Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.473
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 187(3): 659-675.e18, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38215760

RESUMEN

The electron transport chain (ETC) of mitochondria, bacteria, and archaea couples electron flow to proton pumping and is adapted to diverse oxygen environments. Remarkably, in mice, neurological disease due to ETC complex I dysfunction is rescued by hypoxia through unknown mechanisms. Here, we show that hypoxia rescue and hyperoxia sensitivity of complex I deficiency are evolutionarily conserved to C. elegans and are specific to mutants that compromise the electron-conducting matrix arm. We show that hypoxia rescue does not involve the hypoxia-inducible factor pathway or attenuation of reactive oxygen species. To discover the mechanism, we use C. elegans genetic screens to identify suppressor mutations in the complex I accessory subunit NDUFA6/nuo-3 that phenocopy hypoxia rescue. We show that NDUFA6/nuo-3(G60D) or hypoxia directly restores complex I forward activity, with downstream rescue of ETC flux and, in some cases, complex I levels. Additional screens identify residues within the ubiquinone binding pocket as being required for the rescue by NDUFA6/nuo-3(G60D) or hypoxia. This reveals oxygen-sensitive coupling between an accessory subunit and the quinone binding pocket of complex I that can restore forward activity in the same manner as hypoxia.


Asunto(s)
Caenorhabditis elegans , Complejo I de Transporte de Electrón , Hipoxia , Animales , Ratones , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Complejo I de Transporte de Electrón/metabolismo , Hipoxia/genética , Hipoxia/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Oxígeno/metabolismo
2.
Cell ; 179(5): 1222-1238.e17, 2019 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-31730859

RESUMEN

Mitochondrial dysfunction is associated with a spectrum of human conditions, ranging from rare, inborn errors of metabolism to the aging process. To identify pathways that modify mitochondrial dysfunction, we performed genome-wide CRISPR screens in the presence of small-molecule mitochondrial inhibitors. We report a compendium of chemical-genetic interactions involving 191 distinct genetic modifiers, including 38 that are synthetic sick/lethal and 63 that are suppressors. Genes involved in glycolysis (PFKP), pentose phosphate pathway (G6PD), and defense against lipid peroxidation (GPX4) scored high as synthetic sick/lethal. A surprisingly large fraction of suppressors are pathway intrinsic and encode mitochondrial proteins. A striking example of such "intra-organelle" buffering is the alleviation of a chemical defect in complex V by simultaneous inhibition of complex I, which benefits cells by rebalancing redox cofactors, increasing reductive carboxylation, and promoting glycolysis. Perhaps paradoxically, certain forms of mitochondrial dysfunction may best be buffered with "second site" inhibitors to the organelle.


Asunto(s)
Genes Modificadores , Mitocondrias/genética , Mitocondrias/patología , Autoantígenos/metabolismo , Muerte Celular/efectos de los fármacos , Citosol/efectos de los fármacos , Citosol/metabolismo , Complejo I de Transporte de Electrón/metabolismo , Epistasis Genética/efectos de los fármacos , Ferroptosis/efectos de los fármacos , Ferroptosis/genética , Genoma , Glutatión Peroxidasa/metabolismo , Glucólisis/efectos de los fármacos , Glucólisis/genética , Humanos , Células K562 , Mitocondrias/efectos de los fármacos , Oligomicinas/toxicidad , Oxidación-Reducción , Fosforilación Oxidativa/efectos de los fármacos , Vía de Pentosa Fosfato/efectos de los fármacos , Vía de Pentosa Fosfato/genética , Especies Reactivas de Oxígeno/metabolismo , Ribonucleoproteínas/metabolismo , Antígeno SS-B
3.
Cell ; 173(7): 1636-1649.e16, 2018 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-29754813

RESUMEN

Hydrogen gas-evolving membrane-bound hydrogenase (MBH) and quinone-reducing complex I are homologous respiratory complexes with a common ancestor, but a structural basis for their evolutionary relationship is lacking. Here, we report the cryo-EM structure of a 14-subunit MBH from the hyperthermophile Pyrococcus furiosus. MBH contains a membrane-anchored hydrogenase module that is highly similar structurally to the quinone-binding Q-module of complex I while its membrane-embedded ion-translocation module can be divided into a H+- and a Na+-translocating unit. The H+-translocating unit is rotated 180° in-membrane with respect to its counterpart in complex I, leading to distinctive architectures for the two respiratory systems despite their largely conserved proton-pumping mechanisms. The Na+-translocating unit, absent in complex I, resembles that found in the Mrp H+/Na+ antiporter and enables hydrogen gas evolution by MBH to establish a Na+ gradient for ATP synthesis near 100°C. MBH also provides insights into Mrp structure and evolution of MBH-based respiratory enzymes.


Asunto(s)
Proteínas Arqueales/metabolismo , Hidrogenasas/metabolismo , Pyrococcus furiosus/metabolismo , Secuencia de Aminoácidos , Proteínas Arqueales/química , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Sitios de Unión , Membrana Celular/química , Membrana Celular/metabolismo , Microscopía por Crioelectrón , Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/metabolismo , Evolución Molecular , Hidrógeno/metabolismo , Hidrogenasas/química , Hidrogenasas/genética , Mutagénesis , Estructura Cuaternaria de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Alineación de Secuencia , Sodio/química , Sodio/metabolismo , Intercambiadores de Sodio-Hidrógeno/química , Intercambiadores de Sodio-Hidrógeno/metabolismo
4.
Cell ; 170(6): 1247-1257.e12, 2017 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-28844695

RESUMEN

The respiratory megacomplex represents the highest-order assembly of respiratory chain complexes, and it allows mitochondria to respond to energy-requiring conditions. To understand its architecture, we examined the human respiratory chain megacomplex-I2III2IV2 (MCI2III2IV2) with 140 subunits and a subset of associated cofactors using cryo-electron microscopy. The MCI2III2IV2 forms a circular structure with the dimeric CIII located in the center, where it is surrounded by two copies each of CI and CIV. Two cytochrome c (Cyt.c) molecules are positioned to accept electrons on the surface of the c1 state CIII dimer. Analyses indicate that CII could insert into the gaps between CI and CIV to form a closed ring, which we termed the electron transport chain supercomplex. The structure not only reveals the precise assignment of individual subunits of human CI and CIII, but also enables future in-depth analysis of the electron transport chain as a whole.


Asunto(s)
Proteínas del Complejo de Cadena de Transporte de Electrón/química , Complejos Multienzimáticos/química , Microscopía por Crioelectrón , Proteínas del Complejo de Cadena de Transporte de Electrón/aislamiento & purificación , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/aislamiento & purificación , Complejo I de Transporte de Electrón/metabolismo , Complejo II de Transporte de Electrones/química , Complejo II de Transporte de Electrones/aislamiento & purificación , Complejo II de Transporte de Electrones/metabolismo , Humanos , Mitocondrias/química , Mitocondrias/metabolismo , Modelos Moleculares , Complejos Multienzimáticos/aislamiento & purificación , Complejos Multienzimáticos/metabolismo
5.
Mol Cell ; 84(10): 1964-1979.e6, 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38759628

RESUMEN

The role of the mitochondrial electron transport chain (ETC) in regulating ferroptosis is not fully elucidated. Here, we reveal that pharmacological inhibition of the ETC complex I reduces ubiquinol levels while decreasing ATP levels and activating AMP-activated protein kinase (AMPK), the two effects known for their roles in promoting and suppressing ferroptosis, respectively. Consequently, the impact of complex I inhibitors on ferroptosis induced by glutathione peroxidase 4 (GPX4) inhibition is limited. The pharmacological inhibition of complex I in LKB1-AMPK-inactivated cells, or genetic ablation of complex I (which does not trigger apparent AMPK activation), abrogates the AMPK-mediated ferroptosis-suppressive effect and sensitizes cancer cells to GPX4-inactivation-induced ferroptosis. Furthermore, complex I inhibition synergizes with radiotherapy (RT) to selectively suppress the growth of LKB1-deficient tumors by inducing ferroptosis in mouse models. Our data demonstrate a multifaceted role of complex I in regulating ferroptosis and propose a ferroptosis-inducing therapeutic strategy for LKB1-deficient cancers.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Complejo I de Transporte de Electrón , Ferroptosis , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Proteínas Serina-Treonina Quinasas , Ferroptosis/genética , Ferroptosis/efectos de los fármacos , Animales , Humanos , Proteínas Quinasas Activadas por AMP/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Complejo I de Transporte de Electrón/metabolismo , Complejo I de Transporte de Electrón/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Ratones , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa/genética , Línea Celular Tumoral , Neoplasias/genética , Neoplasias/patología , Neoplasias/metabolismo , Neoplasias/tratamiento farmacológico , Quinasas de la Proteína-Quinasa Activada por el AMP/genética , Mitocondrias/metabolismo , Mitocondrias/genética , Mitocondrias/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Transducción de Señal , Femenino
6.
Mol Cell ; 83(6): 942-960.e9, 2023 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-36893757

RESUMEN

Oxygen is toxic across all three domains of life. Yet, the underlying molecular mechanisms remain largely unknown. Here, we systematically investigate the major cellular pathways affected by excess molecular oxygen. We find that hyperoxia destabilizes a specific subset of Fe-S cluster (ISC)-containing proteins, resulting in impaired diphthamide synthesis, purine metabolism, nucleotide excision repair, and electron transport chain (ETC) function. Our findings translate to primary human lung cells and a mouse model of pulmonary oxygen toxicity. We demonstrate that the ETC is the most vulnerable to damage, resulting in decreased mitochondrial oxygen consumption. This leads to further tissue hyperoxia and cyclic damage of the additional ISC-containing pathways. In support of this model, primary ETC dysfunction in the Ndufs4 KO mouse model causes lung tissue hyperoxia and dramatically increases sensitivity to hyperoxia-mediated ISC damage. This work has important implications for hyperoxia pathologies, including bronchopulmonary dysplasia, ischemia-reperfusion injury, aging, and mitochondrial disorders.


Asunto(s)
Hiperoxia , Enfermedades Mitocondriales , Animales , Humanos , Ratones , Complejo I de Transporte de Electrón/metabolismo , Hiperoxia/metabolismo , Hiperoxia/patología , Pulmón/metabolismo , Mitocondrias/metabolismo , Enfermedades Mitocondriales/metabolismo , Oxígeno/metabolismo
7.
Cell ; 160(1-2): 177-90, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25594180

RESUMEN

Reactive oxygen species (ROS) and mitochondrial defects in neurons are implicated in neurodegenerative disease. Here, we find that a key consequence of ROS and neuronal mitochondrial dysfunction is the accumulation of lipid droplets (LD) in glia. In Drosophila, ROS triggers c-Jun-N-terminal Kinase (JNK) and Sterol Regulatory Element Binding Protein (SREBP) activity in neurons leading to LD accumulation in glia prior to or at the onset of neurodegeneration. The accumulated lipids are peroxidated in the presence of ROS. Reducing LD accumulation in glia and lipid peroxidation via targeted lipase overexpression and/or lowering ROS significantly delays the onset of neurodegeneration. Furthermore, a similar pathway leads to glial LD accumulation in Ndufs4 mutant mice with neuronal mitochondrial defects, suggesting that LD accumulation following mitochondrial dysfunction is an evolutionarily conserved phenomenon, and represents an early, transient indicator and promoter of neurodegenerative disease.


Asunto(s)
Gotas Lipídicas/metabolismo , Mitocondrias/metabolismo , Neuroglía/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Complejo I de Transporte de Electrón/genética , Complejo I de Transporte de Electrón/metabolismo , MAP Quinasa Quinasa 4/metabolismo , Ratones , Ratones Noqueados , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Neuroglía/patología , Neuronas/patología , Proteínas de Unión a los Elementos Reguladores de Esteroles/metabolismo
8.
Nature ; 626(7997): 119-127, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38200310

RESUMEN

The evolution of reproductive barriers is the first step in the formation of new species and can help us understand the diversification of life on Earth. These reproductive barriers often take the form of hybrid incompatibilities, in which alleles derived from two different species no longer interact properly in hybrids1-3. Theory predicts that hybrid incompatibilities may be more likely to arise at rapidly evolving genes4-6 and that incompatibilities involving multiple genes should be common7,8, but there has been sparse empirical data to evaluate these predictions. Here we describe a mitonuclear incompatibility involving three genes whose protein products are in physical contact within respiratory complex I of naturally hybridizing swordtail fish species. Individuals homozygous for mismatched protein combinations do not complete embryonic development or die as juveniles, whereas those heterozygous for the incompatibility have reduced complex I function and unbalanced representation of parental alleles in the mitochondrial proteome. We find that the effects of different genetic interactions on survival are non-additive, highlighting subtle complexity in the genetic architecture of hybrid incompatibilities. Finally, we document the evolutionary history of the genes involved, showing signals of accelerated evolution and evidence that an incompatibility has been transferred between species via hybridization.


Asunto(s)
Núcleo Celular , Complejo I de Transporte de Electrón , Peces , Genes Letales , Especiación Genética , Hibridación Genética , Proteínas Mitocondriales , Animales , Alelos , Complejo I de Transporte de Electrón/genética , Peces/clasificación , Peces/embriología , Peces/genética , Peces/crecimiento & desarrollo , Homocigoto , Genes Letales/genética , Especificidad de la Especie , Desarrollo Embrionario/genética , Proteínas Mitocondriales/genética , Núcleo Celular/genética , Heterocigoto , Evolución Molecular
9.
Nature ; 631(8019): 232-239, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38811722

RESUMEN

Mitochondria play a pivotal part in ATP energy production through oxidative phosphorylation, which occurs within the inner membrane through a series of respiratory complexes1-4. Despite extensive in vitro structural studies, determining the atomic details of their molecular mechanisms in physiological states remains a major challenge, primarily because of loss of the native environment during purification. Here we directly image porcine mitochondria using an in situ cryo-electron microscopy approach. This enables us to determine the structures of various high-order assemblies of respiratory supercomplexes in their native states. We identify four main supercomplex organizations: I1III2IV1, I1III2IV2, I2III2IV2 and I2III4IV2, which potentially expand into higher-order arrays on the inner membranes. These diverse supercomplexes are largely formed by 'protein-lipids-protein' interactions, which in turn have a substantial impact on the local geometry of the surrounding membranes. Our in situ structures also capture numerous reactive intermediates within these respiratory supercomplexes, shedding light on the dynamic processes of the ubiquinone/ubiquinol exchange mechanism in complex I and the Q-cycle in complex III. Structural comparison of supercomplexes from mitochondria treated under different conditions indicates a possible correlation between conformational states of complexes I and III, probably in response to environmental changes. By preserving the native membrane environment, our approach enables structural studies of mitochondrial respiratory supercomplexes in reaction at high resolution across multiple scales, from atomic-level details to the broader subcellular context.


Asunto(s)
Respiración de la Célula , Complejo III de Transporte de Electrones , Complejo I de Transporte de Electrón , Mitocondrias , Animales , Microscopía por Crioelectrón , Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/metabolismo , Complejo I de Transporte de Electrón/ultraestructura , Complejo III de Transporte de Electrones/química , Complejo III de Transporte de Electrones/metabolismo , Complejo III de Transporte de Electrones/ultraestructura , Mitocondrias/metabolismo , Mitocondrias/química , Mitocondrias/ultraestructura , Membranas Mitocondriales/metabolismo , Membranas Mitocondriales/química , Membranas Mitocondriales/ultraestructura , Modelos Moleculares , Fosforilación Oxidativa , Porcinos , Ubiquinona/análogos & derivados , Ubiquinona/química , Ubiquinona/metabolismo , Lípidos de la Membrana/química , Lípidos de la Membrana/metabolismo
10.
Nature ; 628(8006): 195-203, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38480879

RESUMEN

Sustained smouldering, or low-grade activation, of myeloid cells is a common hallmark of several chronic neurological diseases, including multiple sclerosis1. Distinct metabolic and mitochondrial features guide the activation and the diverse functional states of myeloid cells2. However, how these metabolic features act to perpetuate inflammation of the central nervous system is unclear. Here, using a multiomics approach, we identify a molecular signature that sustains the activation of microglia through mitochondrial complex I activity driving reverse electron transport and the production of reactive oxygen species. Mechanistically, blocking complex I in pro-inflammatory microglia protects the central nervous system against neurotoxic damage and improves functional outcomes in an animal disease model in vivo. Complex I activity in microglia is a potential therapeutic target to foster neuroprotection in chronic inflammatory disorders of the central nervous system3.


Asunto(s)
Complejo I de Transporte de Electrón , Inflamación , Microglía , Enfermedades Neuroinflamatorias , Animales , Femenino , Humanos , Masculino , Ratones , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Modelos Animales de Enfermedad , Transporte de Electrón/efectos de los fármacos , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Complejo I de Transporte de Electrón/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Inflamación/patología , Microglía/efectos de los fármacos , Microglía/metabolismo , Microglía/patología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/patología , Multiómica , Células Mieloides/metabolismo , Células Mieloides/patología , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Enfermedades Neuroinflamatorias/metabolismo , Enfermedades Neuroinflamatorias/patología , Especies Reactivas de Oxígeno/metabolismo
11.
Mol Cell ; 82(2): 435-446.e7, 2022 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-34847359

RESUMEN

Most mitochondrial proteins are translated in the cytosol and imported into mitochondria. Mutations in the mitochondrial protein import machinery cause human pathologies. However, a lack of suitable tools to measure protein uptake across the mitochondrial proteome has prevented the identification of specific proteins affected by import perturbation. Here, we introduce mePRODmt, a pulsed-SILAC based proteomics approach that includes a booster signal to increase the sensitivity for mitochondrial proteins selectively, enabling global dynamic analysis of endogenous mitochondrial protein uptake in cells. We applied mePRODmt to determine protein uptake kinetics and examined how inhibitors of mitochondrial import machineries affect protein uptake. Monitoring changes in translation and uptake upon mitochondrial membrane depolarization revealed that protein uptake was extensively modulated by the import and translation machineries via activation of the integrated stress response. Strikingly, uptake changes were not uniform, with subsets of proteins being unaffected or decreased due to changes in translation or import capacity.


Asunto(s)
Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Biosíntesis de Proteínas , Proteoma , Proteómica , Carbonil Cianuro m-Clorofenil Hidrazona/farmacología , Complejo I de Transporte de Electrón/metabolismo , Femenino , Células HeLa , Humanos , Cinética , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Biosíntesis de Proteínas/efectos de los fármacos , Transporte de Proteínas , Desacopladores/farmacología
12.
Cell ; 158(6): 1293-1308, 2014 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-25215488

RESUMEN

Fat (Ft) cadherins are enormous cell adhesion molecules that function at the cell surface to regulate the tumor-suppressive Hippo signaling pathway and planar cell polarity (PCP) tissue organization. Mutations in Ft cadherins are found in a variety of tumors, and it is presumed that this is due to defects in either Hippo signaling or PCP. Here, we show Drosophila Ft functions in mitochondria to directly regulate mitochondrial electron transport chain integrity and promote oxidative phosphorylation. Proteolytic cleavage releases a soluble 68 kDa fragment (Ft(mito)) that is imported into mitochondria. Ft(mito) binds directly to NADH dehydrogenase ubiquinone flavoprotein 2 (Ndufv2), a core component of complex I, stabilizing the holoenzyme. Loss of Ft leads to loss of complex I activity, increases in reactive oxygen species, and a switch to aerobic glycolysis. Defects in mitochondrial activity in ft mutants are independent of Hippo and PCP signaling and are reminiscent of the Warburg effect.


Asunto(s)
Cadherinas/metabolismo , Moléculas de Adhesión Celular/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Mitocondrias/metabolismo , Secuencia de Aminoácidos , Animales , Moléculas de Adhesión Celular/química , Polaridad Celular , Proteínas de Drosophila/química , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Complejo I de Transporte de Electrón/metabolismo , Ojo/crecimiento & desarrollo , Genes Supresores de Tumor , Humanos , MAP Quinasa Quinasa 4/metabolismo , Datos de Secuencia Molecular , Transporte de Proteínas , Especies Reactivas de Oxígeno/metabolismo , Alas de Animales/crecimiento & desarrollo
13.
Nature ; 615(7954): 934-938, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36949187

RESUMEN

Mitochondrial energy conversion requires an intricate architecture of the inner mitochondrial membrane1. Here we show that a supercomplex containing all four respiratory chain components contributes to membrane curvature induction in ciliates. We report cryo-electron microscopy and cryo-tomography structures of the supercomplex that comprises 150 different proteins and 311 bound lipids, forming a stable 5.8-MDa assembly. Owing to subunit acquisition and extension, complex I associates with a complex IV dimer, generating a wedge-shaped gap that serves as a binding site for complex II. Together with a tilted complex III dimer association, it results in a curved membrane region. Using molecular dynamics simulations, we demonstrate that the divergent supercomplex actively contributes to the membrane curvature induction and tubulation of cristae. Our findings highlight how the evolution of protein subunits of respiratory complexes has led to the I-II-III2-IV2 supercomplex that contributes to the shaping of the bioenergetic membrane, thereby enabling its functional specialization.


Asunto(s)
Microscopía por Crioelectrón , Complejo III de Transporte de Electrones , Complejo II de Transporte de Electrones , Complejo IV de Transporte de Electrones , Complejo I de Transporte de Electrón , Mitocondrias , Membranas Mitocondriales , Transporte de Electrón , Complejo III de Transporte de Electrones/química , Complejo III de Transporte de Electrones/metabolismo , Complejo III de Transporte de Electrones/ultraestructura , Complejo IV de Transporte de Electrones/química , Complejo IV de Transporte de Electrones/metabolismo , Complejo IV de Transporte de Electrones/ultraestructura , Mitocondrias/química , Mitocondrias/enzimología , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Membranas Mitocondriales/química , Membranas Mitocondriales/enzimología , Membranas Mitocondriales/metabolismo , Membranas Mitocondriales/ultraestructura , Complejo II de Transporte de Electrones/química , Complejo II de Transporte de Electrones/metabolismo , Complejo II de Transporte de Electrones/ultraestructura , Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/metabolismo , Complejo I de Transporte de Electrón/ultraestructura , Multimerización de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Simulación de Dinámica Molecular , Sitios de Unión , Evolución Molecular
14.
Annu Rev Biochem ; 82: 551-75, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23527692

RESUMEN

Complex I (NADH:ubiquinone oxidoreductase) is crucial for respiration in many aerobic organisms. In mitochondria, it oxidizes NADH from the tricarboxylic acid cycle and ß-oxidation, reduces ubiquinone, and transports protons across the inner membrane, contributing to the proton-motive force. It is also a major contributor to cellular production of reactive oxygen species. The redox reaction of complex I is catalyzed in the hydrophilic domain; it comprises NADH oxidation by a flavin mononucleotide, intramolecular electron transfer along a chain of iron-sulfur clusters, and ubiquinone reduction. Redox-coupled proton translocation in the membrane domain requires long-range energy transfer through the protein complex, and the molecular mechanisms that couple the redox and proton-transfer half-reactions are currently unknown. This review evaluates extant data on the mechanisms of energy transduction and superoxide production by complex I, discusses contemporary mechanistic models, and explores how mechanistic studies may contribute to understanding the roles of complex I dysfunctions in human diseases.


Asunto(s)
Complejo I de Transporte de Electrón/metabolismo , Flavinas/metabolismo , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Ubiquinona/metabolismo , Transporte de Electrón , Complejo I de Transporte de Electrón/química , Humanos , Oxidación-Reducción
15.
EMBO J ; 43(2): 225-249, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38177503

RESUMEN

Respiratory complex I (NADH:ubiquinone oxidoreductase) is essential for cellular energy production and NAD+ homeostasis. Complex I mutations cause neuromuscular, mitochondrial diseases, such as Leigh Syndrome, but their molecular-level consequences remain poorly understood. Here, we use a popular complex I-linked mitochondrial disease model, the ndufs4-/- mouse, to define the structural, biochemical, and functional consequences of the absence of subunit NDUFS4. Cryo-EM analyses of the complex I from ndufs4-/- mouse hearts revealed a loose association of the NADH-dehydrogenase module, and discrete classes containing either assembly factor NDUFAF2 or subunit NDUFS6. Subunit NDUFA12, which replaces its paralogue NDUFAF2 in mature complex I, is absent from all classes, compounding the deletion of NDUFS4 and preventing maturation of an NDUFS4-free enzyme. We propose that NDUFAF2 recruits the NADH-dehydrogenase module during assembly of the complex. Taken together, the findings provide new molecular-level understanding of the ndufs4-/- mouse model and complex I-linked mitochondrial disease.


Asunto(s)
Enfermedad de Leigh , Enfermedades Mitocondriales , Animales , Ratones , Complejo I de Transporte de Electrón/genética , Complejo I de Transporte de Electrón/metabolismo , Enfermedad de Leigh/genética , Mitocondrias/metabolismo , Enfermedades Mitocondriales/genética , NAD/metabolismo , NADH Deshidrogenasa/genética , NADH Deshidrogenasa/metabolismo
16.
Nature ; 607(7920): 756-761, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35859172

RESUMEN

Oocytes form before birth and remain viable for several decades before fertilization1. Although poor oocyte quality accounts for most female fertility problems, little is known about how oocytes maintain cellular fitness, or why their quality eventually declines with age2. Reactive oxygen species (ROS) produced as by-products of mitochondrial activity are associated with lower rates of fertilization and embryo survival3-5. Yet, how healthy oocytes balance essential mitochondrial activity with the production of ROS is unknown. Here we show that oocytes evade ROS by remodelling the mitochondrial electron transport chain through elimination of complex I. Combining live-cell imaging and proteomics in human and Xenopus oocytes, we find that early oocytes exhibit greatly reduced levels of complex I. This is accompanied by a highly active mitochondrial unfolded protein response, which is indicative of an imbalanced electron transport chain. Biochemical and functional assays confirm that complex I is neither assembled nor active in early oocytes. Thus, we report a physiological cell type without complex I in animals. Our findings also clarify why patients with complex-I-related hereditary mitochondrial diseases do not experience subfertility. Complex I suppression represents an evolutionarily conserved strategy that allows longevity while maintaining biological activity in long-lived oocytes.


Asunto(s)
Complejo I de Transporte de Electrón , Mitocondrias , Oocitos , Especies Reactivas de Oxígeno , Animales , Transporte de Electrón , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Complejo I de Transporte de Electrón/metabolismo , Femenino , Humanos , Mitocondrias/metabolismo , Oocitos/citología , Oocitos/enzimología , Oocitos/metabolismo , Proteómica , Respuesta de Proteína Desplegada , Xenopus laevis
17.
Nature ; 609(7928): 808-814, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36104567

RESUMEN

Complex I is the first enzyme in the respiratory chain, which is responsible for energy production in mitochondria and bacteria1. Complex I couples the transfer of two electrons from NADH to quinone and the translocation of four protons across the membrane2, but the coupling mechanism remains contentious. Here we present cryo-electron microscopy structures of Escherichia coli complex I (EcCI) in different redox states, including catalytic turnover. EcCI exists mostly in the open state, in which the quinone cavity is exposed to the cytosol, allowing access for water molecules, which enable quinone movements. Unlike the mammalian paralogues3, EcCI can convert to the closed state only during turnover, showing that closed and open states are genuine turnover intermediates. The open-to-closed transition results in the tightly engulfed quinone cavity being connected to the central axis of the membrane arm, a source of substrate protons. Consistently, the proportion of the closed state increases with increasing pH. We propose a detailed but straightforward and robust mechanism comprising a 'domino effect' series of proton transfers and electrostatic interactions: the forward wave ('dominoes stacking') primes the pump, and the reverse wave ('dominoes falling') results in the ejection of all pumped protons from the distal subunit NuoL. This mechanism explains why protons exit exclusively from the NuoL subunit and is supported by our mutagenesis data. We contend that this is a universal coupling mechanism of complex I and related enzymes.


Asunto(s)
Microscopía por Crioelectrón , Complejo I de Transporte de Electrón , Escherichia coli , Animales , Transporte de Electrón , Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/genética , Complejo I de Transporte de Electrón/metabolismo , Complejo I de Transporte de Electrón/ultraestructura , Escherichia coli/enzimología , Escherichia coli/genética , Escherichia coli/metabolismo , Escherichia coli/ultraestructura , Proteínas de Escherichia coli , Mutación , NAD/metabolismo , NADH Deshidrogenasa , Oxidación-Reducción , Subunidades de Proteína , Protones , Quinonas/química , Quinonas/metabolismo , Electricidad Estática , Agua/química
18.
Nature ; 601(7893): 428-433, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34937946

RESUMEN

Although deregulation of transfer RNA (tRNA) biogenesis promotes the translation of pro-tumorigenic mRNAs in cancers1,2, the mechanisms and consequences of tRNA deregulation in tumorigenesis are poorly understood. Here we use a CRISPR-Cas9 screen to focus on genes that have been implicated in tRNA biogenesis, and identify a mechanism by which altered valine tRNA biogenesis enhances mitochondrial bioenergetics in T cell acute lymphoblastic leukaemia (T-ALL). Expression of valine aminoacyl tRNA synthetase is transcriptionally upregulated by NOTCH1, a key oncogene in T-ALL, underlining a role for oncogenic transcriptional programs in coordinating tRNA supply and demand. Limiting valine bioavailability through restriction of dietary valine intake disrupted this balance in mice, resulting in decreased leukaemic burden and increased survival in vivo. Mechanistically, valine restriction reduced translation rates of mRNAs that encode subunits of mitochondrial complex I, leading to defective assembly of complex I and impaired oxidative phosphorylation. Finally, a genome-wide CRISPR-Cas9 loss-of-function screen in differential valine conditions identified several genes, including SLC7A5 and BCL2, whose genetic ablation or pharmacological inhibition synergized with valine restriction to reduce T-ALL growth. Our findings identify tRNA deregulation as a critical adaptation in the pathogenesis of T-ALL and provide a molecular basis for the use of dietary approaches to target tRNA biogenesis in blood malignancies.


Asunto(s)
Leucemia-Linfoma Linfoblástico de Células T Precursoras , Valina-ARNt Ligasa , Valina , Animales , Disponibilidad Biológica , Sistemas CRISPR-Cas , Dieta , Complejo I de Transporte de Electrón/genética , Transportador de Aminoácidos Neutros Grandes 1 , Ratones , Mitocondrias/metabolismo , Fosforilación Oxidativa , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2 , ARN de Transferencia/genética , Valina/metabolismo , Valina-ARNt Ligasa/metabolismo
19.
Nat Rev Mol Cell Biol ; 16(6): 375-88, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25991374

RESUMEN

The mitochondrial respiratory chain, also known as the electron transport chain (ETC), is crucial to life, and energy production in the form of ATP is the main mitochondrial function. Three proton-translocating enzymes of the ETC, namely complexes I, III and IV, generate proton motive force, which in turn drives ATP synthase (complex V). The atomic structures and basic mechanisms of most respiratory complexes have previously been established, with the exception of complex I, the largest complex in the ETC. Recently, the crystal structure of the entire complex I was solved using a bacterial enzyme. The structure provided novel insights into the core architecture of the complex, the electron transfer and proton translocation pathways, as well as the mechanism that couples these two processes.


Asunto(s)
Bacterias/enzimología , Proteínas Bacterianas , Complejo III de Transporte de Electrones , Complejo IV de Transporte de Electrones , Complejo I de Transporte de Electrón , Fuerza Protón-Motriz/fisiología , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/metabolismo , Complejo IV de Transporte de Electrones/química , Complejo IV de Transporte de Electrones/metabolismo , Estructura Cuaternaria de Proteína
20.
Nature ; 599(7886): 650-656, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34732887

RESUMEN

Loss of functional mitochondrial complex I (MCI) in the dopaminergic neurons of the substantia nigra is a hallmark of Parkinson's disease1. Yet, whether this change contributes to Parkinson's disease pathogenesis is unclear2. Here we used intersectional genetics to disrupt the function of MCI in mouse dopaminergic neurons. Disruption of MCI induced a Warburg-like shift in metabolism that enabled neuronal survival, but triggered a progressive loss of the dopaminergic phenotype that was first evident in nigrostriatal axons. This axonal deficit was accompanied by motor learning and fine motor deficits, but not by clear levodopa-responsive parkinsonism-which emerged only after the later loss of dopamine release in the substantia nigra. Thus, MCI dysfunction alone is sufficient to cause progressive, human-like parkinsonism in which the loss of nigral dopamine release makes a critical contribution to motor dysfunction, contrary to the current Parkinson's disease paradigm3,4.


Asunto(s)
Complejo I de Transporte de Electrón/genética , Complejo I de Transporte de Electrón/metabolismo , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/patología , Animales , Axones/efectos de los fármacos , Axones/metabolismo , Axones/patología , Muerte Celular , Dendritas/metabolismo , Dendritas/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Dopamina/metabolismo , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Femenino , Levodopa/farmacología , Levodopa/uso terapéutico , Masculino , Ratones , Destreza Motora/efectos de los fármacos , NADH Deshidrogenasa/deficiencia , NADH Deshidrogenasa/genética , Trastornos Parkinsonianos/tratamiento farmacológico , Trastornos Parkinsonianos/fisiopatología , Fenotipo , Sustancia Negra/citología , Sustancia Negra/efectos de los fármacos , Sustancia Negra/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA