Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Int J Mol Sci ; 24(6)2023 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-36982498

RESUMO

F1·Fo-ATP synthases/ATPases (F1·Fo) are molecular machines that couple either ATP synthesis from ADP and phosphate or ATP hydrolysis to the consumption or production of a transmembrane electrochemical gradient of protons. Currently, in view of the spread of drug-resistant disease-causing strains, there is an increasing interest in F1·Fo as new targets for antimicrobial drugs, in particular, anti-tuberculosis drugs, and inhibitors of these membrane proteins are being considered in this capacity. However, the specific drug search is hampered by the complex mechanism of regulation of F1·Fo in bacteria, in particular, in mycobacteria: the enzyme efficiently synthesizes ATP, but is not capable of ATP hydrolysis. In this review, we consider the current state of the problem of "unidirectional" F1·Fo catalysis found in a wide range of bacterial F1·Fo and enzymes from other organisms, the understanding of which will be useful for developing a strategy for the search for new drugs that selectively disrupt the energy production of bacterial cells.


Assuntos
Adenosina Trifosfatases , Proteínas de Membrana , Adenosina Trifosfatases/metabolismo , Proteínas de Membrana/metabolismo , Óxido Nítrico Sintase/metabolismo , Trifosfato de Adenosina/metabolismo , Catálise , ATPases Translocadoras de Prótons/metabolismo , Hidrólise
2.
Biochemistry (Mosc) ; 87(8): 742-751, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-36171655

RESUMO

Proton-translocating Fo×F1-ATPase/synthase that catalyzes synthesis and hydrolysis of ATP is commonly considered to be a reversibly functioning complex. We have previously shown that venturicidin, a specific Fo-directed inhibitor, blocks the synthesis and hydrolysis of ATP with a significant difference in the affinity [Zharova, T. V. and Vinogradov, A. D. (2017) Biochim. Biophys. Acta, 1858, 939-944]. In this paper, we have studied in detail inhibition of Fo×F1-ATPase/synthase by venturicidin in tightly coupled membranes of Paracoccus denitrificans under conditions of membrane potential generation. ATP hydrolysis was followed by the ATP-dependent succinate-supported NAD+ reduction (potential-dependent reverse electron transfer) catalyzed by the respiratory chain complex I. It has been demonstrated that membrane energization did not affect the affinity of Fo×F1-ATPase/synthase for venturicidin. The dependence of the residual ATP synthase activity on the concentration of venturicidin approximated a linear function, whereas the dependence of ATP hydrolysis was sigmoidal: at low inhibitor concentrations venturicidin strongly inhibited ATP synthesis without decrease in the rate of ATP hydrolysis. A model is proposed suggesting that ATP synthesis and ATP hydrolysis are catalyzed by two different forms of Fo×F1.


Assuntos
Paracoccus denitrificans , Trifosfato de Adenosina , Cinética , NAD , ATPases Translocadoras de Prótons/metabolismo , Prótons , Succinatos , Venturicidinas
3.
Biochim Biophys Acta Bioenerg ; 1858(2): 109-117, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27810396

RESUMO

Bovine heart mitochondrial respiratory complex II generates ROS, mostly as superoxide, at the rate of about 20% of that detected during simultaneous operation of complex I and II when oxidation of ubiquinol is prevented by myxothiazol. ROS generating activity at different fumarate/succinate concentrations ratio implies that an enzyme component with a midpoint potential 40mV more positive than that of fumarate/succinate couple is the donor for one-electron reduction of oxygen. This suggests that the iron-sulfur cluster(s) is(are) involved in superoxide formation. Complex II-mediated ROS production exhibits a maximum at low (about 50µM) succinate concentration and gradually declines to zero activity upon further increase of the substrate. This phenomenology is explained and kinetically modeled to suggest a ping-pong mechanism of ROS generating activity where only dicarboxylate free reduced enzyme is oxidized by oxygen. The succinate:quinone reductase activity catalyzed by purified succinate:ubiquinone reductase also exhibits a ping-pong mechanism where only dicarboxylate free enzyme is oxidized by added quinone. Together these data suggest long distance interaction between the succinate (fumarate) binding and ubiquinone (ubiquinol) reactive sites.


Assuntos
Complexo II de Transporte de Elétrons/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Ubiquinona/metabolismo , Animais , Bovinos , Complexo I de Transporte de Elétrons/metabolismo , Fumaratos/metabolismo , Cinética , Mitocôndrias Cardíacas/metabolismo , Complexos Multienzimáticos/metabolismo , Oxirredução , Ratos , Ácido Succínico/metabolismo , Superóxidos/metabolismo
4.
Biochim Biophys Acta ; 1857(7): 863-71, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26571336

RESUMO

Kinetic characteristics of the proton-pumping NADH:quinone reductases (respiratory complexes I) are reviewed. Unsolved problems of the redox-linked proton translocation activities are outlined. The parameters of complex I-mediated superoxide/hydrogen peroxide generation are summarized, and the physiological significance of mitochondrial ROS production is discussed. This article is part of a Special Issue entitled Respiratory complex I, edited by Volker Zickermann and Ulrich Brandt.


Assuntos
Complexo I de Transporte de Elétrons/química , Complexo I de Transporte de Elétrons/ultraestrutura , NAD/química , Bombas de Próton/química , Bombas de Próton/ultraestrutura , Espécies Reativas de Oxigênio/síntese química , Transporte de Elétrons , Ativação Enzimática , Modelos Químicos , Simulação de Dinâmica Molecular , NAD/ultraestrutura , Oxirredução , Conformação Proteica
5.
Biochim Biophys Acta ; 1827(3): 446-54, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23313413

RESUMO

Membrane-bound respiratory complex I in inside-out submitochondrial particles (SMP) catalyzes both superoxide and hydrogen peroxide formation in NADH- and/or succinate-supported reactions. At optimal NADH concentration (50µM), the complex I-mediated process results in a formation of two superoxide anions and H(2)O(2) as the reaction products in approximately 0.7 ratio. Almost the same ratio is found for purified complex I (0.6) and for the aerobic succinate-supported reverse electron transfer reaction. Superoxide production is depressed at high, more physiologically relevant NADH concentrations, whereas hydrogen peroxide formation is insensitive to the elevated level of NADH. The rates of H(2)O(2) formation at variable NAD(+)/NADH ratios satisfactorily fit the Nernst equation for a single reactive two-electron donor component equilibrated with ambient midpoint redox potential of -347mV (0.13 NAD(+)/NADH ratio, pH 8.0). Half-maximal superoxide production rate proceeds at significantly higher NAD(+)/NADH ratio (0.33). Guanidine strongly stimulates NADH-supported hydrogen peroxide and superoxide production at any NADH concentration and activates NADH:ferricyanide and inhibits NADH:hexaammineruthenium (III) reductase activities while showing no effects on NADH oxidase of SMP. In the low range of NADH concentration, superoxide production rate shows a simple hyperbolic dependence on NADH with apparent K(m)(NADH) of 0.5µM, whereas sigmoidal dependence of hydrogen peroxide production is seen with half-maximal rate at 25µM NADH. We interpret the data as to suggest that at least two sites participate in complex I-mediated ROS generation: FMNH(-) that produces hydrogen peroxide, and an iron-sulfur center (likely N-2) that produces superoxide anion.


Assuntos
Complexo I de Transporte de Elétrons/fisiologia , Peróxido de Hidrogênio/metabolismo , Mitocôndrias/metabolismo , Superóxidos/metabolismo , Guanidina/farmacologia , Espécies Reativas de Oxigênio/metabolismo
6.
Biochim Biophys Acta ; 1817(10): 1879-85, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22503830

RESUMO

The rates of NADH-supported superoxide/hydrogen peroxide production by membrane-bound bovine heart respiratory complex I, soluble pig heart dihydrolipoamide dehydrogenase (DLDH), and by accompanying operation of these enzymes in rat heart mitochondrial matrix were measured as a function of the pool of pyridine nucleotides and its redox state. Each of the activities showed nontrivial dependence on nucleotide pool concentration. The NAD(+)/NADH ratios required for their half maximal capacities were determined. About half of the total NADH-supported H(2)O(2) production by permeabilized mitochondria in the absence of stimulating ammonium could be accounted for by DLDH activity. The significance of the mitochondrial NADH-dependent hydrogen peroxide production under physiologically relevant conditions is discussed. This article is part of a Special Issue entitled: 17th European Bioenergetics Conference (EBEC 2012).


Assuntos
Di-Hidrolipoamida Desidrogenase/metabolismo , Complexo I de Transporte de Elétrons/metabolismo , Peróxido de Hidrogênio/metabolismo , Mitocôndrias Cardíacas/metabolismo , Proteínas Mitocondriais/metabolismo , Proteínas Musculares/metabolismo , NADP/metabolismo , NAD/metabolismo , Animais , Bovinos , Ratos , Suínos
7.
Biochim Biophys Acta Bioenerg ; 1864(2): 148963, 2023 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-36842539

RESUMO

NADH-OH, the specific inhibitor of NADH-binding site of the mammalian complex I, is shown to completely block FMN-dependent reactions of P. denitrificans enzyme in plasma membrane vesicles: NADH oxidation (in a competitive manner with Ki of 1 nM) as well as reduction of pyridine nucleotides, ferricyanide and oxygen in the reverse electron transfer. In contrast to these activities, the reverse electron transfer to hexaammineruthenium (III) catalyzed by plasma membrane vesicles is insensitive to NADH-OH. To explain these results, we hypothesize the existence of a non-FMN redox group of P. denitrificans complex I that is capable of reducing hexaammineruthenium (III), which is corroborated by the complex kinetics of NADH: hexaammineruthenium (III)-reductase activity, catalyzed by this enzyme. A new assay procedure for measuring succinate-driven reverse electron transfer catalyzed by P. denitrificans complex I to hexaammineruthenium (III) is proposed.


Assuntos
NAD , Paracoccus denitrificans , Animais , NAD/metabolismo , Ubiquinona/metabolismo , Prótons , Elétrons , Complexo I de Transporte de Elétrons/metabolismo , Membrana Celular/metabolismo , Mamíferos/metabolismo
8.
Free Radic Biol Med ; 196: 133-144, 2023 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-36649901

RESUMO

The balance between the mitochondrial respiratory chain activity and the cell's needs in ATP ensures optimal cellular function. Cytochrome c is an essential component of the electron transport chain (ETC), which regulates ETC activity, oxygen consumption, ATP synthesis and can initiate apoptosis. The impact of conformational changes in cytochrome c on its function is not understood for the lack of access to these changes in intact mitochondria. We have developed a novel sensor that uses unique properties of label-free surface-enhanced Raman spectroscopy (SERS) to identify conformational changes in heme of cytochrome c and to elucidate their role in functioning mitochondria. We have verified that molecule bond vibrations assessed by SERS are a reliable indicator of the heme conformation during changes in the inner mitochondrial membrane potential and ETC activity. We have demonstrated that cytochrome c heme reversibly switches between planar and ruffled conformations in response to the inner mitochondrial membrane potential (ΔΨ) and H+ concentration in the intermembrane space. This regulates the efficiency of the mitochondrial respiratory chain, thus, adjusting the mitochondrial respiration to the cell's consumption of ATP and the overall activity. We have found that under hypertensive conditions cytochrome c heme loses its sensitivity to ΔΨ that can affect the regulation of ETC activity. The ability of the proposed SERS-based sensor to track mitochondrial function opens broad perspectives in cell bioenergetics.


Assuntos
Citocromos c , Heme , Citocromos c/metabolismo , Heme/metabolismo , Potencial da Membrana Mitocondrial , Mitocôndrias/metabolismo , Trifosfato de Adenosina/metabolismo
9.
Biochim Biophys Acta Bioenerg ; 1863(7): 148594, 2022 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-35850263

RESUMO

6-Ketocholestanol (kCh) is known as a mitochondrial recoupler, i.e. it abolishes uncoupling of mitochondria by such potent agents as carbonyl cyanide m-chlorophenyl hydrazone (CCCP) and 3,5-di(tert-butyl)-4-hydroxybenzylidenemalononitril (SF6847) [Starkov et al., 1997]. Here, we report data on the kCh-induced inhibition of both NADH-oxidase and NADH-ubiquinone oxidoreductase activities of the respiratory complex I in bovine heart submitochondrial particles (SMP). Based on the absence of such inhibition with hexaammineruthenium (III) (HAR) as the complex I electron acceptor, the kCh effect could be associated with the ubiquinone-binding centre of this respiratory enzyme. In isolated rat liver mitochondria (RLM), kCh inhibited oxygen consumption with the glutamate/malate, substrates of NAD-linked dehydrogenases, while no inhibition of RLM respiration was observed with succinate, in agreement with the absence of the kCh effect on the succinate oxidase activity in SMP. Three kCh analogs (cholesterol, 6α-hydroxycholesterol, and 5α,6α-epoxycholesterol) exhibited no effect on the NADH oxidase activities in both SMP and RLM. Importantly, the kCh analogs were ineffective in the recoupling of RLM treated with CCCP or SF6847. Therefore, interaction of kCh with the complex I may be involved in the kCh-mediated mitochondrial recoupling.


Assuntos
Complexo I de Transporte de Elétrons , Mitocôndrias , Animais , Carbonil Cianeto m-Clorofenil Hidrazona/farmacologia , Bovinos , Cetocolesteróis/farmacologia , Ratos
10.
Biochim Biophys Acta ; 1797(6-7): 939-44, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20170624

RESUMO

Coupled rat heart mitochondria produce externally hydrogen peroxide at the rates which correspond to about 0.8 and 0.3% of the total oxygen consumption at State 4 with succinate and glutamate plus malate as the respiratory substrates, respectively. Stimulation of the respiratory activities by ADP (State 4-State 3 transition) decreases the succinate- and glutamate plus malate-supported H2O2 production 8- and 1.3-times, respectively. NH4+ strongly stimulates hydrogen peroxide formation with either substrate without any effect on State 4 and/or State 3 respiration. Rotenone-treated, alamethicin-permeabilized mitochondria catalyze NADH-supported H2O2 production at a rate about 10-fold higher than that seen in intact mitochondria under optimal (State 4 succinate-supported respiration in the presence of ammonium chloride) conditions. NADH-supported hydrogen peroxide production by the rotenone-treated mitochondria devoid of a permeability barrier for H2O2 diffusion by alamethicin treatment are only partially (approximately 50%) sensitive to the Complex I NADH binding site-specific inhibitor, NADH-OH. The residual activity is strongly (approximately 6-fold) stimulated by ammonium chloride. NAD+ inhibits both Complex I-mediated and ammonium-stimulated H2O2 production. In the absence of stimulatory ammonium about half of the total NADH-supported hydrogen peroxide production is catalyzed by Complex I. In the presence of ammonium about 90% of the total hydrogen peroxide production is catalyzed by matrix located, ammonium-dependent enzyme(s).


Assuntos
Peróxido de Hidrogênio/metabolismo , Mitocôndrias Cardíacas/metabolismo , Difosfato de Adenosina/farmacologia , Animais , Complexo I de Transporte de Elétrons/metabolismo , Ácido Glutâmico/metabolismo , Técnicas In Vitro , Malatos/metabolismo , Mitocôndrias Cardíacas/efeitos dos fármacos , Proteínas Mitocondriais/metabolismo , NAD/metabolismo , Oxirredução , Consumo de Oxigênio , Compostos de Amônio Quaternário/farmacologia , Ratos , Ácido Succínico/metabolismo
11.
Biochim Biophys Acta Bioenerg ; 1861(8): 148207, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32315625

RESUMO

Mitochondrial NADH:ubiquinone oxidoreductase (proton translocating respiratory complex I) serves several essential functions in cell metabolism: it maintains the intramitochondrial NADH/NAD+ ratio, contributes to generation of the proton-motive force, and participates in physiological and/or pathophysiological production of so-called reactive oxygen species. A characteristic feature of complex I is a slow, compared with its catalytic turnover, transformation to its inactive (deactivated) state, a phenomenon operationally called A/D transition. Here we report data on several extrinsic factors affecting deactivation as observed in coupled or uncoupled bovine heart submitochondrial particles. The time course of the strongly temperature-dependent deactivation deviates from first-order kinetics, and this deviation is abolished in the presence of an SH-group-specific reagent. The residual fraction of activity attained upon extensive deactivation shows the same kinetics of NADH oxidation as the fully active enzyme does. The rate of complex I deactivation is only slightly pH dependent within the range of 7.0-8.5 and significantly increases at higher pH. ATP∙(Mg) decreases the rate of complex I deactivation in coupled SMP, and this effect is abolished if the proton-motive force generating ATPase activity of Fo∙F1 is precluded. Taken together, the data show that an equilibrium between the A and D forms of complex I exists. Possible mechanistic aspects of the deactivation process are discussed.


Assuntos
Complexo I de Transporte de Elétrons/metabolismo , Mitocôndrias/enzimologia , Animais , Bovinos , Membrana Celular/metabolismo , Complexo I de Transporte de Elétrons/química , Ativação Enzimática , Cinética , Desdobramento de Proteína , Prótons
12.
PLoS One ; 15(12): e0244499, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33378414

RESUMO

The synthesis of a mitochondria-targeted derivative of the classical mitochondrial uncoupler carbonyl cyanide-m-chlorophenylhydrazone (CCCP) by alkoxy substitution of CCCP with n-decyl(triphenyl)phosphonium cation yielded mitoCCCP, which was able to inhibit the uncoupling action of CCCP, tyrphostin A9 and niclosamide on rat liver mitochondria, but not that of 2,4-dinitrophenol, at a concentration of 1-2 µM. MitoCCCP did not uncouple mitochondria by itself at these concentrations, although it exhibited uncoupling action at tens of micromolar concentrations. Thus, mitoCCCP appeared to be a more effective mitochondrial recoupler than 6-ketocholestanol. Both mitoCCCP and 6-ketocholestanol did not inhibit the protonophoric activity of CCCP in artificial bilayer lipid membranes, which might compromise the simple proton-shuttling mechanism of the uncoupling activity on mitochondria.


Assuntos
Carbonil Cianeto m-Clorofenil Hidrazona/farmacologia , Mitocôndrias Hepáticas/efeitos dos fármacos , Acoplamento Oxidativo/efeitos dos fármacos , Fosforilação Oxidativa/efeitos dos fármacos , Animais , Carbonil Cianeto m-Clorofenil Hidrazona/análogos & derivados , Bovinos , Cetocolesteróis/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Mitocôndrias Hepáticas/metabolismo , Ratos , Desacopladores/farmacologia
13.
FEBS Lett ; 582(18): 2719-24, 2008 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-18619961

RESUMO

NADH-supported generation of H2O2 by permeabilized rat heart mitochondria was partially prevented by the specific complex I-directed inhibitor, NADH-OH, and was significantly stimulated by ammonium. Ammonium did not affect H2O2 production by complex I in coupled submitochondrial particles. The soluble mitochondrial matrix protein fraction catalyzed NADH-dependent H2O2 production, which was greatly (approximately 10-fold) stimulated by ammonium. We conclude that complex I is not the major contributor to mitochondrial superoxide (hydrogen peroxide) generation and that there are specific ammonium-sensitive NADH:oxygen oxidoreductase(s) in the mitochondrial matrix which are responsible for mitochondrial H2O2 production.


Assuntos
Peróxido de Hidrogênio/metabolismo , Mitocôndrias Cardíacas/metabolismo , Compostos de Amônio Quaternário/metabolismo , Animais , Catálise , Bovinos , Complexo I de Transporte de Elétrons/metabolismo , Mitocôndrias Cardíacas/efeitos dos fármacos , NAD/metabolismo , Compostos de Amônio Quaternário/farmacologia , Ratos
14.
FEBS Lett ; 592(13): 2213-2219, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29851085

RESUMO

A simple assay procedure for measuring ATP-dependent reverse electron transfer from ubiquinol to hexaammineruthenium (III) (HAR) catalyzed by mitochondrial respiratory complex I is introduced. The specific activity of the enzyme in this reaction and its sensitivity to the standard inhibitors and uncoupling are the same as with other well-known electron acceptors, NAD+ and ferricyanide. In contrast to the reactions with these acceptors, the energy-dependent HAR reduction is not inhibited by NADH-OH, the specific inhibitor of NADH-binding site. These results suggest that a catalytically competent electron connection exists between HAR and a redox component of complex I that is different from flavin mononucleotide bound at the substrate-binding site.


Assuntos
Complexo I de Transporte de Elétrons/metabolismo , Transporte de Elétrons/fisiologia , Mononucleotídeo de Flavina/química , Mononucleotídeo de Flavina/metabolismo , Animais , Sítios de Ligação , Bovinos , Complexo I de Transporte de Elétrons/química , Elétrons , Transferência de Energia/fisiologia , Técnicas In Vitro , Mitocôndrias/metabolismo , Mitocôndrias Cardíacas/efeitos dos fármacos , Mitocôndrias Cardíacas/metabolismo , Oxirredução , Compostos de Rutênio/química , Compostos de Rutênio/farmacologia
15.
Redox Biol ; 17: 192-199, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29702406

RESUMO

The initial rates of superoxide plus hydrogen peroxide (ROS) generation by intact or permeabilized rat heart mitochondria and coupled inside-out bovine heart submitochondrial particles (SMP) oxidizing NAD-dependent substrates, NADH, and succinate were measured by detecting resorufin formation in the Amplex Red assay at various oxygen concentrations. Linear dependences of the initial rates on oxygen concentration within the range of ~125-750 µM were found for all significant mitochondrial generators, i.e. the respiratory complexes and ammonium-stimulated dihydrolipoamide dehydrogenase. At lower oxygen concentrations upon its decrease from air saturation level to zero, the time-course of resorufin formation by SMP catalyzing coupled oxidation of succinate (the total ROS production by respiratory complexes II and III and by the reverse electron transfer (RET)-mediated by complex I) also corresponds to the linear dependence on oxygen with the same first-order rate constant determined in the initial rate studies. Prolonged incubation of SMP generating succinate-supported complex I-mediated ROS affected neither their NADH oxidase nor ROS generating activity. In contrast to SMP significant deviation from the first-order oxygen dependence in the time-course kinetics during coupled oxidation of succinate by intact mitochondria was evident. Complex I catalyzes the NADH:resorufin oxidoreductase reaction resulting in formation of colorless reduced resorufin. Hydrogen peroxide oxidizes reduced resorufin in the presence of peroxidase, thus showing its dihydroresorufin peroxidase activity. Combined NADH:resorufin reductase and dihydroresorufin peroxidase activities result in underestimation of the amount of hydrogen peroxide generated by mitochondria. We conclude that only initial rates of the mitochondrial ROS production, not the amount of resorufin accumulated, should be taken as the reliable measure of the mitochondrial ROS-generating activity, because of the cycling of the oxidized and reduced resorufin during Amplex Red assays fed by NADH and other possible reductant(s) present in mitochondria.


Assuntos
Transporte de Elétrons/genética , Mitocôndrias Cardíacas/metabolismo , Oxazinas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Animais , Complexo I de Transporte de Elétrons/genética , Complexo I de Transporte de Elétrons/metabolismo , Complexo II de Transporte de Elétrons/genética , Complexo II de Transporte de Elétrons/metabolismo , Peróxido de Hidrogênio/química , NAD/metabolismo , Oxazinas/química , Oxazinas/isolamento & purificação , Oxirredução , Oxigênio/metabolismo , Ratos , Ácido Succínico/metabolismo , Superóxidos/química
16.
Biochim Biophys Acta ; 1757(5-6): 553-61, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16678117

RESUMO

Superoxide production by inside-out coupled bovine heart submitochondrial particles, respiring with succinate or NADH, was measured. The succinate-supported production was inhibited by rotenone and uncouplers, showing that most part of superoxide produced during succinate oxidation is originated from univalent oxygen reduction by Complex I. The rate of the superoxide (O2*-)) production during respiration at a high concentration of NADH (1 mM) was significantly lower than that with succinate. Moreover, the succinate-supported O2*- production was significantly decreased in the presence of 1 mM NADH. The titration curves, i.e., initial rates of superoxide production versus NADH concentration, were bell-shaped with the maximal rate (at 50 microM NADH) approaching that seen with succinate. Both NAD+ and acetyl-NAD+ inhibited the succinate-supported reaction with apparent Ki's close to their Km's in the Complex I-catalyzed succinate-dependent energy-linked NAD+ reduction (reverse electron transfer) and NADH:acetyl-NAD+ transhydrogenase reaction, respectively. We conclude that: (i) under the artificial experimental conditions the major part of superoxide produced by the respiratory chain is formed by some redox component of Complex I (most likely FMN in its reduced or free radical form); (ii) two different binding sites for NADH (F-site) and NAD+ (R-site) in Complex I provide accessibility of the substrates-nucleotides to the enzyme red-ox component(s); F-site operates as an entry for NADH oxidation, whereas R-site operates in the reverse electron transfer and univalent oxygen reduction; (iii) it is unlikely that under the physiological conditions (high concentrations of NADH and NAD+) Complex I is responsible for the mitochondrial superoxide generation. We propose that the specific NAD(P)H:oxygen superoxide (hydrogen peroxide) producing oxidoreductase(s) poised in equilibrium with NAD(P)H/NAD(P)+ couple should exist in the mitochondrial matrix, if mitochondria are, indeed, participate in ROS-controlled processes under physiologically relevant conditions.


Assuntos
Complexo I de Transporte de Elétrons/fisiologia , Mitocôndrias Cardíacas/metabolismo , Modelos Biológicos , Partículas Submitocôndricas/metabolismo , Superóxidos/metabolismo , Animais , Sítios de Ligação , Bovinos , Transporte de Elétrons , Peróxido de Hidrogênio/metabolismo , Mitocôndrias Cardíacas/efeitos dos fármacos , NAD/metabolismo , Oxirredução , Oxigênio/metabolismo , Rotenona/farmacologia , Partículas Submitocôndricas/efeitos dos fármacos , Ácido Succínico/metabolismo , Desacopladores/farmacologia
17.
FEBS Lett ; 581(30): 5803-6, 2007 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-18037377

RESUMO

Conditions for the reversible dissociation of flavin mononucleotide (FMN) from the membrane-bound mitochondrial NADH:ubiquinone oxidoreductase (complex I) are described. The catalytic activities of the enzyme, i.e. rotenone-insensitive NADH:hexaammineruthenium III reductase and rotenone-sensitive NADH:quinone reductase decline when bovine heart submitochondrial particles are incubated with NADH in the presence of rotenone or cyanide at alkaline pH. FMN protects and fully restores the NADH-induced inactivation whereas riboflavin and flavin adenine dinucleotide do not. The data show that the reduction of complex I significantly weakens the binding of FMN to protein thus resulting in its dissociation when the concentration of holoenzyme is comparable with K(d ( approximately 10(-8)M at pH 10.0).


Assuntos
Complexo I de Transporte de Elétrons/metabolismo , Mononucleotídeo de Flavina/metabolismo , Membranas Mitocondriais/enzimologia , Animais , Bovinos , Ativação Enzimática/efeitos dos fármacos , Mitocôndrias Cardíacas/efeitos dos fármacos , Mitocôndrias Cardíacas/enzimologia , Membranas Mitocondriais/efeitos dos fármacos , NAD/farmacologia , Rotenona/farmacologia , Partículas Submitocôndricas/efeitos dos fármacos , Partículas Submitocôndricas/enzimologia , Fatores de Tempo
18.
Biochem J ; 387(Pt 3): 677-83, 2005 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-15571492

RESUMO

Palmitate rapidly and reversibly inhibits the uncoupled NADH oxidase activity catalysed by activated complex I in inside-out bovine heart submitochondrial particles (IC50 extrapolated to zero enzyme concentration is equal to 9 microM at 25 degrees C, pH 8.0). The NADH:hexa-ammineruthenium reductase activity of complex I is insensitive to palmitate. Partial (approximately 50%) inhibition of the NADH:external quinone reductase activity is seen at saturating palmitate concentration and the residual activity is fully sensitive to piericidin. The uncoupled succinate oxidase activity is considerably less sensitive to palmitate. Only a slight stimulation of tightly coupled respiration with NADH as the substrate is seen at optimal palmitate concentrations, whereas complete relief of the respiratory control is observed with succinate as the substrate. Palmitate prevents the turnover-induced activation of the de-activated complex I (IC50 extrapolated to zero enzyme concentration is equal to 3 microM at 25 degrees C, pH 8.0). The mode of action of palmitate on the NADH oxidase is qualitatively temperature-dependent. Rapid and reversible inhibition of the complex I catalytic activity and its de-active to active state transition are seen at 25 degrees C, whereas the time-dependent irreversible inactivation of the NADH oxidase proceeds at 37 degrees C. Palmitate drastically increases the rate of spontaneous de-activation of complex I in the absence of NADH. Taken together, these results suggest that free fatty acids act as specific complex I-directed inhibitors; at a physiologically relevant temperature (37 degrees C), their inhibitory effects on mitochondrial NADH oxidation is due to perturbation of the pseudo-reversible active-de-active complex I transition.


Assuntos
Complexo I de Transporte de Elétrons/antagonistas & inibidores , Complexo I de Transporte de Elétrons/metabolismo , Ativação Enzimática , Mitocôndrias Cardíacas/enzimologia , Ácido Palmítico/metabolismo , Animais , Bovinos , Oxirredução , Ratos , Temperatura , Fatores de Tempo
19.
Biochim Biophys Acta ; 1607(2-3): 79-90, 2003 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-14670598

RESUMO

The catalytic properties of the rotenone-sensitive NADH:ubiquinone reductase (Complex I) in bovine heart submitochondrial particles and in inside-out vesicles derived from Paracoccus denitrificans and Rhodobacter capsulatus were compared. The prokaryotic enzymes catalyze the NADH oxidase and NADH:quinone reductase reactions with similar kinetic parameters as those for the mammalian Complex I, except for lower apparent affinities for the substrates--nucleotides. Unidirectional competitive inhibition of NADH oxidation by ADP-ribose, previously discovered for submitochondrial particles, was also evident for tightly coupled P. denitrificans vesicles, thus suggesting that a second, NAD(+)-specific site is present in the simpler prokaryotic enzyme. The inhibitor sensitivity of the forward and reverse electron transfer reactions was compared. In P. denitrificans and Bos taurus vesicles different sensitivities to rotenone and Triton X-100 for the forward and reverse electron transfer reactions were found. In bovine heart preparations, both reactions showed the same sensitivity to piericidin, and the inhibition was titrated as a straight line. In P. denitrificans, the forward and reverse reactions show different sensitivity to piericidin and the titrations of both activities were curvilinear with apparent I(50) (expressed as mole of inhibitor per mole of enzyme) independent of the enzyme concentration. This behavior is explained by a model involving two different sites rapidly interacting with piericidin within the hydrophobic phase.


Assuntos
Complexo I de Transporte de Elétrons/metabolismo , Mitocôndrias/enzimologia , Células Procarióticas/enzimologia , Quinona Redutases/metabolismo , Quinonas/metabolismo , Animais , Antibacterianos/farmacologia , Bovinos , Complexo I de Transporte de Elétrons/antagonistas & inibidores , Cinética , Miocárdio/enzimologia , Piridinas/farmacologia , Quinona Redutases/antagonistas & inibidores
20.
FEBS Lett ; 585(14): 2212-6, 2011 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-21624365

RESUMO

The rotenone-insensitive NADH:hexaammineruthenium III (HAR) oxidoreductase reactions catalyzed by bovine heart and Yarrowia lipolytica submitochondrial particles or purified bovine complex I are stimulated by ATP and other purine nucleotides. The soluble fraction of mammalian complex I (FP) and prokaryotic complex I homolog NDH-1 in Paracoccus denitrificans plasma membrane lack stimulation of their activities by ATP. The stimulation appears as a decrease in apparent K(m) values for NADH and HAR. Thus, the "accessory" subunits of eukaryotic complex I bear an allosteric ATP-binding site.


Assuntos
Regulação Alostérica , Complexo I de Transporte de Elétrons/química , Proteínas Fúngicas/química , Mitocôndrias/enzimologia , NADH NADPH Oxirredutases/química , Nucleotídeos/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Sítios de Ligação , Bovinos , Complexo I de Transporte de Elétrons/metabolismo , Proteínas Fúngicas/metabolismo , NAD/metabolismo , NADH NADPH Oxirredutases/metabolismo , Paracoccus denitrificans/citologia , Paracoccus denitrificans/metabolismo , Compostos de Rutênio/metabolismo , Yarrowia/citologia , Yarrowia/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA