Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 614(7946): 175-181, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36482135

RESUMEN

Mitochondrial ribosomes (mitoribosomes) synthesize proteins encoded within the mitochondrial genome that are assembled into oxidative phosphorylation complexes. Thus, mitoribosome biogenesis is essential for ATP production and cellular metabolism1. Here we used cryo-electron microscopy to determine nine structures of native yeast and human mitoribosomal small subunit assembly intermediates, illuminating the mechanistic basis for how GTPases are used to control early steps of decoding centre formation, how initial rRNA folding and processing events are mediated, and how mitoribosomal proteins have active roles during assembly. Furthermore, this series of intermediates from two species with divergent mitoribosomal architecture uncovers both conserved principles and species-specific adaptations that govern the maturation of mitoribosomal small subunits in eukaryotes. By revealing the dynamic interplay between assembly factors, mitoribosomal proteins and rRNA that are required to generate functional subunits, our structural analysis provides a vignette for how molecular complexity and diversity can evolve in large ribonucleoprotein assemblies.


Asunto(s)
Microscopía por Crioelectrón , Ribosomas Mitocondriales , Ribonucleoproteínas , Subunidades Ribosómicas Pequeñas , Saccharomyces cerevisiae , Humanos , Proteínas Mitocondriales/química , Proteínas Mitocondriales/metabolismo , Proteínas Mitocondriales/ultraestructura , Ribosomas Mitocondriales/química , Ribosomas Mitocondriales/metabolismo , Ribosomas Mitocondriales/ultraestructura , Proteínas Ribosómicas/química , Proteínas Ribosómicas/metabolismo , Proteínas Ribosómicas/ultraestructura , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/metabolismo , ARN Ribosómico , GTP Fosfohidrolasas , Ribonucleoproteínas/química , Ribonucleoproteínas/metabolismo , Ribonucleoproteínas/ultraestructura , Proteínas Fúngicas/química , Proteínas Fúngicas/metabolismo , Proteínas Fúngicas/ultraestructura , Subunidades Ribosómicas Pequeñas/química , Subunidades Ribosómicas Pequeñas/metabolismo , Subunidades Ribosómicas Pequeñas/ultraestructura
2.
Nature ; 590(7844): 163-169, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33408415

RESUMEN

The mitochondrial outer membrane contains so-called ß-barrel proteins, which allow communication between the cytosol and the mitochondrial interior1-3. Insertion of ß-barrel proteins into the outer membrane is mediated by the multisubunit mitochondrial sorting and assembly machinery (SAM, also known as TOB)4-6. Here we use cryo-electron microscopy to determine the structures of two different forms of the yeast SAM complex at a resolution of 2.8-3.2 Å. The dimeric complex contains two copies of the ß-barrel channel protein Sam50-Sam50a and Sam50b-with partially open lateral gates. The peripheral membrane proteins Sam35 and Sam37 cap the Sam50 channels from the cytosolic side, and are crucial for the structural and functional integrity of the dimeric complex. In the second complex, Sam50b is replaced by the ß-barrel protein Mdm10. In cooperation with Sam50a, Sam37 recruits and traps Mdm10 by penetrating the interior of its laterally closed ß-barrel from the cytosolic side. The substrate-loaded SAM complex contains one each of Sam50, Sam35 and Sam37, but neither Mdm10 nor a second Sam50, suggesting that Mdm10 and Sam50b function as placeholders for a ß-barrel substrate released from Sam50a. Our proposed mechanism for dynamic switching of ß-barrel subunits and substrate explains how entire precursor proteins can fold in association with the mitochondrial machinery for ß-barrel assembly.


Asunto(s)
Microscopía por Crioelectrón , Mitocondrias/química , Mitocondrias/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/ultraestructura , Mitocondrias/ultraestructura , Proteínas Mitocondriales/química , Proteínas Mitocondriales/metabolismo , Proteínas Mitocondriales/ultraestructura , Modelos Moleculares , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Multimerización de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Saccharomyces cerevisiae/ultraestructura , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/ultraestructura
3.
Nature ; 571(7765): 429-433, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31292547

RESUMEN

Balanced fusion and fission are key for the proper function and physiology of mitochondria1,2. Remodelling of the mitochondrial inner membrane is mediated by the dynamin-like protein mitochondrial genome maintenance 1 (Mgm1) in fungi or the related protein optic atrophy 1 (OPA1) in animals3-5. Mgm1 is required for the preservation of mitochondrial DNA in yeast6, whereas mutations in the OPA1 gene in humans are a common cause of autosomal dominant optic atrophy-a genetic disorder that affects the optic nerve7,8. Mgm1 and OPA1 are present in mitochondria as a membrane-integral long form and a short form that is soluble in the intermembrane space. Yeast strains that express temperature-sensitive mutants of Mgm19,10 or mammalian cells that lack OPA1 display fragmented mitochondria11,12, which suggests that Mgm1 and OPA1 have an important role in inner-membrane fusion. Consistently, only the mitochondrial outer membrane-not the inner membrane-fuses in the absence of functional Mgm113. Mgm1 and OPA1 have also been shown to maintain proper cristae architecture10,14; for example, OPA1 prevents the release of pro-apoptotic factors by tightening crista junctions15. Finally, the short form of OPA1 localizes to mitochondrial constriction sites, where it presumably promotes mitochondrial fission16. How Mgm1 and OPA1 perform their diverse functions in membrane fusion, scission and cristae organization is at present unknown. Here we present crystal and electron cryo-tomography structures of Mgm1 from Chaetomium thermophilum. Mgm1 consists of a GTPase (G) domain, a bundle signalling element domain, a stalk, and a paddle domain that contains a membrane-binding site. Biochemical and cell-based experiments demonstrate that the Mgm1 stalk mediates the assembly of bent tetramers into helical filaments. Electron cryo-tomography studies of Mgm1-decorated lipid tubes and fluorescence microscopy experiments on reconstituted membrane tubes indicate how the tetramers assemble on positively or negatively curved membranes. Our findings convey how Mgm1 and OPA1 filaments dynamically remodel the mitochondrial inner membrane.


Asunto(s)
Chaetomium/química , Microscopía por Crioelectrón , Proteínas Fúngicas/química , Proteínas Fúngicas/metabolismo , Proteínas de Unión al GTP/química , Membranas Mitocondriales/metabolismo , Proteínas Mitocondriales/química , Cristalografía por Rayos X , Proteínas Fúngicas/ultraestructura , Proteínas de Unión al GTP/metabolismo , Proteínas de Unión al GTP/ultraestructura , Galactosilceramidas/metabolismo , Proteínas Mitocondriales/metabolismo , Proteínas Mitocondriales/ultraestructura , Modelos Moleculares , Dominios Proteicos , Multimerización de Proteína
4.
Proc Natl Acad Sci U S A ; 119(32): e2207459119, 2022 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-35914129

RESUMEN

Twinkle is the mammalian helicase vital for replication and integrity of mitochondrial DNA. Over 90 Twinkle helicase disease variants have been linked to progressive external ophthalmoplegia and ataxia neuropathies among other mitochondrial diseases. Despite the biological and clinical importance, Twinkle represents the only remaining component of the human minimal mitochondrial replisome that has yet to be structurally characterized. Here, we present 3-dimensional structures of human Twinkle W315L. Employing cryo-electron microscopy (cryo-EM), we characterize the oligomeric assemblies of human full-length Twinkle W315L, define its multimeric interface, and map clinical variants associated with Twinkle in inherited mitochondrial disease. Cryo-EM, crosslinking-mass spectrometry, and molecular dynamics simulations provide insight into the dynamic movement and molecular consequences of the W315L clinical variant. Collectively, this ensemble of structures outlines a framework for studying Twinkle function in mitochondrial DNA replication and associated disease states.


Asunto(s)
Microscopía por Crioelectrón , ADN Helicasas , Enfermedades Mitocondriales , Proteínas Mitocondriales , Multimerización de Proteína , ADN Helicasas/química , ADN Helicasas/genética , ADN Helicasas/metabolismo , ADN Helicasas/ultraestructura , Replicación del ADN , ADN Mitocondrial/biosíntesis , Humanos , Espectrometría de Masas , Enfermedades Mitocondriales/genética , Proteínas Mitocondriales/química , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Proteínas Mitocondriales/ultraestructura , Simulación de Dinámica Molecular , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas Mutantes/ultraestructura
5.
Nature ; 558(7710): 401-405, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29899447

RESUMEN

Mitochondrial inheritance, genome maintenance and metabolic adaptation depend on organelle fission by dynamin-related protein 1 (DRP1) and its mitochondrial receptors. DRP1 receptors include the paralogues mitochondrial dynamics proteins of 49 and 51 kDa (MID49 and MID51) and mitochondrial fission factor (MFF); however, the mechanisms by which these proteins recruit and regulate DRP1 are unknown. Here we present a cryo-electron microscopy structure of full-length human DRP1 co-assembled with MID49 and an analysis of structure- and disease-based mutations. We report that GTP induces a marked elongation and rotation of the GTPase domain, bundle-signalling element and connecting hinge loops of DRP1. In this conformation, a network of multivalent interactions promotes the polymerization of a linear DRP1 filament with MID49 or MID51. After co-assembly, GTP hydrolysis and exchange lead to MID receptor dissociation, filament shortening and curling of DRP1 oligomers into constricted and closed rings. Together, these views of full-length, receptor- and nucleotide-bound conformations reveal how DRP1 performs mechanical work through nucleotide-driven allostery.


Asunto(s)
Proteínas Quinasas Asociadas a Muerte Celular/metabolismo , Proteínas Quinasas Asociadas a Muerte Celular/ultraestructura , Proteínas Mitocondriales/metabolismo , Proteínas Mitocondriales/ultraestructura , Factores de Elongación de Péptidos/metabolismo , Factores de Elongación de Péptidos/ultraestructura , Regulación Alostérica , Sitios de Unión/genética , Microscopía por Crioelectrón , Proteínas Quinasas Asociadas a Muerte Celular/química , Proteínas Quinasas Asociadas a Muerte Celular/genética , GTP Fosfohidrolasas/química , GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/metabolismo , GTP Fosfohidrolasas/ultraestructura , Guanosina Trifosfato/metabolismo , Humanos , Hidrólisis , Proteínas Mitocondriales/química , Modelos Moleculares , Mutación , Factores de Elongación de Péptidos/química , Fosforilación , Dominios Proteicos , Rotación , Relación Estructura-Actividad
6.
Nature ; 560(7717): 263-267, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30089917

RESUMEN

Mitochondria maintain their own specialized protein synthesis machinery, which in mammals is used exclusively for the synthesis of the membrane proteins responsible for oxidative phosphorylation1,2. The initiation of protein synthesis in mitochondria differs substantially from bacterial or cytosolic translation systems. Mitochondrial translation initiation lacks initiation factor 1, which is essential in all other translation systems from bacteria to mammals3,4. Furthermore, only one type of methionyl transfer RNA (tRNAMet) is used for both initiation and elongation4,5, necessitating that the initiation factor specifically recognizes the formylated version of tRNAMet (fMet-tRNAMet). Lastly, most mitochondrial mRNAs do not possess 5' leader sequences to promote mRNA binding to the ribosome2. There is currently little mechanistic insight into mammalian mitochondrial translation initiation, and it is not clear how mRNA engagement, initiator-tRNA recruitment and start-codon selection occur. Here we determine the cryo-EM structure of the complete translation initiation complex from mammalian mitochondria at 3.2 Å. We describe the function of an additional domain insertion that is present in the mammalian mitochondrial initiation factor 2 (mtIF2). By closing the decoding centre, this insertion stabilizes the binding of leaderless mRNAs and induces conformational changes in the rRNA nucleotides involved in decoding. We identify unique features of mtIF2 that are required for specific recognition of fMet-tRNAMet and regulation of its GTPase activity. Finally, we observe that the ribosomal tunnel in the initiating ribosome is blocked by insertion of the N-terminal portion of mitochondrial protein mL45, which becomes exposed as the ribosome switches to elongation mode and may have an additional role in targeting of mitochondrial ribosomes to the protein-conducting pore in the inner mitochondrial membrane.


Asunto(s)
Microscopía por Crioelectrón , Mamíferos , Mitocondrias/ultraestructura , Iniciación de la Cadena Peptídica Traduccional , Animales , Codón Iniciador/genética , Factores Eucarióticos de Iniciación/química , Factores Eucarióticos de Iniciación/genética , Factores Eucarióticos de Iniciación/metabolismo , Factores Eucarióticos de Iniciación/ultraestructura , Mitocondrias/química , Mitocondrias/genética , Mitocondrias/metabolismo , Proteínas Mitocondriales/química , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Proteínas Mitocondriales/ultraestructura , Modelos Moleculares , ARN Mitocondrial/química , ARN Mitocondrial/genética , ARN Mitocondrial/metabolismo , ARN Mitocondrial/ultraestructura , ARN de Transferencia de Metionina/genética , ARN de Transferencia de Metionina/metabolismo , ARN de Transferencia de Metionina/ultraestructura
7.
J Biol Chem ; 296: 100474, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33640456

RESUMEN

Respiratory complex I (NADH:ubiquinone oxidoreductase), the first enzyme of the electron-transport chain, captures the free energy released by NADH oxidation and ubiquinone reduction to translocate protons across an energy-transducing membrane and drive ATP synthesis during oxidative phosphorylation. The cofactor that transfers the electrons directly to ubiquinone is an iron-sulfur cluster (N2) located in the NDUFS2/NUCM subunit. A nearby arginine residue (R121), which forms part of the second coordination sphere of the N2 cluster, is known to be posttranslationally dimethylated but its functional and structural significance are not known. Here, we show that mutations of this arginine residue (R121M/K) abolish the quinone-reductase activity, concomitant with disappearance of the N2 signature from the electron paramagnetic resonance (EPR) spectrum. Analysis of the cryo-EM structure of NDUFS2-R121M complex I at 3.7 Å resolution identified the absence of the cubane N2 cluster as the cause of the dysfunction, within an otherwise intact enzyme. The mutation further induced localized disorder in nearby elements of the quinone-binding site, consistent with the close connections between the cluster and substrate-binding regions. Our results demonstrate that R121 is required for the formation and/or stability of the N2 cluster and highlight the importance of structural analyses for mechanistic interpretation of biochemical and spectroscopic data on complex I variants.


Asunto(s)
Complejo I de Transporte de Electrón/química , Proteínas Fúngicas/química , Proteínas Hierro-Azufre/química , Proteínas Mitocondriales/química , Yarrowia/enzimología , Complejo I de Transporte de Electrón/genética , Complejo I de Transporte de Electrón/metabolismo , Complejo I de Transporte de Electrón/ultraestructura , Proteínas Fúngicas/genética , Proteínas Fúngicas/ultraestructura , Proteínas Hierro-Azufre/genética , Proteínas Hierro-Azufre/metabolismo , Proteínas Hierro-Azufre/ultraestructura , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Proteínas Mitocondriales/ultraestructura , Estabilidad Proteica , Yarrowia/genética
8.
Hum Mol Genet ; 28(7): 1090-1099, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30496414

RESUMEN

TWINKLE is the helicase involved in replication and maintenance of mitochondrial DNA (mtDNA) in mammalian cells. Structurally, TWINKLE is closely related to the bacteriophage T7 gp4 protein and comprises a helicase and primase domain joined by a flexible linker region. Mutations in and around this linker region are responsible for autosomal dominant progressive external ophthalmoplegia (adPEO), a neuromuscular disorder associated with deletions in mtDNA. The underlying molecular basis of adPEO-causing mutations remains unclear, but defects in TWINKLE oligomerization are thought to play a major role. In this study, we have characterized these disease variants by single-particle electron microscopy and can link the diminished activities of the TWINKLE variants to altered oligomeric properties. Our results suggest that the mutations can be divided into those that (i) destroy the flexibility of the linker region, (ii) inhibit ring closure and (iii) change the number of subunits within a helicase ring. Furthermore, we demonstrate that wild-type TWINKLE undergoes large-scale conformational changes upon nucleoside triphosphate binding and that this ability is lost in the disease-causing variants. This represents a substantial advancement in the understanding of the molecular basis of adPEO and related pathologies and may aid in the development of future therapeutic strategies.


Asunto(s)
ADN Helicasas/genética , ADN Helicasas/ultraestructura , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/ultraestructura , Oftalmoplejía Externa Progresiva Crónica/genética , Secuencia de Aminoácidos , ADN Primasa , ADN Mitocondrial/genética , ADN Mitocondrial/fisiología , Humanos , Microscopía Electrónica/métodos , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Mutación/genética , Dominios Proteicos/genética
9.
Am J Med Genet A ; 185(1): 203-207, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33037779

RESUMEN

Inherited optic neuropathies (IONs) are neurodegenerative disorders characterized by optic atrophy with or without extraocular manifestations. Optic atrophy-10 (OPA10) is an autosomal recessive ION recently reported to be caused by mutations in RTN4IP1, which encodes reticulon 4 interacting protein 1 (RTN4IP1), a mitochondrial ubiquinol oxydo-reductase. Here we report novel compound heterozygous mutations in RTN4IP1 in a male proband with developmental delay, epilepsy, optic atrophy, ataxia, and choreoathetosis. Workup was notable for transiently elevated lactate and lactate-to-pyruvate ratio, brain magnetic resonance imaging with optic atrophy and T2 signal abnormalities, and a nondiagnostic initial genetic workup, including chromosomal microarray and mitochondrial panel testing. Exome sequencing identified a paternally inherited missense variant (c.263T>G, p.Val88Gly) predicted to be deleterious and a maternally inherited deletion encompassing RTN4IP1. To our knowledge, this is the first report of a non-single nucleotide pathogenic variant associated with OPA10. This case highlights the expanding phenotypic spectrum of OPA10, the association between "syndromic" cases and severe RTN4IP1 mutations, and the importance of nonbiased genetic testing, such as ES, to analyze multiple genes and variants types, in patients suspected of having genetic disease.


Asunto(s)
Proteínas Portadoras/genética , Discapacidades del Desarrollo/genética , Epilepsia/genética , Proteínas Mitocondriales/genética , Atrofia Óptica/genética , Ataxia/diagnóstico por imagen , Ataxia/genética , Ataxia/patología , Proteínas Portadoras/ultraestructura , Preescolar , Discapacidades del Desarrollo/diagnóstico por imagen , Discapacidades del Desarrollo/patología , Epilepsia/diagnóstico por imagen , Epilepsia/patología , Exoma/genética , Femenino , Predisposición Genética a la Enfermedad , Pruebas Genéticas/métodos , Humanos , Lactante , Recién Nacido , Imagen por Resonancia Magnética , Masculino , Proteínas Mitocondriales/ultraestructura , Mutación/genética , Atrofia Óptica/diagnóstico por imagen , Atrofia Óptica/patología , Linaje , Conformación Proteica , Relación Estructura-Actividad , Secuenciación del Exoma
10.
Proc Natl Acad Sci U S A ; 115(47): 11958-11963, 2018 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-30397129

RESUMEN

Biotransformation enzymes ensure a viable homeostasis by regulating reversible cycles of oxidative and reductive reactions. The metabolism of nitrogen-containing compounds is of high pharmaceutical and toxicological relevance because N-oxygenated metabolites derived from reactions mediated by cytochrome P450 enzymes or flavin-dependent monooxygenases are in some cases highly toxic or mutagenic. The molybdenum-dependent mitochondrial amidoxime-reducing component (mARC) was found to be an extremely efficient counterpart, which is able to reduce the full range of N-oxygenated compounds and thereby mediates detoxification reactions. However, the 3D structure of this enzyme was unknown. Here we present the high-resolution crystal structure of human mARC. We give detailed insight into the coordination of its molybdenum cofactor (Moco), the catalytic mechanism, and its ability to reduce a wide range of N-oxygenated compounds. The identification of two key residues will allow future discrimination between mARC paralogs and ensure correct annotation. Since our structural findings contradict in silico predictions that are currently made by online databases, we propose domain definitions for members of the superfamily of Moco sulfurase C-terminal (MOSC) domain-containing proteins. Furthermore, we present evidence for an evolutionary role of mARC for the emergence of the xanthine oxidase protein superfamily. We anticipate the hereby presented crystal structure to be a starting point for future descriptions of MOSC proteins, which are currently poorly structurally characterized.


Asunto(s)
Proteínas Mitocondriales/química , Proteínas Mitocondriales/ultraestructura , Oxidorreductasas/química , Oxidorreductasas/ultraestructura , Catálisis , Coenzimas , Cristalografía por Rayos X/métodos , Células Eucariotas/metabolismo , Humanos , Metaloproteínas , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Molibdeno/metabolismo , Cofactores de Molibdeno , Oxidación-Reducción , Oxidorreductasas/metabolismo , Estructura Terciaria de Proteína , Pteridinas
11.
Int J Mol Sci ; 22(9)2021 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-34063696

RESUMEN

Multiple mitochondrial dysfunctions syndrome (MMDS) is a rare neurodegenerative disorder associated with mutations in genes with a vital role in the biogenesis of mitochondrial [4Fe-4S] proteins. Mutations in one of these genes encoding for BOLA3 protein lead to MMDS type 2 (MMDS2). Recently, a novel phenotype for MMDS2 with complete clinical recovery was observed in a patient containing a novel variant (c.176G > A, p.Cys59Tyr) in compound heterozygosity. In this work, we aimed to rationalize this unique phenotype observed in MMDS2. To do so, we first investigated the structural impact of the Cys59Tyr mutation on BOLA3 by NMR, and then we analyzed how the mutation affects both the formation of a hetero-complex between BOLA3 and its protein partner GLRX5 and the iron-sulfur cluster-binding properties of the hetero-complex by various spectroscopic techniques and by experimentally driven molecular docking. We show that (1) the mutation structurally perturbed the iron-sulfur cluster-binding region of BOLA3, but without abolishing [2Fe-2S]2+ cluster-binding on the hetero-complex; (2) tyrosine 59 did not replace cysteine 59 as iron-sulfur cluster ligand; and (3) the mutation promoted the formation of an aberrant apo C59Y BOLA3-GLRX5 complex. All these aspects allowed us to rationalize the unique phenotype observed in MMDS2 caused by Cys59Tyr mutation.


Asunto(s)
Glutarredoxinas/genética , Mitocondrias/genética , Enfermedades Mitocondriales/genética , Proteínas Mitocondriales/genética , Cisteína/genética , Glutarredoxinas/ultraestructura , Humanos , Proteínas Hierro-Azufre/genética , Mitocondrias/metabolismo , Enfermedades Mitocondriales/metabolismo , Proteínas Mitocondriales/ultraestructura , Simulación del Acoplamiento Molecular , Complejos Multiproteicos , Mutación , Resonancia Magnética Nuclear Biomolecular , Fenotipo
12.
Nature ; 505(7484): 515-9, 2014 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-24362565

RESUMEN

Mitochondrial ribosomes synthesize a number of highly hydrophobic proteins encoded on the genome of mitochondria, the organelles in eukaryotic cells that are responsible for energy conversion by oxidative phosphorylation. The ribosomes in mammalian mitochondria have undergone massive structural changes throughout their evolution, including ribosomal RNA shortening and acquisition of mitochondria-specific ribosomal proteins. Here we present the three-dimensional structure of the 39S large subunit of the porcine mitochondrial ribosome determined by cryo-electron microscopy at 4.9 Å resolution. The structure, combined with data from chemical crosslinking and mass spectrometry experiments, reveals the unique features of the 39S subunit at near-atomic resolution and provides detailed insight into the architecture of the polypeptide exit site. This region of the mitochondrial ribosome has been considerably remodelled compared to its bacterial counterpart, providing a specialized platform for the synthesis and membrane insertion of the highly hydrophobic protein components of the respiratory chain.


Asunto(s)
Mitocondrias/química , Subunidades Ribosómicas/química , Animales , Bovinos , Microscopía por Crioelectrón , Interacciones Hidrofóbicas e Hidrofílicas , Espectrometría de Masas , Mitocondrias/ultraestructura , Proteínas Mitocondriales/química , Proteínas Mitocondriales/ultraestructura , Modelos Moleculares , Conformación de Ácido Nucleico , Conformación Proteica , ARN Ribosómico 16S/química , ARN Ribosómico 16S/ultraestructura , Proteínas Ribosómicas/química , Proteínas Ribosómicas/ultraestructura , Subunidades Ribosómicas/ultraestructura , Porcinos
13.
Nature ; 515(7526): 283-6, 2014 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-25271403

RESUMEN

Mitochondrial ribosomes (mitoribosomes) are extensively modified ribosomes of bacterial descent specialized for the synthesis and insertion of membrane proteins that are critical for energy conversion and ATP production inside mitochondria. Mammalian mitoribosomes, which comprise 39S and 28S subunits, have diverged markedly from the bacterial ribosomes from which they are derived, rendering them unique compared to bacterial, eukaryotic cytosolic and fungal mitochondrial ribosomes. We have previously determined at 4.9 Å resolution the architecture of the porcine (Sus scrofa) 39S subunit, which is highly homologous to the human mitoribosomal large subunit. Here we present the complete atomic structure of the porcine 39S large mitoribosomal subunit determined in the context of a stalled translating mitoribosome at 3.4 Å resolution by cryo-electron microscopy and chemical crosslinking/mass spectrometry. The structure reveals the locations and the detailed folds of 50 mitoribosomal proteins, shows the highly conserved mitoribosomal peptidyl transferase active site in complex with its substrate transfer RNAs, and defines the path of the nascent chain in mammalian mitoribosomes along their idiosyncratic exit tunnel. Furthermore, we present evidence that a mitochondrial tRNA has become an integral component of the central protuberance of the 39S subunit where it architecturally substitutes for the absence of the 5S ribosomal RNA, a ubiquitous component of all cytoplasmic ribosomes.


Asunto(s)
Mitocondrias/química , Proteínas Mitocondriales/química , Proteínas Mitocondriales/ultraestructura , Subunidades Ribosómicas Grandes/química , Subunidades Ribosómicas Grandes/ultraestructura , Animales , Reactivos de Enlaces Cruzados , Microscopía por Crioelectrón , Espectrometría de Masas , Mitocondrias/ultraestructura , Proteínas Mitocondriales/metabolismo , Modelos Moleculares , Conformación Molecular , Peptidil Transferasas/metabolismo , ARN Ribosómico/química , ARN Ribosómico/metabolismo , ARN Ribosómico/ultraestructura , Subunidades Ribosómicas Grandes/genética , Sus scrofa/genética
14.
Biol Chem ; 401(1): 117-129, 2019 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-31513529

RESUMEN

Biogenesis and function of mitochondria depend on the import of about 1000 precursor proteins that are produced on cytosolic ribosomes. The translocase of the outer membrane (TOM) forms the entry gate for most proteins. After passage through the TOM channel, dedicated preprotein translocases sort the precursor proteins into the mitochondrial subcompartments. Many proteins have to be assembled into oligomeric membrane-integrated complexes in order to perform their functions. In this review, we discuss a dual role of mitochondrial preprotein translocases in protein translocation and oligomeric assembly, focusing on the biogenesis of the TOM complex and the respiratory chain. The sorting and assembly machinery (SAM) of the outer mitochondrial membrane forms a dynamic platform for coupling transport and assembly of TOM subunits. The biogenesis of the cytochrome c oxidase of the inner membrane involves a molecular circuit to adjust translation of mitochondrial-encoded core subunits to the availability of nuclear-encoded partner proteins. Thus, mitochondrial protein translocases not only import precursor proteins but can also support their assembly into functional complexes.


Asunto(s)
Proteínas Portadoras/genética , Mitocondrias/genética , Proteínas de Transporte de Membrana Mitocondrial/genética , Proteínas Mitocondriales/genética , Proteínas Portadoras/ultraestructura , Mitocondrias/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/biosíntesis , Membranas Mitocondriales/metabolismo , Membranas Mitocondriales/ultraestructura , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales , Proteínas Mitocondriales/biosíntesis , Proteínas Mitocondriales/ultraestructura , Biosíntesis de Proteínas/genética , Transporte de Proteínas/genética , Ribosomas/genética , Ribosomas/metabolismo , Saccharomyces cerevisiae/genética
15.
Biochem Biophys Res Commun ; 500(1): 2-8, 2018 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-28676393

RESUMEN

The shape and position of mitochondria are intimately connected to both mitochondrial and cellular function. Mitochondrial anchors play a central role in mitochondrial positioning by exerting spatial, temporal, and contextual control over the cellular position of the organelle. Investigations into the molecular mechanisms of mitochondrial anchoring are still in the early stages, and we are beginning to appreciate the number and variety of anchors that exist. From the insight gained thus far, it is clear that mitochondrial anchoring has functional and physiological consequences that extend beyond mitochondrial positioning to other critical cellular processes.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Mitocondrias/metabolismo , Enfermedades Mitocondriales/metabolismo , Membranas Mitocondriales/metabolismo , Proteínas Mitocondriales/metabolismo , Animales , Senescencia Celular/genética , Proteínas del Citoesqueleto/química , Proteínas del Citoesqueleto/ultraestructura , Drosophila melanogaster/embriología , Drosophila melanogaster/metabolismo , Drosophila melanogaster/ultraestructura , Embrión no Mamífero , Expresión Génica , Humanos , Mitocondrias/ultraestructura , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/patología , Membranas Mitocondriales/ultraestructura , Proteínas Mitocondriales/química , Proteínas Mitocondriales/ultraestructura , Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/ultraestructura , Células Madre/metabolismo , Células Madre/ultraestructura , Toxoplasma/metabolismo , Toxoplasma/ultraestructura
16.
J Biol Chem ; 291(1): 493-507, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26578513

RESUMEN

Multiple isoforms of the mitochondrial fission GTPase dynamin-related protein 1 (Drp1) arise from the alternative splicing of its single gene-encoded pre-mRNA transcript. Among these, the longer Drp1 isoforms, expressed selectively in neurons, bear unique polypeptide sequences within their GTPase and variable domains, known as the A-insert and the B-insert, respectively. Their functions remain unresolved. A comparison of the various biochemical and biophysical properties of the neuronally expressed isoforms with that of the ubiquitously expressed, and shortest, Drp1 isoform (Drp1-short) has revealed the effect of these inserts on Drp1 function. Utilizing various biochemical, biophysical, and cellular approaches, we find that the A- and B-inserts distinctly alter the oligomerization propensity of Drp1 in solution as well as the preferred curvature of helical Drp1 self-assembly on membranes. Consequently, these sequences also suppress Drp1 cooperative GTPase activity. Mitochondrial fission factor (Mff), a tail-anchored membrane protein of the mitochondrial outer membrane that recruits Drp1 to sites of ensuing fission, differentially stimulates the disparate Drp1 isoforms and alleviates the autoinhibitory effect imposed by these sequences on Drp1 function. Moreover, the differential stimulatory effects of Mff on Drp1 isoforms are dependent on the mitochondrial lipid, cardiolipin (CL). Although Mff stimulation of the intrinsically cooperative Drp1-short isoform is relatively modest, CL-independent, and even counter-productive at high CL concentrations, Mff stimulation of the much less cooperative longest Drp1 isoform (Drp1-long) is robust and occurs synergistically with increasing CL content. Thus, membrane-anchored Mff differentially regulates various Drp1 isoforms by functioning as an allosteric effector of cooperative GTPase activity.


Asunto(s)
Dinaminas/genética , GTP Fosfohidrolasas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Mitocondriales/metabolismo , Empalme del ARN/genética , Animales , Cardiolipinas/metabolismo , Membrana Celular/metabolismo , Dinaminas/metabolismo , GTP Fosfohidrolasas/química , GTP Fosfohidrolasas/ultraestructura , Guanosina Trifosfato/metabolismo , Humanos , Hidrólisis , Cinética , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/química , Proteínas Asociadas a Microtúbulos/ultraestructura , Proteínas Mitocondriales/química , Proteínas Mitocondriales/ultraestructura , Multimerización de Proteína , Estructura Secundaria de Proteína , Ratas
17.
Biochim Biophys Acta ; 1857(7): 980-90, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27040506

RESUMEN

Mammalian complex I is composed of 44 different subunits and its assembly requires at least 13 specific assembly factors. Proper function of the mitochondrial respiratory chain enzyme is of crucial importance for cell survival due to its major participation in energy production and cell signaling. Complex I assembly depends on the coordination of several crucial processes that need to be tightly interconnected and orchestrated by a number of assembly factors. The understanding of complex I assembly evolved from simple sequential concept to the more sophisticated modular assembly model describing a convoluted process. According to this model, the different modules assemble independently and associate afterwards with each other to form the final enzyme. In this review, we aim to unravel the complexity of complex I assembly and provide the latest insights in this fundamental and fascinating process. This article is part of a Special Issue entitled Respiratory complex I, edited by Volker Zickermann and Ulrich Brandt.


Asunto(s)
Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/ultraestructura , Proteínas Mitocondriales/química , Proteínas Mitocondriales/ultraestructura , Modelos Químicos , Simulación de Dinámica Molecular , Animales , Sitios de Unión , Activación Enzimática , Humanos , Unión Proteica , Conformación Proteica
18.
Biochim Biophys Acta ; 1857(7): 1001-14, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26801215

RESUMEN

In the mitochondrial inner membrane, oxidative phosphorylation generates ATP via the operation of several multimeric enzymes. The proton-pumping Complex I (NADH:ubiquinone oxidoreductase) is the first and most complicated enzyme required in this process. Complex I is an L-shaped enzyme consisting of more than 40 subunits, one FMN molecule and eight Fe-S clusters. In recent years, genetic and proteomic analyses of Complex I mutants in various model systems, including plants, have provided valuable insights into the assembly of this multimeric enzyme. Assisted by a number of key players, referred to as "assembly factors", the assembly of Complex I takes place in a sequential and modular manner. Although a number of factors have been identified, their precise function in mediating Complex I assembly still remains to be elucidated. This review summarizes our current knowledge of plant Complex I composition and assembly derived from studies in plant model systems such as Arabidopsis thaliana and Chlamydomonas reinhardtii. Plant Complex I is highly conserved and comprises a significant number of subunits also present in mammalian and fungal Complexes I. Plant Complex I also contains additional subunits absent from the mammalian and fungal counterpart, whose function in enzyme activity and assembly is not clearly understood. While 14 assembly factors have been identified for human Complex I, only two proteins, namely GLDH and INDH, have been established as bona fide assembly factors for plant Complex I. This article is part of a Special Issue entitled Respiratory complex I, edited by Volker Zickermann and Ulrich Brandt.


Asunto(s)
Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/ultraestructura , Proteínas Mitocondriales/química , Proteínas Mitocondriales/ultraestructura , Proteínas de Plantas/química , Proteínas de Plantas/ultraestructura , Sitios de Unión , Activación Enzimática , Modelos Químicos , Simulación de Dinámica Molecular , Unión Proteica , Conformación Proteica
19.
Biochim Biophys Acta ; 1857(7): 902-14, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26921811

RESUMEN

Proton-pumping NADH:ubiquinone oxidoreductase (complex I) is the largest and most complicated enzyme of the respiratory chain. Fourteen central subunits represent the minimal form of complex I and can be assigned to functional modules for NADH oxidation, ubiquinone reduction, and proton pumping. In addition, the mitochondrial enzyme comprises some 30 accessory subunits surrounding the central subunits that are not directly associated with energy conservation. Complex I is known to release deleterious oxygen radicals (ROS) and its dysfunction has been linked to a number of hereditary and degenerative diseases. We here review recent progress in structure determination, and in understanding the role of accessory subunits and functional analysis of mitochondrial complex I. For the central subunits, structures provide insight into the arrangement of functional modules including the substrate binding sites, redox-centers and putative proton channels and pump sites. Only for two of the accessory subunits, detailed structures are available. Nevertheless, many of them could be localized in the overall structure of complex I, but most of these assignments have to be considered tentative. Strikingly, redox reactions and proton pumping machinery are spatially completely separated and the site of reduction for the hydrophobic substrate ubiquinone is found deeply buried in the hydrophilic domain of the complex. The X-ray structure of complex I from Yarrowia lipolytica provides clues supporting the previously proposed two-state stabilization change mechanism, in which ubiquinone redox chemistry induces conformational states and thereby drives proton pumping. The same structural rearrangements may explain the active/deactive transition of complex I implying an integrated mechanistic model for energy conversion and regulation. This article is part of a Special Issue entitled Respiratory complex I, edited by Volker Zickermann and Ulrich Brandt.


Asunto(s)
Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/ultraestructura , Proteínas Mitocondriales/química , Proteínas Mitocondriales/ultraestructura , Bombas de Protones/química , Especies Reactivas de Oxígeno/síntesis química , Secuencia de Aminoácidos , Transporte de Electrón , Activación Enzimática , Modelos Químicos , Simulación de Dinámica Molecular , Datos de Secuencia Molecular , Oxidación-Reducción , Conformación Proteica , Estructura Terciaria de Proteína , Bombas de Protones/ultraestructura , Relación Estructura-Actividad
20.
Biochim Biophys Acta ; 1857(7): 946-57, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26777588

RESUMEN

Mitochondrial complex I (NADH:ubiquinone oxidoreductase) is a key enzyme in cellular energy metabolism and provides approximately 40% of the proton-motive force that is utilized during mitochondrial ATP production. The dysregulation of complex I function--either genetically, pharmacologically, or metabolically induced--has severe pathophysiological consequences that often involve an imbalance in the production of reactive oxygen species (ROS). Slow transition of the active (A) enzyme to the deactive, dormant (D) form takes place during ischemia in metabolically active organs such as the heart and brain. The reactivation of complex I occurs upon reoxygenation of ischemic tissue, a process that is usually accompanied by an increase in cellular ROS production. Complex I in the D-form serves as a protective mechanism preventing the oxidative burst upon reperfusion. Conversely, however, the D-form is more vulnerable to oxidative/nitrosative damage. Understanding the so-called active/deactive (A/D) transition may contribute to the development of new therapeutic interventions for conditions like stroke, cardiac infarction, and other ischemia-associated pathologies. In this review, we summarize current knowledge on the mechanism of A/D transition of mitochondrial complex I considering recently available structural data and site-specific labeling experiments. In addition, this review discusses in detail the impact of the A/D transition on ROS production by complex I and the S-nitrosation of a critical cysteine residue of subunit ND3 as a strategy to prevent oxidative damage and tissue damage during ischemia-reperfusion injury. This article is part of a Special Issue entitled Respiratory complex I, edited by Volker Zickermann and Ulrich Brandt.


Asunto(s)
Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/metabolismo , Isquemia/metabolismo , Proteínas Mitocondriales/química , Proteínas Mitocondriales/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Transporte de Electrón , Activación Enzimática , Humanos , Proteínas Mitocondriales/ultraestructura , Modelos Biológicos , Modelos Químicos , Modelos Moleculares , Oxidación-Reducción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA