin could be seen during the process of Hb-DNIC destruction. Study of Hb-DNIC interaction with reactive oxygen metabolites is important for understanding NO and Hb roles in pathological processes that could result from oxidative stress.


Assuntos
Hemoglobinas/química , Hemoglobinas/metabolismo , Ferro/química , Ferro/metabolismo , Óxidos de Nitrogênio/química , Óxidos de Nitrogênio/metabolismo , Animais , Espectroscopia de Ressonância de Spin Eletrônica , Feminino , Radicais Livres/metabolismo , Peróxido de Hidrogênio/metabolismo , Peróxido de Hidrogênio/farmacologia , Oxirredução , Estresse Oxidativo , Ligação Proteica , Coelhos , Marcadores de Spin , Superóxidos/metabolismo , terc-Butil Hidroperóxido/metabolismo , terc-Butil Hidroperóxido/farmacologia
8.
Nitric Oxide ; 18(3): 195-203, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18222183

RESUMO

EPR studies have shown that water-soluble mononitrosyl iron complexes with N-methyl-d-glucamine dithiocarbamate (MNIC-MGD) (3 micromol) injected to intact mice were decomposed virtually completely within 1h. The total content of MNIC-MGD in animal urine did not exceed 30 nmol/ml. In the liver, a small amount of MNIC-MGD were converted into dinitrosyl iron complexes (30 nmol/g of liver tissue). The same was observed in intact rabbits in which MNIC-MGD formation was induced by endogenous or exogenous NO binding to NO traps, viz., iron complexes with MGD. In mice, the content of MNIC-MGD in urine samples did not change after bacterial lipopolysaccharide-induced expression of iNOS. It was supposed that MNIC-MGD decomposition in intact animals was largely due to the release of NO from the complexes and its further transfer to other specific acceptors. In mice with iNOS expression, the main contribution to MNIC-MGD decomposition was made by superoxide ions whose destructive effect is mediated by an oxidative mechanism. This effect could fully compensate the augmented synthesis of MNIC-MGD involving endogenous NO whose production was supported by iNOS. Water-soluble dinitrosyl iron complexes (DNIC) with various thiol-containing ligands and thiosulfate injected to intact mice were also decomposed; however, in this case the effect was less pronounced than in the case of MNIC-MGD. It was concluded that DNIC decomposition was largely due to the oxidative effect of superoxide ions on these complexes.


Assuntos
Compostos Ferrosos/metabolismo , Ferro/metabolismo , Fígado/metabolismo , Óxidos de Nitrogênio/metabolismo , Sorbitol/análogos & derivados , Compostos de Sulfidrila/metabolismo , Tiocarbamatos/metabolismo , Animais , Espectroscopia de Ressonância de Spin Eletrônica/métodos , Feminino , Compostos Ferrosos/química , Compostos Ferrosos/farmacocinética , Injeções Intraperitoneais , Ferro/química , Ligantes , Lipopolissacarídeos/farmacologia , Fígado/química , Masculino , Camundongos , Óxido Nítrico/biossíntese , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/biossíntese , Óxido Nítrico Sintase Tipo II/efeitos dos fármacos , Óxidos de Nitrogênio/química , Coelhos , Solubilidade , Sorbitol/química , Sorbitol/metabolismo , Sorbitol/farmacocinética , Marcadores de Spin , Compostos de Sulfidrila/química , Compostos de Sulfidrila/farmacocinética , Tiocarbamatos/química , Tiocarbamatos/farmacocinética , Distribuição Tecidual , Água/química
9.
Chemphyschem ; 8(12): 1863-9, 2007 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-17634998

RESUMO

The unexpected effects of Ca(2+) on the free-radical chain reactions of dopamine, norepinephrine, isoproterenol, and pyrocatechol oxidation are studied using oxygen consumption measurements, EPR-spectroscopy, UV/VIS spectrophotometry, and by potentiometric titration. It is found that the formation of Ca(2+)-catecholate complexes is accompanied by an increase in the dissociation constants (K(ai) ) of their phenolic hydroxyls. At pH>pK(ai) and in the presence of alkaline-earth metal cations, the rate of catecholate oxidation increases (Ca(2+), Mg(2+)> Sr(2+), Ba(2+)), whereas on addition of Zn ions the rate decreases. The effects of Group II metal cations on catecholate autoxidation are concomitant with a transient increase of the EPR signal for metal-semiquinonate complexes. Therefore, the effects of Ca(2+) and other alkaline-earth metal cations on catecholate autoxidation can be defined as 1) additional deprotonation of catechol OH-groups involved in the formation of M(2+)-catecholate complexes, the latter exceeding catechols in the susceptibility to dioxygen-induced oxidation and 2) formation of relatively stable free-radical intermediates responsible for chain propagation.


Assuntos
Catecóis/química , Metais Alcalinoterrosos/química , Oxigênio/química , Zinco/química , Cátions Bivalentes , Dopamina/química , Radicais Livres/química , Concentração de Íons de Hidrogênio , Hidrólise , Isoproterenol/química , Norepinefrina/química , Oxirredução
10.
Nitric Oxide ; 16(4): 413-8, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17478115

RESUMO

Previously we established the hypotensive action of nitric oxide donors, dinitrosyl-iron complexes (DNIC) with thiol-containing ligands, stored in frozen solution at 77K. In the present study, we tested recently designed water soluble dry powder preparations of DNICs keeping their characteristics in dry air for a long time. The complexes dissolved in PBS were injected intravenously into normotensive Wistar and spontaneously hypertensive SHR rats. The average arterial pressure (AAP) was recorded through preliminary implanted catheter in a carotid artery. The initial hypotensive action of DNIC with cysteine (DNIC-cys) was comparable to action of nitroprusside (SNP) but, in contrast to the latter, lasted for 20-120min depending on a doze. The blood DNIC content as detected by electronic paramagnetic resonance steadily decreased at this time. The hypotensive action of S-nitrosocysteine was similar to SNP while binding of iron in DNIC by batophenantroline-disulphonate prevented its hypotensive effect. These data suggest that long-lasting hypotensive action of DNICs may be caused by stable protein-bound DNICs forming in the process of transfer of Fe(+)(NO(+))(2) moieties from low-molecular DNICs to thiol protein ligands. The relative initial dose-dependent effect of DNIC-cys was similar in Wistar and SHR but secondary AAP reduction was more profound in SHR. A substitution of cysteine in DNIC by thiosulphate resulted in markedly less initial AAP reduction while long-lasting effect was similar and substitution by glutathione smoothed initial AAP decline and stabilized AAP level in the second phase. Prolonged AAP reduction induced by DNIC-cys was considerably shortened in narcotized rats. Thus, dry preparations of DNICs preserve prolonged hypotensive activity.


Assuntos
Anti-Hipertensivos/farmacologia , Hipertensão/tratamento farmacológico , Hipotensão , Ferro/farmacologia , Óxidos de Nitrogênio/farmacologia , Compostos de Sulfidrila , Animais , Anti-Hipertensivos/química , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Estabilidade de Medicamentos , Hipertensão/fisiopatologia , Hipotensão/induzido quimicamente , Ferro/química , Ligantes , Masculino , Doadores de Óxido Nítrico/química , Doadores de Óxido Nítrico/farmacologia , Óxidos de Nitrogênio/química , Ratos , Ratos Endogâmicos SHR , Ratos Wistar , Vigília , Água/química
11.
Nitric Oxide ; 16(2): 286-93, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17088088

RESUMO

The formation of protein-bound dinitrosyl-iron complexes (DNIC) in blood plasma and packed red cell fraction has been demonstrated by the EPR method in the experiments on rabbits which were i/v injected with the low-molecular DNIC with thiosulphate. This formation was ensured by transfer of Fe(+)(NO(+))(2) moieties from low-molecular DNIC onto serum albumin or hemoglobin molecules. Protein-bound DNICs appeared immediately after low-molecular DNIC injection followed with gradually decreasing their amounts. The complexes could be detected by EPR technique during more than two days. The addition of water-soluble NO scavenger, the iron complex with N-methyl-d-glucamine dithiocarbamate (MGD) resulted in decomposition of a part of protein-bound DNICs and in effective excretion of secondary products (mainly mononitrosyl-iron complexes with MGD) from the blood flow.


Assuntos
Proteínas Sanguíneas/metabolismo , Ferro/sangue , Óxidos de Nitrogênio/sangue , Animais , Espectroscopia de Ressonância de Spin Eletrônica , Feminino , Ferro/química , Óxidos de Nitrogênio/química , Coelhos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA