Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 524
Filtrar
1.
Biochem J ; 481(7): 499-514, 2024 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-38572757

RESUMEN

Respiratory complex I is a redox-driven proton pump. Several high-resolution structures of complex I have been determined providing important information about the putative proton transfer paths and conformational transitions that may occur during catalysis. However, how redox energy is coupled to the pumping of protons remains unclear. In this article, we review biochemical, structural and molecular simulation data on complex I and discuss several coupling models, including the key unresolved mechanistic questions. Focusing both on the quinone-reductase domain as well as the proton-pumping membrane-bound domain of complex I, we discuss a molecular mechanism of proton pumping that satisfies most experimental and theoretical constraints. We suggest that protonation reactions play an important role not only in catalysis, but also in the physiologically-relevant active/deactive transition of complex I.


Asunto(s)
Complejo I de Transporte de Electrón , Protones , Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/metabolismo , Antiportadores/metabolismo , Electrones , Simulación de Dinámica Molecular , Oxidación-Reducción , Benzoquinonas
2.
Acta Crystallogr D Struct Biol ; 80(Pt 3): 159-173, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38372588

RESUMEN

Complex I (proton-pumping NADH:ubiquinone oxidoreductase) is the first component of the mitochondrial respiratory chain. In recent years, high-resolution cryo-EM studies of complex I from various species have greatly enhanced the understanding of the structure and function of this important membrane-protein complex. Less well studied is the structural basis of complex I biogenesis. The assembly of this complex of more than 40 subunits, encoded by nuclear or mitochondrial DNA, is an intricate process that requires at least 20 different assembly factors in humans. These are proteins that are transiently associated with building blocks of the complex and are involved in the assembly process, but are not part of mature complex I. Although the assembly pathways have been studied extensively, there is limited information on the structure and molecular function of the assembly factors. Here, the insights that have been gained into the assembly process using cryo-EM are reviewed.


Asunto(s)
Complejo I de Transporte de Electrón , Mitocondrias , Humanos , Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/genética , Complejo I de Transporte de Electrón/metabolismo , Microscopía por Crioelectrón , Mitocondrias/metabolismo
3.
Pest Manag Sci ; 80(6): 2679-2688, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38284296

RESUMEN

BACKGROUND: Bacterial leaf blight caused by Xanthomonas oryzae pv. oryzae (Xoo) is one of the most serious diseases of rice, and there is a lack of bactericides for controlling this disease. We previously found parthenolide (PTL) is a potential lead for developing bactericides against Xoo, and subunit F of respiratory chain complex I (NuoF) is an important target protein of PTL. However, the binding modes of PTL with NuoF need further elucidation. RESULTS: In this study, we obtained the crystal structure of Xoo NuoEF (complex of subunit E and F of respiratory chain complex I) with a resolution of 2.36 Å, which is the first report on the protein structure of NuoEF in plant-pathogenic bacteria. The possible binding sites of PTL with NuoF (Cys105 and Cys187) were predicted with molecular docking and mutated into alanine using a base mismatch method. The mutated proteins were expressed in Escherichia coli and purified with affinity chromatography. The binding abilities of PTL with mutated proteins were investigated via pull-down assay and BIAcore analysis, which revealed that double mutation of Cys105 and Cys187 in NuoF severely affected the binding ability of PTL with NuoF. In addition, the binding modes were further simulated with combined quantum mechanical/molecular mechanical calculations, and the results indicated that PTL may have a stronger binding with Cys105 than Cys187. CONCLUSION: NuoEF protein structure of Xoo was resolved, and Cys105 and Cys187 in NuoF are important binding sites of PTL. This study further clarified the action mechanism of PTL against Xoo, and will promote the innovation of bactericides targeting Xoo complex I. © 2024 Society of Chemical Industry.


Asunto(s)
Proteínas Bacterianas , Simulación del Acoplamiento Molecular , Sesquiterpenos , Xanthomonas , Xanthomonas/efectos de los fármacos , Xanthomonas/genética , Xanthomonas/enzimología , Xanthomonas/metabolismo , Sesquiterpenos/farmacología , Sesquiterpenos/metabolismo , Sesquiterpenos/química , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Complejo I de Transporte de Electrón/metabolismo , Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Complejo I de Transporte de Electrón/genética , Sitios de Unión
4.
Biochem J ; 480(5): 319-333, 2023 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-36920092

RESUMEN

My group and myself have studied respiratory complex I for almost 30 years, starting in 1994 when it was known as a L-shaped giant 'black box' of bioenergetics. First breakthrough was the X-ray structure of the peripheral arm, followed by structures of the membrane arm and finally the entire complex from Thermus thermophilus. The developments in cryo-EM technology allowed us to solve the first complete structure of the twice larger, ∼1 MDa mammalian enzyme in 2016. However, the mechanism coupling, over large distances, the transfer of two electrons to pumping of four protons across the membrane remained an enigma. Recently we have solved high-resolution structures of mammalian and bacterial complex I under a range of redox conditions, including catalytic turnover. This allowed us to propose a robust and universal mechanism for complex I and related protein families. Redox reactions initially drive conformational changes around the quinone cavity and a long-distance transfer of substrate protons. These set up a stage for a series of electrostatically driven proton transfers along the membrane arm ('domino effect'), eventually resulting in proton expulsion from the distal antiporter-like subunit. The mechanism radically differs from previous suggestions, however, it naturally explains all the unusual structural features of complex I. In this review I discuss the state of knowledge on complex I, including the current most controversial issues.


Asunto(s)
Complejo I de Transporte de Electrón , Protones , Animales , Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/metabolismo , Modelos Moleculares , Oxidación-Reducción , Metabolismo Energético , Mamíferos/metabolismo
5.
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
6.
Nat Plants ; 9(1): 142-156, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36585502

RESUMEN

Protein complexes of the mitochondrial respiratory chain assemble into respiratory supercomplexes. Here we present the high-resolution electron cryo-microscopy structure of the Arabidopsis respiratory supercomplex consisting of complex I and a complex III dimer, with a total of 68 protein subunits and numerous bound cofactors. A complex I-ferredoxin, subunit B14.7 and P9, a newly defined subunit of plant complex I, mediate supercomplex formation. The component complexes stabilize one another, enabling new detailed insights into their structure. We describe (1) an interrupted aqueous passage for proton translocation in the membrane arm of complex I; (2) a new coenzyme A within the carbonic anhydrase module of plant complex I defining a second catalytic centre; and (3) the water structure at the proton exit pathway of complex III2 with a co-purified ubiquinone in the QO site. We propose that the main role of the plant supercomplex is to stabilize its components in the membrane.


Asunto(s)
Arabidopsis , Arabidopsis/metabolismo , Microscopía por Crioelectrón , Complejo III de Transporte de Electrones/metabolismo , Protones , Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/metabolismo
7.
Nat Plants ; 9(1): 157-168, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36581760

RESUMEN

The last steps of cellular respiration-an essential metabolic process in plants-are carried out by mitochondrial oxidative phosphorylation. This process involves a chain of multi-subunit membrane protein complexes (complexes I-V) that form higher-order assemblies called supercomplexes. Although supercomplexes are the most physiologically relevant form of the oxidative phosphorylation complexes, their functions and structures remain mostly unknown. Here we present the cryogenic electron microscopy structure of the supercomplex I + III2 from Vigna radiata (mung bean). The structure contains the full subunit complement of complex I, including a newly assigned, plant-specific subunit. It also shows differences in the mitochondrial processing peptidase domain of complex III2 relative to a previously determined supercomplex with complex IV. The supercomplex interface, while reminiscent of that in other organisms, is plant specific, with a major interface involving complex III2's mitochondrial processing peptidase domain and no participation of complex I's bridge domain. The complex I structure suggests that the bridge domain sets the angle between the enzyme's two arms, limiting large-scale conformational changes. Moreover, complex I's catalytic loops and its response in active-to-deactive assays suggest that, in V. radiata, the resting complex adopts a non-canonical state and can sample deactive- or open-like conformations even in the presence of substrate. This study widens our understanding of the possible conformations and behaviour of complex I and supercomplex I + III2. Further studies of complex I and its supercomplexes in diverse organisms are needed to determine the universal and clade-specific mechanisms of respiration.


Asunto(s)
Vigna , Complejo III de Transporte de Electrones/metabolismo , Mitocondrias/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/metabolismo
8.
FEBS Lett ; 597(2): 224-236, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36180980

RESUMEN

Complex I converts oxidoreduction energy into a proton electrochemical gradient across the inner mitochondrial or bacterial cell membrane. This gradient is the primary source of energy for aerobic synthesis of ATP. Oxidation of reduced nicotinamide adenine dinucleotide (NADH) by ubiquinone (Q) yields NAD+ and ubiquinol (QH2 ), which is tightly coupled to translocation of four protons from the negatively to the positively charged side of the membrane. Electrons from NADH oxidation reach the iron-sulfur centre N2 positioned near the bottom of a tunnel that extends circa 30 Å from the membrane domain into the hydrophilic domain of the complex. The tunnel is occupied by ubiquinone, which can take a distal position near the N2 centre or proximal positions closer to the membrane. Here, we review important structural, kinetic and thermodynamic properties of ubiquinone that define its role in complex I function. We suggest that this function exceeds that of a mere substrate or electron acceptor and propose that ubiquinone may be the redox element of complex I coupling electron transfer to proton translocation.


Asunto(s)
Complejo I de Transporte de Electrón , Ubiquinona , Ubiquinona/química , Complejo I de Transporte de Electrón/química , Protones , NAD/metabolismo , Oxidación-Reducción , Transporte de Electrón
9.
Curr Opin Struct Biol ; 77: 102447, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36087446

RESUMEN

Respiratory complex I (NADH:ubiquinone oxidoreductase) is a multi-subunit, energy-transducing mitochondrial enzyme that is essential for oxidative phosphorylation and regulating NAD+/NADH pools. Despite recent advances in structural knowledge and a long history of biochemical analyses, the mechanism of redox-coupled proton translocation by complex I remains unknown. Due to its ability to separate molecules in a mixed population into distinct classes, single-particle electron cryomicroscopy has enabled identification and characterisation of different complex I conformations. However, deciding on their catalytic and/or regulatory properties to underpin mechanistic hypotheses, especially without detailed biochemical characterisation of the structural samples, has proven challenging. In this review we explore different mechanistic interpretations of the closed and open states identified in cryoEM analyses of mammalian complex I.


Asunto(s)
Complejo I de Transporte de Electrón , NAD , Animales , Complejo I de Transporte de Electrón/química , Microscopía por Crioelectrón , NAD/química , NAD/metabolismo , Ubiquinona/química , Ubiquinona/metabolismo , Oxidación-Reducción , Mamíferos/metabolismo
10.
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
11.
Proc Natl Acad Sci U S A ; 119(27): e2123090119, 2022 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-35759670

RESUMEN

Energy-converting NADH:ubiquinone oxidoreductase, respiratory complex I, is essential for cellular energy metabolism coupling NADH oxidation to proton translocation. The mechanism of proton translocation by complex I is still under debate. Its membrane arm contains an unusual central axis of polar and charged amino acid residues connecting the quinone binding site with the antiporter-type subunits NuoL, NuoM, and NuoN, proposed to catalyze proton translocation. Quinone chemistry probably causes conformational changes and electrostatic interactions that are propagated through these subunits by a conserved pattern of predominantly lysine, histidine, and glutamate residues. These conserved residues are thought to transfer protons along and across the membrane arm. The distinct charge distribution in the membrane arm is a prerequisite for proton translocation. Remarkably, the central subunit NuoM contains a conserved glutamate residue in a position that is taken by a lysine residue in the two other antiporter-type subunits. It was proposed that this charge asymmetry is essential for proton translocation, as it should enable NuoM to operate asynchronously with NuoL and NuoN. Accordingly, we exchanged the conserved glutamate in NuoM for a lysine residue, introducing charge symmetry in the membrane arm. The stably assembled variant pumps protons across the membrane, but with a diminished H+/e- stoichiometry of 1.5. Thus, charge asymmetry is not essential for proton translocation by complex I, casting doubts on the suggestion of an asynchronous operation of NuoL, NuoM, and NuoN. Furthermore, our data emphasize the importance of a balanced charge distribution in the protein for directional proton transfer.


Asunto(s)
Membrana Celular , Complejo I de Transporte de Electrón , Proteínas de Escherichia coli , NADH Deshidrogenasa , Sustitución de Aminoácidos , Membrana Celular/química , Secuencia Conservada , Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/genética , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Glutamatos/química , Glutamatos/genética , Lisina/química , Lisina/genética , NADH Deshidrogenasa/química , NADH Deshidrogenasa/genética , Protones , Quinonas/química
12.
J Phys Chem B ; 126(22): 4080-4088, 2022 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-35612955

RESUMEN

We apply linear response theory to calculate mechanical allosteric couplings in respiratory complex I between the iron sulfur cluster N2, located in the catalytic cavity, and the membrane part of the enzyme, separated from it by more than 50 Å. According to our hypothesis, the redox reaction of ubiquinone in the catalytic cavity of the enzyme generates an unbalanced charge that via repulsion of the charged redox center N2 produces local mechanical stress that transmits into the membrane part of the enzyme where it induces proton pumping. Using coarse-grained simulations of the enzyme, we calculated mechanistic allosteric couplings that reveal the pathways of the mechanical transmission of the stress along the enzyme. The results shed light on the recent experimental studies where a stabilization of the enzyme with an introduced disulfide bridge resulted in the abolishing of proton pumping. Simulation of the disulfide bond action indicates a dramatic change of the mechanistic coupling pathways in line with experimental findings.


Asunto(s)
Complejo I de Transporte de Electrón , Proteínas Hierro-Azufre , Disulfuros/metabolismo , Complejo I de Transporte de Electrón/química , Proteínas Hierro-Azufre/química , Oxidación-Reducción , Protones
14.
Curr Opin Struct Biol ; 74: 102350, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35316665

RESUMEN

Complex I is one of the major respiratory complexes, conserved from bacteria to mammals. It oxidises NADH, reduces quinone and pumps protons across the membrane, thus playing a central role in the oxidative energy metabolism. In this review we discuss our current state of understanding the structure of complex I from various species of mammals, plants, fungi, and bacteria, as well as of several complex I-related proteins. By comparing the structural evidence from these systems in different redox states and data from mutagenesis and molecular simulations, we formulate the mechanisms of electron transfer and proton pumping and explain how they are conformationally and electrostatically coupled. Finally, we discuss the structural basis of the deactivation phenomenon in mammalian complex I.


Asunto(s)
Complejo I de Transporte de Electrón , Protones , Animales , Transporte de Electrón , Complejo I de Transporte de Electrón/química , Metabolismo Energético , Mamíferos/metabolismo , Oxidación-Reducción
15.
Molecules ; 27(4)2022 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-35209128

RESUMEN

The finding that the most common mitochondrial DNA mutation m.11778G>A/MT-ND4 (p.R340H) associated with Leber's hereditary optic neuropathy (LHON) induces rotenone resistance has produced a long-standing debate, because it contrasts structural evidence showing that the ND4 subunit is far away from the quinone-reaction site in complex I, where rotenone acts. However, recent cryo-electron microscopy data revealed that rotenone also binds to the ND4 subunit. We investigated the possible structural modifications induced by the LHON mutation and found that its amino acid replacement would disrupt a possible hydrogen bond between native R340 and Q139 in ND4, thereby destabilizing rotenone binding. Our analysis thus explains rotenone resistance in LHON patients as a biochemical signature of its pathogenic effect on complex I.


Asunto(s)
Alelos , Sustitución de Aminoácidos , Resistencia a Medicamentos/genética , Complejo I de Transporte de Electrón/genética , Mutación , Atrofia Óptica Hereditaria de Leber/genética , Rotenona/farmacología , Secuencia de Aminoácidos , Sitios de Unión , Secuencia Conservada , Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/metabolismo , Modelos Moleculares , Atrofia Óptica Hereditaria de Leber/metabolismo , Unión Proteica , Conformación Proteica , Rotenona/química , Relación Estructura-Actividad , Desacopladores/farmacología
16.
Nat Struct Mol Biol ; 29(2): 172-182, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35145322

RESUMEN

Mammalian respiratory complex I (CI) is a 45-subunit, redox-driven proton pump that generates an electrochemical gradient across the mitochondrial inner membrane to power ATP synthesis in mitochondria. In the present study, we report cryo-electron microscopy structures of CI from Sus scrofa in six treatment conditions at a resolution of 2.4-3.5 Å, in which CI structures of each condition can be classified into two biochemical classes (active or deactive), with a notably higher proportion of active CI particles. These structures illuminate how hydrophobic ubiquinone-10 (Q10) with its long isoprenoid tail is bound and reduced in a narrow Q chamber comprising four different Q10-binding sites. Structural comparisons of active CI structures from our decylubiquinone-NADH and rotenone-NADH datasets reveal that Q10 reduction at site 1 is not coupled to proton pumping in the membrane arm, which might instead be coupled to Q10 oxidation at site 2. Our data overturn the widely accepted previous proposal about the coupling mechanism of CI.


Asunto(s)
Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/metabolismo , Animales , Sitios de Unión , Microscopía por Crioelectrón , Complejo I de Transporte de Electrón/ultraestructura , Mitocondrias Cardíacas/metabolismo , Modelos Moleculares , Oxidación-Reducción , Conformación Proteica , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Sus scrofa , Ubiquinona/análogos & derivados , Ubiquinona/química , Ubiquinona/metabolismo
17.
IUBMB Life ; 74(7): 629-644, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35166025

RESUMEN

The flavin mononucleotide (FMN) cofactor of respiratory complex I occupies a key position in the electron transport chain. Here, the electrons coming from NADH start the sequence of oxidoreduction reactions, which drives the generation of the proton-motive force necessary for ATP synthesis. The overall architecture and the general catalytic proprieties of the FMN site are mostly well established. However, several aspects regarding the complex I flavin cofactor are still unknown. For example, the flavin binding to the N-module, the NADH-oxidizing portion of complex I, lacks a molecular description. The dissociation of FMN from the enzyme is beginning to emerge as an important regulatory mechanism of complex I activity and ROS production. Finally, how mitochondria import and metabolize FMN is still uncertain. This review summarizes the current knowledge on complex I flavin cofactor and discusses the open questions for future research.


Asunto(s)
Complejo I de Transporte de Electrón , Mononucleótido de Flavina , Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/genética , Complejo I de Transporte de Electrón/metabolismo , Mononucleótido de Flavina/química , Mononucleótido de Flavina/metabolismo , Flavinas/química , Flavinas/metabolismo , NAD/química , NAD/metabolismo , Oxidación-Reducción
18.
Structure ; 30(1): 80-94.e4, 2022 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-34562374

RESUMEN

Respiratory complex I drives proton translocation across energy-transducing membranes by NADH oxidation coupled with (ubi)quinone reduction. In humans, its dysfunction is associated with neurodegenerative diseases. The Escherichia coli complex represents the structural minimal form of an energy-converting NADH:ubiquinone oxidoreductase. Here, we report the structure of the peripheral arm of the E. coli complex I consisting of six subunits, the FMN cofactor, and nine iron-sulfur clusters at 2.7 Å resolution obtained by cryo electron microscopy. While the cofactors are in equivalent positions as in the complex from other species, individual subunits are adapted to the absence of supernumerary proteins to guarantee structural stability. The catalytically important subunits NuoC and D are fused resulting in a specific architecture of functional importance. Striking features of the E. coli complex are scrutinized by mutagenesis and biochemical characterization of the variants. Moreover, the arrangement of the subunits sheds light on the unknown assembly of the complex.


Asunto(s)
Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/metabolismo , Escherichia coli/metabolismo , Mutación , Sitios de Unión , Microscopía por Crioelectrón , Complejo I de Transporte de Electrón/genética , Escherichia coli/química , Escherichia coli/genética , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Conformación Proteica , Estabilidad Proteica , Subunidades de Proteína/química , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo
19.
Am J Physiol Cell Physiol ; 322(1): C12-C23, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34757853

RESUMEN

Mitochondrial reactive oxygen species (ROS) have emerged as an important mechanism of disease and redox signaling in the cellular system. Under basal or pathological conditions, electron leakage for ROS production is primarily mediated by complexes I and III of the electron transport chain (ETC) and by the proton motive force (PMF), consisting of a membrane potential (ΔΨ) and a proton gradient (ΔpH). Several factors control redox status in mitochondria, including ROS, the PMF, oxidative posttranslational modifications (OPTM) of the ETC subunits, SOD2, and cytochrome c heme lyase (HCCS). In the mitochondrial PMF, increased ΔpH-supported backpressure due to diminishing electron transport and chemiosmosis promotes a more reductive mitochondrial physiological setting. OPTM by protein cysteine sulfonation in complex I and complex III has been shown to affect enzymatic catalysis, the proton gradient, redox status, and enzyme-mediated ROS production. Pathological conditions associated with oxidative or nitrosative stress, such as myocardial ischemia and reperfusion (I/R), increase mitochondrial ROS production and redox dysfunction via oxidative injury to complexes I and III, intensely enhancing protein cysteine sulfonation and impairing heme integrity. The physiological conditions of reductive stress induced by gains in SOD2 function normalize I/R-mediated ROS overproduction and redox dysfunction. Further insight into the cellular mechanisms by which HCCS, biogenesis of c-type cytochrome, and OPTM regulate PMF and ROS production in mitochondria will enrich our understanding of redox signal transduction and identify new therapeutic targets for cardiovascular diseases in which oxidative stress perturbs normal redox signaling.


Asunto(s)
Mitocondrias Cardíacas/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Fosforilación Oxidativa , Especies Reactivas de Oxígeno/metabolismo , Animales , Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/metabolismo , Humanos , Mitocondrias Cardíacas/patología , Daño por Reperfusión Miocárdica/patología , Oxidación-Reducción , Estructura Secundaria de Proteína
20.
Angew Chem Int Ed Engl ; 60(52): 27277-27281, 2021 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-34612584

RESUMEN

NADH:ubiquinone oxidoreductase, respiratory complex I, plays a central role in cellular energy metabolism. As a major source of reactive oxygen species (ROS) it affects ageing and mitochondrial dysfunction. The novel inhibitor NADH-OH specifically blocks NADH oxidation and ROS production by complex I in nanomolar concentrations. Attempts to elucidate its structure by NMR spectroscopy have failed. Here, by using X-ray crystallographic analysis, we report the structure of NADH-OH bound in the active site of a soluble fragment of complex I at 2.0 Šresolution. We have identified key amino acid residues that are specific and essential for binding NADH-OH. Furthermore, the structure sheds light on the specificity of NADH-OH towards the unique Rossmann-fold of complex I and indicates a regulatory role in mitochondrial ROS generation. In addition, NADH-OH acts as a lead-structure for the synthesis of a novel class of ROS suppressors.


Asunto(s)
Complejo I de Transporte de Electrón/antagonistas & inhibidores , Inhibidores Enzimáticos/química , NAD/análogos & derivados , Aquifex/enzimología , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/metabolismo , Inhibidores Enzimáticos/farmacología , Humanos , Enlace de Hidrógeno , Modelos Moleculares , NAD/química , NAD/metabolismo , NAD/farmacología , Unión Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA