Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
2.
J Clin Invest ; 98(3): 650-61, 1996 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-8698856

RESUMO

Anticancer therapy with doxorubicin (DOX) and other quinone anthracyclines is limited by severe cardiotoxicity, reportedly because semiquinone metabolites delocalize Fe(II) from ferritin and generate hydrogen peroxide, thereby promoting hydroxyl radical formation and lipid peroxidation. Cardioprotective interventions with antioxidants or chelators have nevertheless produced conflicting results. To investigate the role and mechanism(s) of cardiac lipid peroxidation in a clinical setting, we measured lipid conjugated dienes (CD) and hydroperoxides in blood plasma samples from the coronary sinus and femoral artery of nine cancer patients undergoing intravenous treatments with DOX. Before treatment, CD were unexpectedly higher in coronary sinus than in femoral artery (342 +/- 131 vs 112 +/- 44 nmol/ml, mean +/- SD; P < 0.01), showing that cardiac tissues were spontaneously involved in lipid peroxidation. This was not observed in ten patients undergoing cardiac catheterization for the diagnosis of arrhythmias or valvular dysfunctions, indicating that myocardial lipid peroxidation was specifically increased by the presence of cancer. The infusion of a standard dose of 60 mg DOX/m(2) rapidly ( approximately 5 min) abolished the difference in CD levels between coronary sinus and femoral artery (134 +/- 95 vs 112 +/- 37 nmol/ml); moreover, dose fractionation studies showed that cardiac release of CD and hydroperoxides decreased by approximately 80% in response to the infusion of as little as 13 mg DOX/m(2). Thus, DOX appeared to inhibit cardiac lipid peroxidation in a rather potent manner. Corollary in vitro experiments were performed using myocardial biopsies from patients undergoing aortocoronary bypass grafting. These experiments suggested that the spontaneous exacerbation of lipid peroxidation probably involved preexisting Fe(II) complexes, which could not be sequestered adequately by cardiac isoferritins and became redox inactive when hydrogen peroxide was included to simulate DOX metabolism and hydroxyl radical formation. Collectively, these in vitro and in vivo studies provide novel evidence for a possible inhibition of cardiac lipid peroxidation in DOX-treated patients. Other processes might therefore contribute to the cardiotoxicity of DOX.


Assuntos
Antibióticos Antineoplásicos/efeitos adversos , Doxorrubicina/efeitos adversos , Coração/efeitos dos fármacos , Peroxidação de Lipídeos/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Difosfato de Adenosina/farmacologia , Adulto , Idoso , Relação Dose-Resposta a Droga , Feminino , Humanos , Ferro/metabolismo , Ferro/farmacologia , Masculino , Pessoa de Meia-Idade , Miocárdio/metabolismo , Neoplasias/metabolismo
3.
J Clin Invest ; 95(4): 1595-605, 1995 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-7706466

RESUMO

The cardiotoxicity of doxorubicin (DOX) and other quinone-containing antitumor anthracyclines has been tentatively attributed to the formation of drug semiquinones which generate superoxide anion and reduce ferritin-bound Fe(III), favoring the release of Fe(II) and its subsequent involvement in free radical reactions. In the present study NADPH- and DOX-supplemented cytosolic fractions from human myocardial biopsies are shown to support a two-step reaction favoring an alternative mechanism of Fe(II) mobilization. The first step is an enzymatic two-electron reduction of the C-13 carbonyl group in the side chain of DOX, yielding a secondary alcohol metabolite which is called doxorubicinol (3.9 +/- 0.4 nmoles/mg protein per 4 h, mean +/- SEM). The second step is a nonenzymatic and superoxide anion-independent redox coupling of a large fraction of doxorubicinol (3.2 +/- 0.4 nmol/mg protein per 4 h) with Fe(III)-binding proteins distinct from ferritin, regenerating stoichiometric amounts of DOX, and mobilizing a twofold excess of Fe(II) ions (6.1 +/- 0.7 nmol/mg protein per 4 h). The formation of secondary alcohol metabolites decreases significantly (Pi < 0.01) when DOX is replaced by less cardiotoxic anthracyclines such as daunorubicin, 4'-epi DOX, and 4-demethoxy daunorubicin (2.1 +/- 0.1, 1.2 +/- 0.2, and 0.6 +/- 0.2 nmol/mg protein per 4 h, respectively). Therefore, daunorubicin, 4'-epi DOX, and 4-demethoxy daunorubicin are significantly (P < 0.01) less effective than DOX in mobilizing Fe(II) (3.5 +/- 0.1, 1.8 +/- 0.2, and 0.9 +/- 0.3 nmol/mg protein per 4 h, respectively). These results highlight the formation of secondary alcohol metabolites and the availability of nonferritin sources of Fe(III) as novel and critical determinants of Fe(II) delocalization and cardiac damage by structurally distinct anthracyclines, thus providing alternative routes to the design of cardioprotectants for anthracycline-treated patients.


Assuntos
Antibióticos Antineoplásicos/metabolismo , Doxorrubicina/análogos & derivados , Doxorrubicina/metabolismo , Ferro/metabolismo , Miocárdio/metabolismo , Álcoois/metabolismo , Antibióticos Antineoplásicos/farmacologia , Biópsia , Citosol/metabolismo , Doxorrubicina/farmacologia , Feminino , Ferritinas/análise , Átrios do Coração , Humanos , Masculino , Pessoa de Meia-Idade , Miocárdio/citologia , Miocárdio/enzimologia , NADP/metabolismo , Oxirredução , Frações Subcelulares
4.
Cancer Res ; 61(23): 8422-8, 2001 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-11731422

RESUMO

Changes in iron homeostasis have been implicated in cardiotoxicity induced by the anticancer anthracycline doxorubicin (DOX). Certain products of DOX metabolism, like the secondary alcohol doxorubicinol (DOXol) or reactive oxygen species (ROS), may contribute to cardiotoxicity by inactivating iron regulatory proteins (IRP) that modulate the fate of mRNAs for transferrin receptor and ferritin. It is important to know whether DOXol and ROS act by independent or combined mechanisms. Therefore, we monitored IRP activities in H9c2 rat embryo cardiomyocytes exposed to DOX or to analogues which were selected to achieve a higher formation of secondary alcohol metabolite (daunorubicin), a concomitant increase of alcohol metabolite and decrease of ROS (5-iminodaunorubicin), or a defective conversion to alcohol metabolite (mitoxantrone). On the basis of such multiple comparisons, we characterized that DOXol was able to remove iron from the catalytic Fe-S cluster of cytoplasmic aconitase, making this enzyme switch to the cluster-free IRP-1. ROS were not involved in this step, but they converted the IRP-1 produced by DOXol into a null protein which did not bind to mRNA, nor was it able to switch back to aconitase. DOX was also shown to inactivate IRP-2, which does not assemble or disassemble a Fe-S cluster. Comparisons between DOX and the analogues revealed that IRP-2 was inactivated only by ROS. Thus, DOX can inactivate both IRP through a sequential action of DOXol and ROS on IRP-1 or an independent action of ROS on IRP-2. This information serves guidelines for designing anthracyclines that spare iron homeostasis and induce less severe cardiotoxicity.


Assuntos
Antibióticos Antineoplásicos/toxicidade , Doxorrubicina/toxicidade , Cardiopatias/induzido quimicamente , Coração/efeitos dos fármacos , Proteínas Ferro-Enxofre/antagonistas & inibidores , Proteínas de Ligação a RNA/antagonistas & inibidores , Animais , Antibióticos Antineoplásicos/metabolismo , Células Cultivadas , Doxorrubicina/metabolismo , Cardiopatias/metabolismo , Proteína 1 Reguladora do Ferro , Proteína 2 Reguladora do Ferro , Proteínas Reguladoras de Ferro , Miocárdio/citologia , Miocárdio/metabolismo , Ratos , Espécies Reativas de Oxigênio/metabolismo
5.
Biochim Biophys Acta ; 876(2): 220-5, 1986 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-3955061

RESUMO

Lipid peroxidation of microsomes from rat liver and Morris hepatoma 9618A was induced by means of tert-butyl hydroperoxide (t-BuOOH). In rat liver microsomes t-BuOOH stimulated an early formation of lipid hydroperoxides (LOOH) and an increasing accumulation of malondialdehyde; t-BuOOH was completely consumed and cytochrome P-450 was rapidly destroyed. In hepatoma microsomes (60% deficiency of cytochrome P-450) a remarkable inhibition of both malondialdehyde and LOOH was observed; t-BuOOH was consumed only partially and cytochrome P-450 was destroyed slowly. In the presence of aminopyrine, malondialdehyde production was inhibited to the same extent (about 70%) in normal and tumour microsomes. The concentration of t-BuOOH required to achieve half-maximal velocity of malondialdehyde accumulation was comparable in the two microsome types. It is proposed that the deficiency of cytochrome P-450 limits the activation of t-BuOOH to the free radical species which initiate lipid peroxidation. Low cytochrome P-450 content would also affect the LOOH-dependent propagation of lipid peroxidation.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Peróxidos Lipídicos/metabolismo , Neoplasias Hepáticas Experimentais/metabolismo , Microssomos Hepáticos/metabolismo , Microssomos/metabolismo , Peróxidos/farmacologia , Animais , Linhagem Celular , Cinética , Microssomos/efeitos dos fármacos , Ratos , Ratos Endogâmicos ACI , Ratos Endogâmicos BUF , Vitamina E/metabolismo , terc-Butil Hidroperóxido
6.
Clin Cancer Res ; 7(6): 1511-5, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11410484

RESUMO

Doxorubicin cardiotoxicity is a multifactorial process in which the alcohol metabolite doxorubicinol mediates the transition from reversible to irreversible damage. We investigated whether the tubulin-active taxane paclitaxel increases conversion of doxorubicin to doxorubicinol, thus explaining the high incidence of congestive heart failure when doxorubicin is used with paclitaxel. Specimens of human myocardium from patients undergoing bypass surgery were processed to obtain cytosolic fractions in which doxorubicin was converted to doxorubicinol by NADPH-dependent aldo/keto or carbonyl reductases. In this model, clinically relevant concentrations of paclitaxel (1-2.5 microM) increased doxorubicinol formation by mechanisms consistent with allosteric modulation of the reductases. Stimulation was observed over a broad range of basal enzymatic activity, and was accompanied by a similar pattern of enhanced formation of doxorubicinol aglycone, a metabolite potentially involved in the reversible phase of cardiotoxicity. The closely related analogue docetaxel had effects similar to paclitaxel, but increased doxorubicinol formation over a narrower range of enzymatic activity. The unrelated tubulin-active alkaloid vinorelbine had no effect. These results demonstrate that taxanes have a unique potential for enhancing doxorubicin metabolism to toxic species in human myocardium. The effects on doxorubicinol formation provide clues to explain the clinical pattern of doxorubicin-paclitaxel cardiotoxicity and also caution against the potential toxicity of combining docetaxel with high cumulative doses of doxorubicin.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Doxorrubicina/administração & dosagem , Doxorrubicina/metabolismo , Coração/efeitos dos fármacos , Miocárdio/metabolismo , Paclitaxel/administração & dosagem , Taxoides , Antineoplásicos/farmacologia , Antineoplásicos Fitogênicos/farmacologia , Hidrocarbonetos Aromáticos com Pontes/farmacologia , Docetaxel , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Humanos , Paclitaxel/análogos & derivados , Tubulina (Proteína)/metabolismo , Vimblastina/análogos & derivados , Vimblastina/farmacologia , Vinorelbina
7.
Free Radic Biol Med ; 3(6): 379-87, 1987.
Artigo em Inglês | MEDLINE | ID: mdl-3123331

RESUMO

Chelation by citrate was found to promote the autoxidation of Fe2+, measured as the disappearance of 1,10-phenanthroline-chelatable Fe2+. The autoxidation of citrate-Fe2+ could in turn promote the peroxidation of microsomal phospholipid liposomes, as judged by malondialdehyde formation. At low citrate-Fe2+ ratios the autoxidation of Fe2+ was slow and the formation of malondialdehyde was preceded by a lag phase. The lag phase was eliminated by increasing the citrate-Fe2+ ratio, which also increased the rate of Fe2+ autoxidation. The Fe2+ autoxidation product required for the initiation of lipid peroxidation was characterized as being Fe3+. As direct evidence of this, linear initial rates of lipid peroxidation were obtained via the combination of citrate-Fe2+ and citrate-Fe3+, optimum activity occurring at a Fe3+-Fe2+ ratio of 1:1. Evidence is also presented to suggest that the superoxide and the hydrogen peroxide that are formed during the autoxidation of citrate-Fe2+ can either stimulate or inhibit lipid peroxidation by affecting the yield of citrate-Fe3+ from citrate-Fe2+. No evidence was obtained for the participation of the hydroxyl radical in the initiation of lipid peroxidation by citrate-Fe2+.


Assuntos
Citratos/metabolismo , Ferro/metabolismo , Peróxidos Lipídicos/metabolismo , Animais , Catalase/farmacologia , Compostos Ferrosos/metabolismo , Técnicas In Vitro , Metabolismo dos Lipídeos , Lipossomos/metabolismo , Masculino , Manitol/farmacologia , Microssomos Hepáticos/metabolismo , Oxirredução , Ratos , Ratos Endogâmicos , Superóxido Dismutase/farmacologia , Xantina Oxidase/farmacologia
8.
FEBS Lett ; 209(2): 305-10, 1986 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-3792550

RESUMO

Microsomal membranes from the slow-growing Morris hepatoma 9618A catalyze, in the presence of t-butyl hydroperoxide, lower rates of lipid peroxidation than rat liver microsomes. The cytochrome P-450 content of hepatoma microsomes is about 40% that of the liver. SKF 525-A, an inhibitor of mixed-function oxidase, produces in hepatoma microsomes a P-450 type I binding spectrum similar to that of hepatic microsomes. The concentration of the inhibitor required for half-maximal spectral change is about 2 microM in both microsome types. SKF 525-A or ethylmorphine inhibit lipid peroxidation of normal and tumor microsomes to the same extent (about 60%). Treatment of the tumor-bearing rats with 3-methylcholanthrene increases the hepatoma cytochrome P-450 to values comparable to those of control membranes, although the hemoprotein has a peak in the CO-reduced difference absorption spectrum at 448 nm. The cytochrome P-448 induction is accompanied by an almost complete restoration of the hydroperoxide-dependent lipid peroxidation.


Assuntos
Citocromos/metabolismo , Peróxido de Hidrogênio/metabolismo , Peróxidos Lipídicos/metabolismo , Neoplasias Hepáticas Experimentais/metabolismo , Metilcolantreno/farmacologia , Microssomos Hepáticos/metabolismo , Microssomos/metabolismo , Animais , Citocromo P-450 CYP1A2 , Etilmorfina/farmacologia , Cinética , Microssomos/efeitos dos fármacos , Microssomos Hepáticos/efeitos dos fármacos , Proadifeno/farmacologia , Progesterona/farmacologia , Ratos , Ratos Endogâmicos BUF
9.
FEBS Lett ; 198(2): 301-6, 1986 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-3956738

RESUMO

Microsomal membranes from rat liver and from the fast-growing Morris hepatoma 3942A have been peroxidized to different extents and the order parameter of the membranes measured by fluorescence depolarization of the probe 1,6-diphenyl-1,3,5-hexatriene. The data have been analysed by applying a mathematical approach that takes into account simultaneously static and dynamic fluorescence parameters. It appears that tumour membranes are more ordered than the control and their order parameter does not increase with greater exposure to the action of O2 radicals in contrast to liver membranes. The fatty acid composition of the membrane lipids has been studied under different experimental conditions and correlated to the behaviour of the physical parameter.


Assuntos
Peróxidos Lipídicos/metabolismo , Neoplasias Hepáticas Experimentais/metabolismo , Microssomos Hepáticos/metabolismo , Oxigênio/metabolismo , Animais , Difenilexatrieno , Ácidos Graxos/análise , Fluorescência , Bicamadas Lipídicas/metabolismo , Masculino , Lipídeos de Membrana/análise , Ratos , Temperatura
10.
Eur J Cancer ; 40(4): 563-70, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14962724

RESUMO

The aim of this study was to define the maximum tolerated dose (MTD) and the pharmacological profile of the paclitaxel analogue BMS-184476 given once every 3 weeks, or on days 1 and 8 every 3 weeks (d1&8), in combination with a fixed dose of 50 mg/m(2) of Doxorubicin (Doxo) administered on day 1 of a 21-day cycle. Adult patients with advanced solid malignancies received escalating doses of BMS-184476 infused over 1 h after bolus Doxo. Pharmacokinetics (PK) of BMS-184476, Doxo and metabolites were investigated. The effect of BMS-184476 on doxorubicinol formation was studied in the cytosol from human myocardium. The MTD of 3-weekly BMS-184476 was 30 mg/m(2). The MTD/recommended Phase II dose was 35 mg/m(2)/week (70 mg/m(2) per cycle) in the d1&8 schedule. The dose-limiting toxicity was neutropenia for both schedules. Other toxicities were loss of appetite, asthenia, and mild, cumulative peripheral neuropathy. The objective response rate in 17 previously untreated or minimally pretreated patients with breast cancer treated at 35 mg/m(2)/week of BMS-184476 was 59% (95% Confidence Interval (CI): 33-82%). Two of the 7 patients not responding to the study regimen later responded to Doxo and paclitaxel. Plasma disposition of BMS-184476 at 30, 35 and 40 mg/m(2) was linear without evidence of a PK interaction with Doxo. In studies with cytosol from human myocardium, the formation of cardiotoxic doxorubicinol was not enhanced by BMS-184476. Dosing of BMS-184476 for 2 consecutive weeks allowed the administration of larger doses of the taxane with a promising antitumour activity in patients with untreated or minimally pretreated breast cancer. The higher than expected myelotoxicity of the 3-weekly schedule is unexplained by the investigated interactions. Lack of enhanced doxorubicinol formation in human myocardium is consistent with the cardiac safety of the regimen.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Neoplasias/tratamento farmacológico , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Doxorrubicina/administração & dosagem , Doxorrubicina/efeitos adversos , Doxorrubicina/farmacocinética , Coração/efeitos dos fármacos , Humanos , Dose Máxima Tolerável , Pessoa de Meia-Idade , Miocárdio , Taxoides/administração & dosagem , Taxoides/efeitos adversos , Taxoides/farmacocinética
11.
Br J Pharmacol ; 134(6): 1271-8, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11704647

RESUMO

1. The anticancer anthracycline doxorubicin (DOX) causes cardiotoxicity. Enzymatic reduction of a side chain carbonyl group converts DOX to a secondary alcohol metabolite that has been implicated in cardiotoxicity. We therefore monitored negative inotropism, assessed as inhibition of post-rest contractions, in rat right ventricle strips exposed to DOX or to analogues forming fewer amounts of their alcohol metabolites (epirubicin, EPI, and the novel disaccharide anthracycline MEN 10755). 2. Thirty microM EPI exhibited higher uptake than equimolar DOX, but formed comparable amounts of alcohol metabolite due to its resistance to carbonyl reduction. MEN 10755 exhibited also an impaired uptake, and consequently formed the lowest levels of alcohol metabolite. Accordingly, DOX and EPI inhibited post-rest contractions by approximately 40-50%, whereas MEN 10755 inhibited by approximately 6%. 3. One hundred microM EPI exhibited the same uptake as equimolar DOX, but formed approximately 50% less alcohol metabolite. One hundred microM MEN 10755 still exhibited the lowest uptake, forming approximately 60% less alcohol metabolite than EPI. Under these conditions DOX inhibited post-rest contractions by 88%. EPI and MEN 10755 were approximately 18% (P<0.05) or approximately 80% (P<0.001) less inhibitory than DOX, respectively. 4. The negative inotropism of 30-100 microM DOX, EPI, or MEN 10755 correlated with cellular levels of both alcohol metabolites (r=0.88, P<0.0001) and carbonyl anthracyclines (r=0.79, P<0.0001). Nonetheless, multiple comparisons showed that alcohol metabolites were approximately 20-40 times more effective than carbonyl anthracyclines in inhibiting contractility. The negative inotropism of MEN 10755 was therefore increased by chemical procedures, like side chain valeryl esterification, that facilitated its uptake and conversion to alcohol metabolite but not its retention in a carbonyl form. 5. These results demonstrate that secondary alcohol metabolites are important mediators of cardiotoxicity. A combination of reduced uptake and limited conversion to alcohol metabolite formation might therefore render MEN 10755 more cardiac tolerable than DOX and EPI.


Assuntos
Antraciclinas/farmacologia , Antineoplásicos/farmacologia , Dissacarídeos/farmacologia , Doxorrubicina/análogos & derivados , Doxorrubicina/farmacologia , Contração Miocárdica/efeitos dos fármacos , Álcoois/metabolismo , Animais , Antraciclinas/química , Antineoplásicos/química , Dissacarídeos/química , Relação Dose-Resposta a Droga , Doxorrubicina/química , Coração/efeitos dos fármacos , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/metabolismo , Técnicas In Vitro , Masculino , Ratos , Ratos Sprague-Dawley , Função Ventricular
12.
Brain Res Mol Brain Res ; 50(1-2): 267-76, 1997 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-9406943

RESUMO

Heme oxygenase (HO)-catalyzed degradation of cellular heme moieties generates biliverdin and equimolar amounts of carbon monoxide (CO), which has been implicated as a possible modulator of neural function. Technical difficulties preclude direct measurements of CO within intact nervous tissues; hence, alternative procedures are needed to monitor the formation and possible biologic functions of this gas. In the present study rat hypothalamic explants were found to generate 114 +/- 5 or 127 +/- 11 pmol biliverdin/hypothalamus/1 h (n = 3) upon incubation with 1 or 10 microM hemin, respectively. Ten micromolar zinc-protoporphyrin IX (Zn-PP-IX), a known inhibitor of HO, significantly decreased the degradation of 10 microM hemin from 127 +/- 11 to 26 +/- 11 pmol biliverdin/hypothalamus/1 h (n = 3; P < 0.01). Biliverdin was the principal product of HO-dependent heme degradation, as its possible conversion into bilirubin was precluded by hemin-dependent inhibition of biliverdin reductase. Basal or hemin-supplemented hypothalamic incubations were also shown to generate sizable amounts of propentdyopents (PDPs), reflecting HO-independent degradation pathways which do not liberate CO and cannot be inhibited by Zn-PP-IX. Plotting the ratio of biliverdin to PDPs thus provided an index of the efficiency with which hemin was degraded through biochemical pathways involving CO. Under the experimental conditions of our study, the biliverdin/PDPs ratio varied from 0 to 32 or 15%, depending on the absence or presence of 1 or 10 microM hemin respectively: this suggested that the formation of CO was most efficient at 1 microM hemin. Under these defined conditions, 1 microM hemin was also found to inhibit the release of arginine vasopressin (AVP) evoked by depolarizing solutions of KCl. A series of experiments showed that the effect of hemin was counteracted by Zn-PP-IX, and also by tin-mesoporphyrin IX, which is even more selective in inhibiting HO; it was also attenuated in the presence of the gaseous scavenger ferrous hemoglobin. Furthermore, the inhibition of AVP release could be reproduced by omitting hemin and by incubating hypothalami under CO, whereas treatment with biliverdin had no effect. This suggested that the release of AVP was suppressed by HO degradation of hemin, yielding CO as a modulator of hypothalamic function. These observations may be relevant to diseases characterized by inappropriate secretion of AVP and enzymatic disturbances affecting the synthesis of heme and the formation of CO through the HO pathway (e.g., acute intermittent porphyria or lead intoxication).


Assuntos
Arginina Vasopressina/metabolismo , Monóxido de Carbono/metabolismo , Heme Oxigenase (Desciclizante)/metabolismo , Heme/metabolismo , Hipotálamo/metabolismo , Sistemas Neurossecretores/fisiologia , Oxirredutases atuantes sobre Doadores de Grupo CH-CH , Animais , Bilirrubina/biossíntese , Bilirrubina/farmacologia , Biliverdina/biossíntese , Ativação Enzimática , Hemina/farmacologia , Técnicas In Vitro , Masculino , Oxirredutases/metabolismo , Ratos , Ratos Wistar
13.
Ann N Y Acad Sci ; 488: 468-80, 1986.
Artigo em Inglês | MEDLINE | ID: mdl-3555261

RESUMO

Membranes isolated from tumor cells present profound alterations in their composition, structural organization, and functional properties. In this study we have reported some of these alterations in microsomal and plasma membranes of hepatomas with different growth rate and degree of differentiation. The chemical parameters studied were the phospholipid-to-protein, the cholesterol-to-protein, and the cholesterol-to-phospholipid ratios and the fatty acid composition of the phospholipids. The physical parameters were the molecular order (static) and the fluidity (dynamic), determined, respectively, as the order parameter [P2] and the correlation time tau R of the fluorescent probe 1,6-diphenyl-1,3,5-hexatriene (DPH). The functional property investigated was the ability of the membranes to undergo superoxide-induced lipid peroxidation, determined as byproduct (malondialdehyde and lipid hydroperoxides) formation and as changes in the fatty acid acyl residues. Changes in the physical state of the membrane, induced by oxy radicals, were also monitored during lipid peroxidation. A study of the antioxidant activity of the tumor cell, in terms of oxy radical enzymatic defenses (superoxide dismutase, glutathione peroxidase and catalase) was also performed. The main results obtained are the following: hepatoma membranes possess a lower phospholipid content and a lower degree of fatty acid unsaturation; on the other hand, the cholesterol-to-phospholipid ratio is increased; the physical state appears characterized by an increased rigidity (increased molecular order of the lipids and decreased fluidity); the membrane peroxidizability is markedly depressed and its order parameter, in contrast to liver membranes, does not increase with exposure to the action of O2- radicals; and the oxy radical enzymatic defense mechanisms are decreased. All these alterations increase with increasing growth rate and dedifferentiation of the tumor. Considering all of the data, we are inclined to think that tumor membranes are altered structurally and functionally in part as the result of an oxy radical-induced damage that takes place in vivo under conditions of increased oxygen toxicity.


Assuntos
Neoplasias Hepáticas Experimentais/metabolismo , Oxigênio/metabolismo , Animais , Membrana Celular/metabolismo , Radicais Livres , Membranas Intracelulares/metabolismo , Peróxidos Lipídicos/metabolismo , Lipídeos de Membrana/metabolismo , Microssomos Hepáticos/metabolismo , Ratos
14.
Ann N Y Acad Sci ; 851: 179-86, 1998 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-9668619

RESUMO

Iron may be important in catalyzing excessive production of reactive oxygen species (ROS). Cellular iron homeostasis is regulated by iron regulatory proteins (IRPs), which bind to iron-responsive elements (IRE) of mRNAs for ferritin and transferrin receptor (TfR) modulating iron uptake and sequestration, respectively. Although iron is the main regulator of IRP activity, IRP is also influenced by other factors, including the redox state. Therefore, IRP might be sensitive to pathophysiological alterations of redox state caused by ROS. However, previous studies have produced diverging evidence on the effect of oxidative injury on IRP. Results obtained in an animal model close to a pathophysiological condition, such as ischemia reperfusion of the liver as well as in a cell-free system involving an enzymatic source of O2 and H2O2, indicate that IRP is downregulated by oxidative stress. In fact, IRP activity is inhibited at early times of post-ischemic reperfusion. Moreover, the concerted action of O2 and H2O2 produced by xanthine oxidase in a cell-free system caused a remarkable inhibition of IRP activity. IRP seems a direct target of ROS; in fact, in vivo inhibition can be prevented by the antioxidant N-acetylcysteine and by interleukin-1 receptor antagonist. In addition, modulation of iron levels of the cell-free assay did not affect the downregulation imposed by xanthine oxidase. Conceivably, downregulation of IRP activity by O2 and H2O2 may facilitate iron sequestration into ferritin, thus limiting the pro-oxidant challenge of iron.


Assuntos
Proteínas Ferro-Enxofre/metabolismo , Ferro/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas de Ligação a RNA/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Acetilcisteína/farmacologia , Animais , Regulação para Baixo/fisiologia , Ferritinas/metabolismo , Proteínas Reguladoras de Ferro , Isquemia/fisiopatologia , Fígado/fisiopatologia , Oxirredução , Estresse Oxidativo/fisiologia , Proteínas de Ligação a RNA/fisiologia , Ratos , Proteína Wnt2 , Xantina/metabolismo , Xantina Oxidase/metabolismo
15.
Chem Phys Lipids ; 44(2-4): 191-208, 1987.
Artigo em Inglês | MEDLINE | ID: mdl-2822270

RESUMO

Iron is required for the initiation of lipid peroxidation. Evidence is presented that lipid peroxidation requires both Fe3+ and Fe2+, perhaps with oxygen to form a Fe3+-dioxygen-Fe2+ complex. Other mechanisms of initiation, mostly involving the iron-catalyzed formation of hydroxyl radical, are described and discussed from both theoretical and experimental view points.


Assuntos
Ferro/metabolismo , Peróxidos Lipídicos/metabolismo , Peroxidases/metabolismo , Radicais Livres , Hidróxidos , Radical Hidroxila
16.
Chem Biol Interact ; 71(1): 1-19, 1989.
Artigo em Inglês | MEDLINE | ID: mdl-2550151

RESUMO

The role of iron in the peroxidation of polyunsaturated fatty acids is reviewed, especially with respect to the involvement of oxygen radicals. The hydroxyl radical can be generated by a superoxide-driven Haber-Weiss reaction or by Fenton's reaction; and the hydroxyl radical can initiate lipid peroxidation. However, lipid peroxidation is frequently insensitive to hydroxyl radical scavengers or superoxide dismutase. We propose that the hydroxyl radical may not be involved in the peroxidation of membrane lipids, but instead lipid peroxidation requires both Fe2+ and Fe3+. The inability of superoxide dismutase to affect lipid peroxidation can be explained by the fact that the direct reduction of iron can occur, exemplified by rat liver microsomal NADPH-dependent lipid peroxidation. Catalase can be stimulatory, inhibitory or without affect because H2O2 may oxidize some Fe2+ to form the required Fe3+, or, alternatively, excess H2O2 may inhibit by excessive oxidation of the Fe2+. In an analogous manner reductants can form the initiating complex by reduction of Fe3+, but complete reduction would inhibit lipid peroxidation. All of these redox reactions would be influenced by iron chelation.


Assuntos
Hidróxidos/metabolismo , Ferro/metabolismo , Peroxidação de Lipídeos , Superóxidos/metabolismo , Animais , Catalase/metabolismo , Radicais Livres , Radical Hidroxila
17.
J Investig Med ; 47(3): 151-60, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10198571

RESUMO

BACKGROUND: gamma-Glutamyl transpeptidase (gamma-GT) is found in serum and in the plasma membranes of virtually all cell types. Its physiologic role is to initiate the hydrolysis of extracellular glutathione (GSH), a tripeptide in which cysteine lies between alpha-glycine and gamma-glutamate residues. Cysteine and other thiol compounds are known to promote LDL oxidation by reducing Fe(III) to redox active Fe(II); therefore, we sought to determine whether similar reactions can be sustained by GSH and influenced by gamma-GT. METHODS: Fe(III) reduction and LDL oxidation were studied by monitoring the formation bathophenanthroline-chelatable Fe(II) and the accumulation of thiobarbituric acid-reactive substances, respectively. Human atheromatous tissues were examined by histochemical techniques for the presence of oxidized LDL and their colocalization with cells expressing gamma-GT activity. RESULTS: A series of experiments showed that the gamma-glutamate residue of GSH affected interactions of the juxtaposed cysteine thiol with iron, precluding Fe(III) reduction and hence LDL oxidation. Both processes increased remarkably after addition of purified gamma-GT, which acts by removing the gamma-glutamate residue. GSH-dependent LDL oxidation was similarly promoted by gamma-GT associated with the plasma membrane of human monoblastoid cells, and this process required iron traces that can be found in advanced or late stage atheromas. Collectively, these findings suggested a possible role for gamma-GT in the cellular processes of LDL oxidation and atherogenesis. Histochemical analyses confirmed that this may be the case, showing that gamma-GT activity is expressed by macrophage-derived foam cells within human atheromas, and that these cells colocalize with oxidized LDL. CONCLUSIONS: Biochemical and histochemical correlates indicate that gamma-GT can promote LDL oxidation by hydrolyzing GSH into more potent iron reductants. These findings may provide mechanistic clues to the epidemiologic evidence for a possible correlation between persistent elevation of gamma-GT and the risk of fatal reinfarction in patients with ischemic heart disease.


Assuntos
Arteriosclerose/metabolismo , Ferro/metabolismo , Lipoproteínas LDL/metabolismo , gama-Glutamiltransferase/metabolismo , Adulto , Idoso , Catalase/farmacologia , Quelantes/metabolismo , Células Espumosas/enzimologia , Células Espumosas/patologia , Glutationa/farmacologia , Humanos , Técnicas Imunoenzimáticas , Ferro/química , Peroxidação de Lipídeos/fisiologia , Lipoproteínas LDL/química , Masculino , Pessoa de Meia-Idade , Fenantrolinas/metabolismo , Superóxido Dismutase/metabolismo , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo , Células U937
18.
Lipids ; 27(3): 219-26, 1992 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-1326072

RESUMO

Mechanisms of iron-catalyzed lipid peroxidation depend on the presence or absence of preformed lipid hydroperoxides (LOOH). Preformed LOOH are decomposed by Fe(II) to highly reactive lipid alkoxyl radicals, which in turn promote the formation of new LOOH. However, in the absence of LOOH, both Fe2+ and Fe3+ must be available to initiate lipid peroxidation, with optimum activity occurring as the Fe2+/Fe3+ ratio approaches unity. The simultaneous availability of Fe2+ and Fe3+ can be achieved by oxidizing some Fe2+ with hydrogen peroxide or with chelators that favor autoxidation of Fe2+ by molecular oxygen. Alternatively, one can use Fe3+ and reductants like superoxide, ascorbate or thiols. In either case excess Fe2+ oxidation or Fe3+ reduction will inhibit lipid peroxidation by converting all the iron to the Fe3+ or Fe2+ form, respectively. Superoxide dismutase and catalase can affect lipid peroxidation by affecting iron reduction/oxidation and the formation of a (1:1) Fe2+/Fe3+ ratio. Hydroxyl radical scavengers can also increase or decrease lipid peroxidation by affecting the redox cycling of iron.


Assuntos
Ferro , Peroxidação de Lipídeos , Animais , Catalase/metabolismo , Radicais Livres , Hidróxidos , Radical Hidroxila , Ferro/farmacologia , Cinética , Peroxidação de Lipídeos/efeitos dos fármacos , Microssomos/metabolismo , Oxirredução
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa