Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Cancer ; 143(7): 1706-1719, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29672841

RESUMEN

Familial aggregation is a significant risk factor for the development of thyroid cancer and familial non-medullary thyroid cancer (FNMTC) accounts for 5-7% of all NMTC. Whole exome sequencing analysis in the family affected by FNMTC with oncocytic features where our group previously identified a predisposing locus on chromosome 19p13.2, revealed a novel heterozygous mutation (c.400G > A, NM_012335; p.Gly134Ser) in exon 5 of MYO1F, mapping to the linkage locus. In the thyroid FRTL-5 cell model stably expressing the mutant MYO1F p.Gly134Ser protein, we observed an altered mitochondrial network, with increased mitochondrial mass and a significant increase in both intracellular and extracellular reactive oxygen species, compared to cells expressing the wild-type (wt) protein or carrying the empty vector. The mutation conferred a significant advantage in colony formation, invasion and anchorage-independent growth. These data were corroborated by in vivo studies in zebrafish, since we demonstrated that the mutant MYO1F p.Gly134Ser, when overexpressed, can induce proliferation in whole vertebrate embryos, compared to the wt one. MYO1F screening in additional 192 FNMTC families identified another variant in exon 7, which leads to exon skipping, and is predicted to alter the ATP-binding domain in MYO1F. Our study identified for the first time a role for MYO1F in NMTC.


Asunto(s)
Proliferación Celular , Embrión no Mamífero/patología , Mitocondrias/patología , Mutación , Miosina Tipo I/genética , Cáncer Papilar Tiroideo/patología , Neoplasias de la Tiroides/patología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Apoptosis , Células Cultivadas , Niño , Cromosomas Humanos Par 19 , Embrión no Mamífero/metabolismo , Femenino , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Mitocondrias/genética , Mitocondrias/metabolismo , Miosina Tipo I/química , Miosina Tipo I/metabolismo , Consumo de Oxígeno , Linaje , Conformación Proteica , Cáncer Papilar Tiroideo/genética , Cáncer Papilar Tiroideo/metabolismo , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/metabolismo , Adulto Joven , Pez Cebra
2.
Biochim Biophys Acta ; 1857(7): 991-1000, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26820434

RESUMEN

This review discusses the functional properties of mitochondrial Complex I originating from its presence in an assembled form as a supercomplex comprising Complex III and Complex IV in stoichiometric ratios. In particular several lines of evidence are presented favouring the concept that electron transfer from Complex I to Complex III is operated by channelling of electrons through Coenzyme Q molecules bound to the supercomplex, in contrast with the hypothesis that the transfer of reducing equivalents from Complex I to Complex III occurs via random diffusion of the Coenzyme Q molecules in the lipid bilayer. Furthermore, another property provided by the supercomplex assembly is the control of generation of reactive oxygen species by Complex I. This article is part of a Special Issue entitled Respiratory Complex I, edited by Volker Zickermann and Ulrich Brandt.


Asunto(s)
Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/metabolismo , Mitocondrias/enzimología , Especies Reactivas de Oxígeno/síntesis química , Ubiquinona/química , Ubiquinona/metabolismo , Animales , Transporte de Electrón , Complejo I de Transporte de Electrón/ultraestructura , Activación Enzimática , Humanos , Modelos Químicos , Simulación de Dinámica Molecular , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Complejos Multiproteicos/ultraestructura , Oxidación-Reducción , Conformación Proteica , Bombas de Protones/química , Bombas de Protones/ultraestructura , Relación Estructura-Actividad , Ubiquinona/ultraestructura
3.
Biochim Biophys Acta ; 1837(4): 427-43, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24246637

RESUMEN

Recent experimental evidence has replaced the random diffusion model of electron transfer with a model of supramolecular organisation based upon specific interactions between individual respiratory complexes. These supercomplexes were found to be functionally relevant by flux control analysis and to confer a kinetic advantage to NAD-linked respiration (channelling). However, the Coenzyme Q pool is still required for FAD-linked oxidations and for the proper equilibrium with Coenzyme Q bound in the supercomplex. Channelling in the cytochrome c region probably also occurs but does not seem to confer a particular kinetic advantage. The supramolecular association of individual complexes strongly depends on membrane lipid amount and composition and is affected by lipid peroxidation; it also seems to be modulated by membrane potential and protein phosphorylation. Additional properties of supercomplexes are stabilisation of Complex I, as evidenced by the destabilising effect on Complex I of mutations in either Complex III or IV, and prevention of excessive generation of reactive oxygen species. The dynamic character of the supercomplexes allows their involvement in metabolic adaptations and in control of cellular signalling pathways. This article is part of a Special Issue entitled: Dynamic and ultrastructure of bioenergetic membranes and their components.


Asunto(s)
Respiración de la Célula , Mitocondrias/metabolismo , Membranas Mitocondriales/metabolismo , Complejos Multienzimáticos/metabolismo , Cinética , Lípidos de la Membrana/metabolismo , Modelos Biológicos , Oxidación-Reducción , Fosforilación Oxidativa , Ubiquinona/metabolismo
4.
Mitochondrion ; 74: 101822, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-38040170

RESUMEN

Over the past decades, models of the organization of mitochondrial respiratory system have been controversial. The goal of this perspective is to assess this "conflict of models" by focusing on specific kinetic evidence in the two distinct segments of Coenzyme Q- and Cytochrome c-mediated electron transfer. Respiratory supercomplexes provide kinetic advantage by allowing a restricted diffusion of Coenzyme Q and Cytochrome c, and short-range interaction with their partner enzymes. In particular, electron transfer from NADH is compartmentalized by channeling of Coenzyme Q within supercomplexes, whereas succinate oxidation proceeds separately using the free Coenzyme Q pool. Previous evidence favoring Coenzyme Q random diffusion in the NADH-dependent electron transfer is due to downstream flux interference and misinterpretation of results. Indeed, electron transfer by complexes III and IV via Cytochrome c is less strictly dependent on substrate channeling in mammalian mitochondria. We briefly describe these differences and their physiological implications.


Asunto(s)
Grupo Citocromo c , Proteínas del Complejo de Cadena de Transporte de Electrón , Mitocondrias , Ubiquinona , Ubiquinona/metabolismo , Grupo Citocromo c/metabolismo , Animales , Bovinos , Mamíferos/metabolismo , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Mitocondrias/metabolismo , Corazón/fisiología , Porcinos
5.
Biochim Biophys Acta ; 1817(2): 363-9, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22086148

RESUMEN

We have studied the effects of idebenone on mitochondrial function in cybrids derived from one normal donor (HQB17) and one patient harboring the G3460A/MT-ND1 mutation of Leber's Hereditary Optic Neuropathy (RJ206); and in XTC.UC1 cells bearing a premature stop codon at amino acid 101 of MT-ND1 that hampers complex I assembly. Addition of idebenone to HQB17 cells caused mitochondrial depolarization and NADH depletion, which were inhibited by cyclosporin (Cs) A and decylubiquinone, suggesting an involvement of the permeability transition pore (PTP). On the other hand, addition of dithiothreitol together with idebenone did not cause PTP opening and allowed maintenance of the mitochondrial membrane potential even in the presence of rotenone. Addition of dithiothreitol plus idebenone, or of idebenol, to HQB17, RJ206 and XTC.UC1 cells sustained membrane potential in intact cells and ATP synthesis in permeabilized cells even in the presence of rotenone and malonate, and restored a good level of coupled respiration in complex I-deficient XTC.UC1 cells. These findings demonstrate that idebenol can feed electrons at complex III. If the quinone is maintained in the reduced state, a task that in some cell types appears to be performed by dicoumarol-sensitive NAD(P)H:quinone oxidoreductase 1 [Haefeli et al. (2011) PLoS One 6, e17963], electron transfer to complex III may allow reoxidation of NADH in complex I deficiencies.


Asunto(s)
Metabolismo Energético/efectos de los fármacos , Mitocondrias Hepáticas/efectos de los fármacos , Mitocondrias Hepáticas/metabolismo , Ubiquinona/análogos & derivados , Adenosina Trifosfato/metabolismo , Animales , Antioxidantes/farmacología , Respiración de la Célula/efectos de los fármacos , Células Cultivadas , Ditiotreitol/farmacología , Evaluación Preclínica de Medicamentos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Mitocondrias Hepáticas/fisiología , Consumo de Oxígeno/efectos de los fármacos , Ubiquinona/farmacología
6.
Biol Chem ; 394(5): 631-9, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23449521

RESUMEN

Substantial evidence exists that the mitochondrial respiratory chain is organized in supramolecular units called supercomplexes or respirasomes. While the structural evidence of the supercomplexes is overwhelming, fewer studies have focused on their functional relevance. Although the presence of coenzyme Q channeling between complexes I and III has been ascertained, no such clear demonstration has been carried out for cytochrome c between complexes III and IV, at least in mammalian mitochondria. This review also discusses the implications concerning the number of respiratory complexes organized in supercomplexes and the possibility that they represent associations in dynamic equilibrium with the individual complexes.


Asunto(s)
Citocromos c/metabolismo , Mitocondrias/metabolismo , Ubiquinona/metabolismo , Respiración de la Célula/fisiología , Citocromos c/análisis , Transporte de Electrón , Humanos , Mitocondrias/química , Ubiquinona/análisis
7.
Biochim Biophys Acta Bioenerg ; 1864(3): 148977, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37059413

RESUMEN

We have investigated NADH and succinate aerobic oxidation in frozen and thawed swine heart mitochondria. Simultaneous oxidation of NADH and succinate showed complete additivity under a variety of experimental conditions, suggesting that the electron fluxes originating from NADH and succinate are completely independent and do not mix at the level of the so-called mobile diffusible components. We ascribe the results to mixing of the fluxes at the level of cytochrome c in bovine mitochondria: the Complex IV flux control coefficient in NADH oxidation was high in swine mitochondria but very low in bovine mitochondria, suggesting a stronger interaction of cytochrome c with the supercomplex in the former. This was not the case in succinate oxidation, in which Complex IV exerted little control also in swine mitochondria. We interpret the data in swine mitochondria as restriction of the NADH flux by channelling within the I-III2-IV supercomplex, whereas the flux from succinate shows pool mixing for both Coenzyme Q and probably cytochrome c. The difference between the two types of mitochondria may be ascribed to different lipid composition affecting the cytochrome c binding properties, as suggested by breaks in Arrhenius plots of Complex IV activity occurring at higher temperatures in bovine mitochondria.


Asunto(s)
Mitocondrias Cardíacas , Ácido Succínico , Animales , Bovinos , Porcinos , Mitocondrias Cardíacas/metabolismo , NAD/metabolismo , Citocromos c/metabolismo , Electrones , Succinatos/metabolismo , Complejo IV de Transporte de Electrones/metabolismo
8.
Adv Exp Med Biol ; 942: 93-136, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22399420

RESUMEN

Oxidative stress is among the major causes of toxicity due to interaction of Reactive Oxygen Species (ROS) with cellular macromolecules and structures and interference with signal transduction pathways. The mitochondrial respiratory chain, specially from Complexes I and III, is considered the main origin of ROS particularly under conditions of high membrane potential, but several other sources may be important for ROS generation, such as mitochondrial p66(Shc), monoamine oxidase, α-ketoglutarate dehydogenase, besides redox cycling of redox-active molecules. ROS are able to oxidatively modify lipids, proteins, carbohydrates and nucleic acids in mitochondria and to activate/inactivate signalling pathways by oxidative modification of redox-active factors. Cells are endowed with several defence mechanisms including repair or removal of damaged molecules, and antioxidant systems, either enzymatic or non-enzymatic. Oxidative stress is at the basis of ageing and many pathological disorders, such as ischemic diseases, neurodegenerative diseases, diabetes, and cancer, although the underlying mechanisms are not always completely understood.


Asunto(s)
Mitocondrias/fisiología , Especies Reactivas de Oxígeno/metabolismo , Animales , Mitocondrias/enzimología , Estrés Oxidativo
9.
Adv Exp Med Biol ; 748: 107-44, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22729856

RESUMEN

Recent experimental evidence has replaced the random diffusion model of electron transfer with a model of supramolecular organisation based on specific interactions between individual respiratory complexes. These supercomplexes are detected by blue-native electrophoresis and are found to be functionally relevant by flux control analysis; moreover, they have been isolated and characterised by single-particle electron microscopy. The supramolecular association of individual complexes strongly depends on membrane lipid amount and composition and is affected by lipid peroxidation; it also seems to be modulated by membrane potential and protein phosphorylation. Supercomplex association confers several new properties with respect to the non-associated respiratory complexes to the respiratory chain: the most obvious is substrate channelling, specifically addressing Coenzyme Q and cytochrome c to interact directly with the partner enzymes without the need of a less efficient random diffusion step; in addition, supramolecular association may provide a further rate advantage by conferring long-range conformational changes to the individual complexes. Additional properties are stabilisation of Complex I, as evidenced by the destabilising effect on Complex I of mutations in either Complex III or Complex IV, and prevention of excessive generation of reactive oxygen species. On the basis of the properties described above, we hypothesise that an oxidative stress acts primarily by disassembling supercomplex associations thereby establishing a vicious circle of oxidative stress and energy failure, ultimately leading to cell damage and disease. We provide evidence that in physiological ageing and in some disease states, characterised by oxidative stress and mitochondrial damage, such as heart failure, neurodegenerative disorders and cancer, a loss of supercomplex association occurs, in line with our working hypothesis.


Asunto(s)
Transporte de Electrón , Mitocondrias/metabolismo , Complejos Multienzimáticos/fisiología , Fosforilación Oxidativa , Envejecimiento/metabolismo , Animales , Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/fisiología , Complejo IV de Transporte de Electrones/química , Complejo IV de Transporte de Electrones/fisiología , Humanos , Potencial de la Membrana Mitocondrial
10.
Life (Basel) ; 12(2)2022 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-35207491

RESUMEN

Mitochondria are the powerhouses of cells; however, mitochondrial dysfunction causes energy depletion and cell death in various diseases [...].

11.
Biochim Biophys Acta ; 1797(6-7): 1171-7, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20153717

RESUMEN

It is now clear that mitochondrial defects are associated with a large variety of clinical phenotypes. This is the result of the mitochondria's central role in energy production, reactive oxygen species homeostasis, and cell death. These processes are interdependent and may occur under various stressing conditions, among which low oxygen levels (hypoxia) are certainly prominent. Cells exposed to hypoxia respond acutely with endogenous metabolites and proteins promptly regulating metabolic pathways, but if low oxygen levels are prolonged, cells activate adapting mechanisms, the master switch being the hypoxia-inducible factor 1 (HIF-1). Activation of this factor is strictly bound to the mitochondrial function, which in turn is related with the oxygen level. Therefore in hypoxia, mitochondria act as [O2] sensors, convey signals to HIF-1 directly or indirectly, and contribute to the cell redox potential, ion homeostasis, and energy production. Although over the last two decades cellular responses to low oxygen tension have been studied extensively, mechanisms underlying these functions are still indefinite. Here we review current knowledge of the mitochondrial role in hypoxia, focusing mainly on their role in cellular energy and reactive oxygen species homeostasis in relation with HIF-1 stabilization. In addition, we address the involvement of HIF-1 and the inhibitor protein of F1F0 ATPase in the hypoxia-induced mitochondrial autophagy.


Asunto(s)
Hipoxia de la Célula/fisiología , Mitocondrias/metabolismo , Oxígeno/metabolismo , Animales , Transporte de Electrón , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Metabolismo Energético , Humanos , Factor 1 Inducible por Hipoxia/metabolismo , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Modelos Biológicos , Proteínas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteína Inhibidora ATPasa
12.
Biochim Biophys Acta ; 1797(2): 314-23, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19931505

RESUMEN

Many cancer cells are characterized by high rate of glycolysis and reduced rate of aerobic respiration, whose mechanism is still elusive. Here we investigate the down-regulation of oxidative phosphorylation (OXPHOS) in K-ras transformed mouse fibroblasts as compared to a control counterpart. Transcriptional analysis showed different expression levels of several OXPHOS nuclear genes in the two cell lines. In particular, during the exponential growth phase most genes encoding proteins of Complex I were expressed at lower levels in transformed cells. Consistently, a significant decrease of Complex I content was found in transformed cells. Moreover, analysis of NAD-dependent respiration and ATP synthesis indicated a strong decrease of Complex I activity in the mitochondria from neoplastic cells, that was confirmed by direct assay of the enzyme redox activity. At variance, succinate-dependent respiration and ATP synthesis were not significantly affected. Taken together, our results provide the new insight that the reduction of respiration observed in K-ras transformed cells is specifically due to a Complex I activity decrease.


Asunto(s)
Transformación Celular Neoplásica , Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/metabolismo , Metabolismo Energético , Fibroblastos/metabolismo , Genes ras , Mitocondrias/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Biomarcadores/metabolismo , Proliferación Celular , Respiración de la Célula/fisiología , Células Cultivadas , Perfilación de la Expresión Génica , Potencial de la Membrana Mitocondrial/fisiología , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Oxidación-Reducción , Fosforilación Oxidativa , Oxígeno/metabolismo
13.
Biochim Biophys Acta ; 1797(6-7): 633-40, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20116362

RESUMEN

Recent investigations by native gel electrophoresis showed the existence of supramolecular associations of the respiratory complexes, confirmed by electron microscopy analysis and single particle image processing. Flux control analysis demonstrated that Complex I and Complex III in mammalian mitochondria kinetically behave as a single unit with control coefficients approaching unity for each component, suggesting the existence of substrate channeling within the super-complex. The formation of this supramolecular unit largely depends on the lipid content and composition of the inner mitochondrial membrane. The function of the super-complexes appears not to be restricted to kinetic advantages in electron transfer: we discuss evidence on their role in the stability and assembly of the individual complexes, particularly Complex I, and in preventing excess oxygen radical formation. There is increasing evidence that disruption of the super-complex organization leads to functional derangements responsible for pathological changes, as we have found in K-ras-transformed fibroblasts.


Asunto(s)
Proteínas del Complejo de Cadena de Transporte de Electrón/fisiología , Mitocondrias/patología , Mitocondrias/fisiología , Envejecimiento/patología , Envejecimiento/fisiología , Animales , Transporte de Electrón , Proteínas del Complejo de Cadena de Transporte de Electrón/química , Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/fisiología , Complejo II de Transporte de Electrones/química , Complejo II de Transporte de Electrones/fisiología , Complejo III de Transporte de Electrones/química , Complejo III de Transporte de Electrones/fisiología , Humanos , Cinética , Enfermedades Mitocondriales/patología , Enfermedades Mitocondriales/fisiopatología , Modelos Biológicos , Multimerización de Proteína , Estabilidad Proteica , Especies Reactivas de Oxígeno/metabolismo
14.
Life (Basel) ; 11(3)2021 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-33804034

RESUMEN

Under aerobic conditions, mitochondrial oxidative phosphorylation (OXPHOS) converts the energy released by nutrient oxidation into ATP, the currency of living organisms. The whole biochemical machinery is hosted by the inner mitochondrial membrane (mtIM) where the protonmotive force built by respiratory complexes, dynamically assembled as super-complexes, allows the F1FO-ATP synthase to make ATP from ADP + Pi. Recently mitochondria emerged not only as cell powerhouses, but also as signaling hubs by way of reactive oxygen species (ROS) production. However, when ROS removal systems and/or OXPHOS constituents are defective, the physiological ROS generation can cause ROS imbalance and oxidative stress, which in turn damages cell components. Moreover, the morphology of mitochondria rules cell fate and the formation of the mitochondrial permeability transition pore in the mtIM, which, most likely with the F1FO-ATP synthase contribution, permeabilizes mitochondria and leads to cell death. As the multiple mitochondrial functions are mutually interconnected, changes in protein composition by mutations or in supercomplex assembly and/or in membrane structures often generate a dysfunctional cascade and lead to life-incompatible diseases or severe syndromes. The known structural/functional changes in mitochondrial proteins and structures, which impact mitochondrial bioenergetics because of an impaired or defective energy transduction system, here reviewed, constitute the main biochemical damage in a variety of genetic and age-related diseases.

15.
J Biol Chem ; 284(47): 32331-5, 2009 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-19776013

RESUMEN

Metabolic control analysis was applied to intact HepG2 cells. The effect on the control coefficient of cytochrome c oxidase (CcOX) over cell respiration of both the electrical (Delta psi) and chemical (Delta pH) component of the mitochondrial transmembrane proton electrochemical gradient (Delta mu(H(+))) was investigated. The overall O(2) consumption and specific CcOX activity of actively phosphorylating cells were titrated with cyanide under conditions in which Delta psi and Delta pH were selectively modulated by addition of ionophores. In the absence of ionophores, CcOX displayed a high control coefficient (C(IV) = 0.73), thus representing an important site of regulation of mitochondrial oxidative phosphorylation. A high control coefficient value (C(IV) = 0.85) was also measured in the presence of nigericin, i.e. when Delta psi is maximal, and in the presence of nigericin and valinomycin (C(IV) = 0.77), when Delta mu(H(+)) is abolished. In contrast, CcOX displayed a markedly lower control coefficient (C(IV) = 0.30) upon addition of valinomycin, when Delta psi is converted into Delta pH. These results show that Delta psi is responsible for the tight control of CcOX over respiration in actively phosphorylating cells.


Asunto(s)
Complejo IV de Transporte de Electrones/metabolismo , Potenciales de la Membrana/fisiología , Consumo de Oxígeno , Línea Celular , Cianuros/farmacología , Relación Dosis-Respuesta a Droga , Humanos , Ionóforos/farmacología , Modelos Estadísticos , Nigericina/farmacología , Fosforilación , Valinomicina/farmacología
16.
Biochim Biophys Acta ; 1787(6): 563-73, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19268424

RESUMEN

The kinetic analysis by Kröger and Klingenberg on electron transfer in the Coenzyme Q region led to the conclusion that the quinone behaves kinetically as a homogeneous pool freely diffusing in the lipid bilayer, thus setting the basis for the widely accepted random diffusion model of electron transfer. The recent description of supramolecular complexes of the respiratory chain enzymes, in particular Complex I-III supercomplexes, has reopened the problem of electron transfer in the Coenzyme Q region. Flux control analysis has revealed that Complexes I and III indeed function as a single unit indicating substrate channelling by Coenzyme Q in transferring electrons from Complex I to Complex III. In this review we analyse in detail the reasons that suggested Coenzyme Q pool behaviour; although electron transfer between Complexes I and III indeed appears to be effected by substrate channelling, the Coenzyme Q pool is in equilibrium with bound quinone and is required to fill the site(s) within the supercomplex. In addition, the pool equation of Kröger and Klingenberg still describes in the most adequate way the electron transfer from Complex II and other Coenzyme Q-reducing enzymes to Complex III, besides the energy-dependent reverse electron transfer from Complex II to Complex I.


Asunto(s)
Transporte de Electrón/fisiología , Mitocondrias/metabolismo , Ubiquinona/metabolismo , Animales , Complejo I de Transporte de Electrón/metabolismo , Complejo III de Transporte de Electrones/metabolismo , Cinética
18.
Biochim Biophys Acta ; 1787(5): 384-92, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19059197

RESUMEN

We have investigated the production of reactive oxygen species (ROS) by Complex I in isolated open bovine heart submitochondrial membrane fragments during forward electron transfer in presence of NADH, by means of the probe 2',7'-Dichlorodihydrofluorescein diacetate. ROS production by Complex I is strictly related to its inhibited state. Our results indicate that different Complex I inhibitors can be grouped into two classes: Class A inhibitors (Rotenone, Piericidin A and Rolliniastatin 1 and 2) increase ROS production; Class B inhibitors (Stigmatellin, Mucidin, Capsaicin and Coenzyme Q(2)) prevent ROS production also in the presence of Class A inhibitors. Addition of the hydrophilic Coenzyme Q(1) as an electron acceptor potentiates the effect of Rotenone-like inhibitors in increasing ROS production, but has no effect in the presence of Stigmatellin-like inhibitors; the effect is not shared by more hydrophobic quinones such as decyl-ubiquinone. This behaviour relates the prooxidant CoQ(1) activity to a hydrophilic electron escape site. Moreover the two classes of Complex I inhibitors have an opposite effect on the increase of NADH-DCIP reduction induced by short chain quinones: only Class B inhibitors allow this increase, indicating the presence of a Rotenone-sensitive but Stigmatellin-insensitive semiquinone species in the active site of the enzyme. The presence of this semiquinone was also suggested by preliminary EPR data. The results suggest that electron transfer from the iron-sulphur clusters (N2) to Coenzyme Q occurs in two steps gated by two different conformations, the former being sensitive to Rotenone and the latter to Stigmatellin.


Asunto(s)
Complejo I de Transporte de Electrón/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Partículas Submitocóndricas/metabolismo , Animales , Bovinos , Espectroscopía de Resonancia por Spin del Electrón , Complejo I de Transporte de Electrón/efectos de los fármacos , Complejo I de Transporte de Electrón/metabolismo , Furanos/farmacología , Peróxido de Hidrógeno/metabolismo , Insecticidas/farmacología , Cinética , Mitocondrias/metabolismo , Mitocondrias Cardíacas/metabolismo , Subunidades de Proteína/metabolismo , Piridinas/farmacología , Rotenona/farmacología , Superóxido Dismutasa/metabolismo , Ubiquinona/metabolismo
19.
Biochim Biophys Acta ; 1777(7-8): 941-5, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18486591

RESUMEN

Mitochondrial F(1)F(0)-ATPase was studied in lymphocytes from patients with neuropathy, ataxia, and retinitis pigmentosa (NARP), caused by a mutation at leu-156 in the ATPase 6 subunit. The mutation giving the milder phenotype (Leu156Pro) suffered a 30% reduction in proton flux, and a similar loss in ATP synthetic activity. The more severe mutation (Leu156Arg) also suffered a 30% reduction in proton flux, but ATP synthesis was virtually abolished. Oligomycin sensitivity of the proton translocation through F(0) was enhanced by both mutations. We conclude that in the Leu156Pro mutation, rotation of the c-ring is slowed but coupling of ATP synthesis to proton flux is maintained, whereas in the Leu156Arg mutation, proton flux appears to be uncoupled. Modelling indicated that, in the Leu156Arg mutation, transmembrane helix III of ATPase 6 is unable to span the membrane, terminating in an intramembrane helix II-helix III loop. We propose that the integrity of transmembrane helix III is essential for the mechanical function of ATPase 6 as a stator element in the ATP synthase, but that it is not relevant for oligomycin inhibition.


Asunto(s)
Metabolismo Energético , Mitocondrias/metabolismo , Enfermedades Mitocondriales/metabolismo , ATPasas de Translocación de Protón/metabolismo , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Sustitución de Aminoácidos , Humanos , Cinética , Linfocitos/enzimología , Linfocitos/metabolismo , Mitocondrias/enzimología , Enfermedades Mitocondriales/enzimología , Modelos Moleculares , Conformación Proteica , ATPasas de Translocación de Protón/química , ATPasas de Translocación de Protón/genética
20.
Biochim Biophys Acta ; 1767(7): 913-9, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17568559

RESUMEN

Two point mutations (T>G and T>C) at the same 8993 nucleotide of mitochondrial DNA (at comparable mutant load), affecting the ATPase 6 subunit of the F1F0-ATPase, result in neurological phenotypes of variable severity in humans. We have investigated mitochondrial function in lymphocytes from individuals carrying the 8993T>C mutation: the results were compared with data from five 8993T>G NARP (Neuropathy, Ataxia and Retinitis Pigmentosa) patients. Both 8993T>G and 8993T>C mutations led to energy deprivation and ROS overproduction. However, the relative contribution of the two pathogenic components is different depending on the mutation considered. The 8993T>G change mainly induces an energy deficiency, whereas the 8993T>C favours an increased ROS production. These results possibly highlight the different pathogenic mechanism generated by the two mutations at position 8993 and provide useful information to better characterize the biochemical role of the highly conserved Leu-156 in ATPase 6 subunit of the mitochondrial ATP synthase complex.


Asunto(s)
Ataxia/genética , Genes Mitocondriales , Enfermedad de Leigh/genética , ATPasas de Translocación de Protón Mitocondriales/genética , Enfermedades del Sistema Nervioso Periférico/genética , Retinitis Pigmentosa/genética , Adenosina Trifosfato/metabolismo , Adulto , Análisis Mutacional de ADN , ADN Mitocondrial/genética , Femenino , Humanos , Leucina/química , Leucina/genética , Linfocitos/metabolismo , Potenciales de la Membrana , Persona de Mediana Edad , Mutación , Fenotipo , Especies Reactivas de Oxígeno/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA