Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
EMBO J ; 43(2): 225-249, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38177503

RESUMO

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.


Assuntos
Doença de Leigh , Doenças Mitocondriais , Animais , Camundongos , Complexo I de Transporte de Elétrons/genética , Complexo I de Transporte de Elétrons/metabolismo , Doença de Leigh/genética , Mitocôndrias/metabolismo , Doenças Mitocondriais/genética , NAD/metabolismo , NADH Desidrogenase/genética , NADH Desidrogenase/metabolismo
2.
Sci Adv ; 9(31): eadi1359, 2023 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-37531432

RESUMO

Respiratory complex I, a key enzyme in mammalian metabolism, captures the energy released by reduction of ubiquinone by NADH to drive protons across the inner mitochondrial membrane, generating the proton-motive force for ATP synthesis. Despite remarkable advances in structural knowledge of this complicated membrane-bound enzyme, its mechanism of catalysis remains controversial. In particular, how ubiquinone reduction is coupled to proton pumping and the pathways and mechanisms of proton translocation are contested. We present a 2.4-Å resolution cryo-EM structure of complex I from mouse heart mitochondria in the closed, active (ready-to-go) resting state, with 2945 water molecules modeled. By analyzing the networks of charged and polar residues and water molecules present, we evaluate candidate pathways for proton transfer through the enzyme, for the chemical protons for ubiquinone reduction, and for the protons transported across the membrane. Last, we compare our data to the predictions of extant mechanistic models, and identify key questions to answer in future work to test them.


Assuntos
Complexo I de Transporte de Elétrons , Prótons , Camundongos , Animais , Oxirredução , Complexo I de Transporte de Elétrons/metabolismo , Ubiquinona/química , Ubiquinona/metabolismo , Microscopia Crioeletrônica , Mamíferos/metabolismo , Água/metabolismo
3.
Elife ; 122023 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-36622099

RESUMO

Respiratory complex I powers ATP synthesis by oxidative phosphorylation, exploiting the energy from NADH oxidation by ubiquinone to drive protons across an energy-transducing membrane. Drosophila melanogaster is a candidate model organism for complex I due to its high evolutionary conservation with the mammalian enzyme, well-developed genetic toolkit, and complex physiology for studies in specific cell types and tissues. Here, we isolate complex I from Drosophila and determine its structure, revealing a 43-subunit assembly with high structural homology to its 45-subunit mammalian counterpart, including a hitherto unknown homologue to subunit NDUFA3. The major conformational state of the Drosophila enzyme is the mammalian-type 'ready-to-go' active resting state, with a fully ordered and enclosed ubiquinone-binding site, but a subtly altered global conformation related to changes in subunit ND6. The mammalian-type 'deactive' pronounced resting state is not observed: in two minor states, the ubiquinone-binding site is unchanged, but a deactive-type π-bulge is present in ND6-TMH3. Our detailed structural knowledge of Drosophila complex I provides a foundation for new approaches to disentangle mechanisms of complex I catalysis and regulation in bioenergetics and physiology.


Assuntos
Drosophila melanogaster , Complexo I de Transporte de Elétrons , Animais , Microscopia Crioeletrônica , Drosophila melanogaster/metabolismo , Complexo I de Transporte de Elétrons/metabolismo , Complexo I de Transporte de Elétrons/ultraestrutura , Mitocôndrias/metabolismo , Ubiquinona/metabolismo
4.
J Biol Chem ; 298(3): 101602, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35063503

RESUMO

Mitochondrial complex I (NADH:ubiquinone oxidoreductase), a crucial enzyme in energy metabolism, captures the redox potential energy from NADH oxidation/ubiquinone reduction to create the proton motive force used to drive ATP synthesis in oxidative phosphorylation. High-resolution single-particle electron cryo-EM analyses have provided detailed structural knowledge of the catalytic machinery of complex I, but not of the molecular principles of its energy transduction mechanism. Although ubiquinone is considered to bind in a long channel at the interface of the membrane-embedded and hydrophilic domains, with channel residues likely involved in coupling substrate reduction to proton translocation, no structures with the channel fully occupied have yet been described. Here, we report the structure (determined by cryo-EM) of mouse complex I with a tight-binding natural product acetogenin inhibitor, which resembles the native substrate, bound along the full length of the expected ubiquinone-binding channel. Our structure reveals the mode of acetogenin binding and the molecular basis for structure-activity relationships within the acetogenin family. It also shows that acetogenins are such potent inhibitors because they are highly hydrophobic molecules that contain two specific hydrophilic moieties spaced to lock into two hydrophilic regions of the otherwise hydrophobic channel. The central hydrophilic section of the channel does not favor binding of the isoprenoid chain when the native substrate is fully bound but stabilizes the ubiquinone/ubiquinol headgroup as it transits to/from the active site. Therefore, the amphipathic nature of the channel supports both tight binding of the amphipathic inhibitor and rapid exchange of the ubiquinone/ubiquinol substrate and product.


Assuntos
Acetogeninas , Complexo I de Transporte de Elétrons , Acetogeninas/antagonistas & inibidores , Acetogeninas/metabolismo , Acetogeninas/farmacologia , Animais , Microscopia Crioeletrônica , Complexo I de Transporte de Elétrons/metabolismo , Camundongos , NAD/metabolismo , Oxirredução , Relação Estrutura-Atividade , Ubiquinona/metabolismo
5.
Nat Commun ; 11(1): 5261, 2020 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-33067417

RESUMO

Respiratory complex I (NADH:ubiquinone oxidoreductase) captures the free energy from oxidising NADH and reducing ubiquinone to drive protons across the mitochondrial inner membrane and power oxidative phosphorylation. Recent cryo-EM analyses have produced near-complete models of the mammalian complex, but leave the molecular principles of its long-range energy coupling mechanism open to debate. Here, we describe the 3.0-Å resolution cryo-EM structure of complex I from mouse heart mitochondria with a substrate-like inhibitor, piericidin A, bound in the ubiquinone-binding active site. We combine our structural analyses with both functional and computational studies to demonstrate competitive inhibitor binding poses and provide evidence that two inhibitor molecules bind end-to-end in the long substrate binding channel. Our findings reveal information about the mechanisms of inhibition and substrate reduction that are central for understanding the principles of energy transduction in mammalian complex I.


Assuntos
Complexo I de Transporte de Elétrons/química , Complexo I de Transporte de Elétrons/metabolismo , Inibidores Enzimáticos/metabolismo , Mamíferos/metabolismo , Animais , Sítios de Ligação , Microscopia Crioeletrônica , Complexo I de Transporte de Elétrons/antagonistas & inibidores , Complexo I de Transporte de Elétrons/genética , Inibidores Enzimáticos/química , Feminino , Mamíferos/genética , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias Cardíacas/genética , Mitocôndrias Cardíacas/metabolismo , Simulação de Dinâmica Molecular , Fosforilação Oxidativa , Piridinas/química , Piridinas/metabolismo
6.
Annu Rev Biophys ; 48: 165-184, 2019 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-30786232

RESUMO

Single-particle electron cryomicroscopy (cryo-EM) has led to a revolution in structural work on mammalian respiratory complex I. Complex I (mitochondrial NADH:ubiquinone oxidoreductase), a membrane-bound redox-driven proton pump, is one of the largest and most complicated enzymes in the mammalian cell. Rapid progress, following the first 5-Å resolution data on bovine complex I in 2014, has led to a model for mouse complex I at 3.3-Å resolution that contains 96% of the 8,518 residues and to the identification of different particle classes, some of which are assigned to biochemically defined states. Factors that helped improve resolution, including improvements to biochemistry, cryo-EM grid preparation, data collection strategy, and image processing, are discussed. Together with recent structural data from an ancient relative, membrane-bound hydrogenase, cryo-EM on mammalian complex I has provided new insights into the proton-pumping machinery and a foundation for understanding the enzyme's catalytic mechanism.


Assuntos
Microscopia Crioeletrônica , Complexo I de Transporte de Elétrons/ultraestrutura , Animais , Humanos , Lentes , Mitocôndrias , Oxirredução
7.
Nat Med ; 24(7): 1036-1046, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29892070

RESUMO

Metabolic reprograming is an emerging hallmark of tumor biology and an actively pursued opportunity in discovery of oncology drugs. Extensive efforts have focused on therapeutic targeting of glycolysis, whereas drugging mitochondrial oxidative phosphorylation (OXPHOS) has remained largely unexplored, partly owing to an incomplete understanding of tumor contexts in which OXPHOS is essential. Here, we report the discovery of IACS-010759, a clinical-grade small-molecule inhibitor of complex I of the mitochondrial electron transport chain. Treatment with IACS-010759 robustly inhibited proliferation and induced apoptosis in models of brain cancer and acute myeloid leukemia (AML) reliant on OXPHOS, likely owing to a combination of energy depletion and reduced aspartate production that leads to impaired nucleotide biosynthesis. In models of brain cancer and AML, tumor growth was potently inhibited in vivo following IACS-010759 treatment at well-tolerated doses. IACS-010759 is currently being evaluated in phase 1 clinical trials in relapsed/refractory AML and solid tumors.


Assuntos
Neoplasias/patologia , Fosforilação Oxidativa , Animais , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Metabolismo Energético , Glicólise/efeitos dos fármacos , Células HEK293 , Humanos , Ácido Láctico/metabolismo , Leucemia Mieloide Aguda/patologia , Camundongos , Mitocôndrias/metabolismo , Nucleotídeos/biossíntese , Carga Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Nat Struct Mol Biol ; 25(7): 548-556, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29915388

RESUMO

Complex I (NADH:ubiquinone oxidoreductase) uses the reducing potential of NADH to drive protons across the energy-transducing inner membrane and power oxidative phosphorylation in mammalian mitochondria. Recent cryo-EM analyses have produced near-complete models of all 45 subunits in the bovine, ovine and porcine complexes and have identified two states relevant to complex I in ischemia-reperfusion injury. Here, we describe the 3.3-Å structure of complex I from mouse heart mitochondria, a biomedically relevant model system, in the 'active' state. We reveal a nucleotide bound in subunit NDUFA10, a nucleoside kinase homolog, and define mechanistically critical elements in the mammalian enzyme. By comparisons with a 3.9-Å structure of the 'deactive' state and with known bacterial structures, we identify differences in helical geometry in the membrane domain that occur upon activation or that alter the positions of catalytically important charged residues. Our results demonstrate the capability of cryo-EM analyses to challenge and develop mechanistic models for mammalian complex I.


Assuntos
Complexo I de Transporte de Elétrons/química , Mitocôndrias Cardíacas/enzimologia , Animais , Sítios de Ligação , Microscopia Crioeletrônica , Complexo I de Transporte de Elétrons/metabolismo , Complexo I de Transporte de Elétrons/ultraestrutura , Ativação Enzimática , Camundongos , Modelos Moleculares , NADH Desidrogenase/química , NADH Desidrogenase/metabolismo , NADH Desidrogenase/ultraestrutura , Nucleotídeos/química , Nucleotídeos/metabolismo , Fosfolipídeos/química , Fosfolipídeos/metabolismo , Elementos Estruturais de Proteínas , Estrutura Quaternária de Proteína , Subunidades Proteicas , Ubiquinona/química , Ubiquinona/metabolismo
9.
BMC Biol ; 14: 65, 2016 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-27506389

RESUMO

BACKGROUND: The biguanides are a family of drugs with diverse clinical applications. Metformin, a widely used anti-hyperglycemic biguanide, suppresses mitochondrial respiration by inhibiting respiratory complex I. Phenformin, a related anti-hyperglycemic biguanide, also inhibits respiration, but proguanil, which is widely used for the prevention of malaria, does not. The molecular structures of phenformin and proguanil are closely related and both inhibit isolated complex I. Proguanil does not inhibit respiration in cells and mitochondria because it is unable to access complex I. The molecular features that determine which biguanides accumulate in mitochondria, enabling them to inhibit complex I in vivo, are not known. RESULTS: Here, a family of seven biguanides are used to reveal the molecular features that determine why phenformin enters mitochondria and inhibits respiration whereas proguanil does not. All seven biguanides inhibit isolated complex I, but only four of them inhibit respiration in cells and mitochondria. Direct conjugation of a phenyl group and bis-substitution of the biguanide moiety prevent uptake into mitochondria, irrespective of the compound hydrophobicity. This high selectivity suggests that biguanide uptake into mitochondria is protein mediated, and is not by passive diffusion. Only those biguanides that enter mitochondria and inhibit complex I activate AMP kinase, strengthening links between complex I and the downstream effects of biguanide treatments. CONCLUSIONS: Biguanides inhibit mitochondrial complex I, but specific molecular features control the uptake of substituted biguanides into mitochondria, so only some biguanides inhibit mitochondrial respiration in vivo. Biguanides with restricted intracellular access may be used to determine physiologically relevant targets of biguanide action, and for the rational design of substituted biguanides for diverse clinical applications.


Assuntos
Adenilato Quinase/metabolismo , Biguanidas/química , Biguanidas/farmacologia , Complexo I de Transporte de Elétrons/metabolismo , Animais , Bovinos , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular/efeitos dos fármacos , Respiração Celular/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Membranas Mitocondriais/efeitos dos fármacos , Membranas Mitocondriais/metabolismo , Consumo de Oxigênio/efeitos dos fármacos , Fenformin/farmacologia , Ratos , Rotenona/farmacologia , Solubilidade
10.
Genome Biol Evol ; 5(10): 1969-77, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24068653

RESUMO

Respiratory electron transport in mitochondria is coupled to ATP synthesis while generating mutagenic oxygen free radicals. Mitochondrial DNA mutation then accumulates with age, and may set a limit to the lifespan of individual, multicellular organisms. Why is this mutation not inherited? Here we demonstrate that female gametes-oocytes-have unusually small and simple mitochondria that are suppressed for DNA transcription, electron transport, and free radical production. By contrast, male gametes-sperm-and somatic cells of both sexes transcribe mitochondrial genes for respiratory electron carriers and produce oxygen free radicals. This germ-line division between mitochondria of sperm and egg is observed in both the vinegar fruitfly and the zebrafish-species spanning a major evolutionary divide within the animal kingdom. We interpret these findings as an evidence that oocyte mitochondria serve primarily as genetic templates, giving rise, irreversibly and in each new generation, to the familiar energy-transducing mitochondria of somatic cells and male gametes. Suppressed mitochondrial metabolism in the female germ line may therefore constitute a mechanism for increasing the fidelity of mitochondrial DNA inheritance.


Assuntos
Evolução Biológica , DNA Mitocondrial/genética , Oócitos/metabolismo , Espermatozoides/metabolismo , Transcrição Gênica , Trifosfato de Adenosina/biossíntese , Envelhecimento/genética , Animais , Transporte de Elétrons/genética , Feminino , Radicais Livres/metabolismo , Células Germinativas/metabolismo , Masculino , Mitocôndrias/genética , Mitocôndrias/metabolismo , Oxigênio/metabolismo , Peixe-Zebra/metabolismo
11.
Philos Trans R Soc Lond B Biol Sci ; 368(1622): 20120263, 2013 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-23754815

RESUMO

Oxidative phosphorylation couples ATP synthesis to respiratory electron transport. In eukaryotes, this coupling occurs in mitochondria, which carry DNA. Respiratory electron transport in the presence of molecular oxygen generates free radicals, reactive oxygen species (ROS), which are mutagenic. In animals, mutational damage to mitochondrial DNA therefore accumulates within the lifespan of the individual. Fertilization generally requires motility of one gamete, and motility requires ATP. It has been proposed that oxidative phosphorylation is nevertheless absent in the special case of quiescent, template mitochondria, that these remain sequestered in oocytes and female germ lines and that oocyte mitochondrial DNA is thus protected from damage, but evidence to support that view has hitherto been lacking. Here we show that female gametes of Aurelia aurita, the common jellyfish, do not transcribe mitochondrial DNA, lack electron transport, and produce no free radicals. In contrast, male gametes actively transcribe mitochondrial genes for respiratory chain components and produce ROS. Electron microscopy shows that this functional division of labour between sperm and egg is accompanied by contrasting mitochondrial morphology. We suggest that mitochondrial anisogamy underlies division of any animal species into two sexes with complementary roles in sexual reproduction. We predict that quiescent oocyte mitochondria contain DNA as an unexpressed template that avoids mutational accumulation by being transmitted through the female germ line. The active descendants of oocyte mitochondria perform oxidative phosphorylation in somatic cells and in male gametes of each new generation, and the mutations that they accumulated are not inherited. We propose that the avoidance of ROS-dependent mutation is the evolutionary pressure underlying maternal mitochondrial inheritance and the developmental origin of the female germ line.


Assuntos
Envelhecimento , DNA Mitocondrial/genética , Metabolismo Energético/fisiologia , Oócitos/metabolismo , Cifozoários/fisiologia , Animais , Feminino , Fertilização , Regulação da Expressão Gênica/fisiologia , Masculino , Potencial da Membrana Mitocondrial , Mitocôndrias/ultraestrutura , Oócitos/citologia , Espécies Reativas de Oxigênio
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...