Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.840
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Cell ; 170(6): 1247-1257.e12, 2017 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-28844695

RESUMO

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


Assuntos
Complexo de Proteínas da Cadeia de Transporte de Elétrons/química , Complexos Multienzimáticos/química , Microscopia Crioeletrônica , Complexo de Proteínas da Cadeia de Transporte de Elétrons/isolamento & purificação , Complexo de Proteínas da Cadeia de Transporte de Elétrons/metabolismo , Complexo I de Transporte de Elétrons/química , Complexo I de Transporte de Elétrons/isolamento & purificação , Complexo I de Transporte de Elétrons/metabolismo , Complexo II de Transporte de Elétrons/química , Complexo II de Transporte de Elétrons/isolamento & purificação , Complexo II de Transporte de Elétrons/metabolismo , Humanos , Mitocôndrias/química , Mitocôndrias/metabolismo , Modelos Moleculares , Complexos Multienzimáticos/isolamento & purificação , Complexos Multienzimáticos/metabolismo
2.
Annu Rev Biochem ; 84: 631-57, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25839341

RESUMO

Oxygenic photosynthesis is the principal converter of sunlight into chemical energy. Cyanobacteria and plants provide aerobic life with oxygen, food, fuel, fibers, and platform chemicals. Four multisubunit membrane proteins are involved: photosystem I (PSI), photosystem II (PSII), cytochrome b6f (cyt b6f), and ATP synthase (FOF1). ATP synthase is likewise a key enzyme of cell respiration. Over three billion years, the basic machinery of oxygenic photosynthesis and respiration has been perfected to minimize wasteful reactions. The proton-driven ATP synthase is embedded in a proton tight-coupling membrane. It is composed of two rotary motors/generators, FO and F1, which do not slip against each other. The proton-driven FO and the ATP-synthesizing F1 are coupled via elastic torque transmission. Elastic transmission decouples the two motors in kinetic detail but keeps them perfectly coupled in thermodynamic equilibrium and (time-averaged) under steady turnover. Elastic transmission enables operation with different gear ratios in different organisms.


Assuntos
Células Vegetais/enzimologia , ATPases Translocadoras de Prótons/química , ATPases Translocadoras de Prótons/metabolismo , Bactérias/classificação , Bactérias/citologia , Bactérias/enzimologia , Respiração Celular , Cloroplastos/química , Cloroplastos/enzimologia , Cianobactérias/citologia , Cianobactérias/enzimologia , Mitocôndrias/química , Mitocôndrias/enzimologia , Fotossíntese
3.
Nature ; 626(7997): 169-176, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38267577

RESUMO

To coordinate cellular physiology, eukaryotic cells rely on the rapid exchange of molecules at specialized organelle-organelle contact sites1,2. Endoplasmic reticulum-mitochondrial contact sites (ERMCSs) are particularly vital communication hubs, playing key roles in the exchange of signalling molecules, lipids and metabolites3,4. ERMCSs are maintained by interactions between complementary tethering molecules on the surface of each organelle5,6. However, due to the extreme sensitivity of these membrane interfaces to experimental perturbation7,8, a clear understanding of their nanoscale organization and regulation is still lacking. Here we combine three-dimensional electron microscopy with high-speed molecular tracking of a model organelle tether, Vesicle-associated membrane protein (VAMP)-associated protein B (VAPB), to map the structure and diffusion landscape of ERMCSs. We uncovered dynamic subdomains within VAPB contact sites that correlate with ER membrane curvature and undergo rapid remodelling. We show that VAPB molecules enter and leave ERMCSs within seconds, despite the contact site itself remaining stable over much longer time scales. This metastability allows ERMCSs to remodel with changes in the physiological environment to accommodate metabolic needs of the cell. An amyotrophic lateral sclerosis-associated mutation in VAPB perturbs these subdomains, likely impairing their remodelling capacity and resulting in impaired interorganelle communication. These results establish high-speed single-molecule imaging as a new tool for mapping the structure of contact site interfaces and reveal that the diffusion landscape of VAPB at contact sites is a crucial component of ERMCS homeostasis.


Assuntos
Retículo Endoplasmático , Mitocôndrias , Membranas Mitocondriais , Movimento , Proteínas de Transporte Vesicular , Humanos , Esclerose Lateral Amiotrófica/genética , Retículo Endoplasmático/química , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/ultraestrutura , Mitocôndrias/química , Mitocôndrias/metabolismo , Mitocôndrias/ultraestrutura , Membranas Mitocondriais/química , Membranas Mitocondriais/metabolismo , Membranas Mitocondriais/ultraestrutura , Transdução de Sinais , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo , Proteínas de Transporte Vesicular/ultraestrutura , Microscopia Eletrônica , Imageamento Tridimensional , Sítios de Ligação , Difusão , Fatores de Tempo , Mutação , Homeostase
4.
Nature ; 631(8019): 232-239, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38811722

RESUMO

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


Assuntos
Respiração Celular , Complexo III da Cadeia de Transporte de Elétrons , Complexo I de Transporte de Elétrons , Mitocôndrias , Animais , Microscopia Crioeletrônica , Complexo I de Transporte de Elétrons/química , Complexo I de Transporte de Elétrons/metabolismo , Complexo I de Transporte de Elétrons/ultraestrutura , Complexo III da Cadeia de Transporte de Elétrons/química , Complexo III da Cadeia de Transporte de Elétrons/metabolismo , Complexo III da Cadeia de Transporte de Elétrons/ultraestrutura , Mitocôndrias/metabolismo , Mitocôndrias/química , Mitocôndrias/ultraestrutura , Membranas Mitocondriais/metabolismo , Membranas Mitocondriais/química , Membranas Mitocondriais/ultraestrutura , Modelos Moleculares , Fosforilação Oxidativa , Suínos , Ubiquinona/análogos & derivados , Ubiquinona/química , Ubiquinona/metabolismo , Lipídeos de Membrana/química , Lipídeos de Membrana/metabolismo
5.
Cell ; 158(6): 1415-1430, 2014 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-25215496

RESUMO

The manner by which genotype and environment affect complex phenotypes is one of the fundamental questions in biology. In this study, we quantified the transcriptome--a subset of the metabolome--and, using targeted proteomics, quantified a subset of the liver proteome from 40 strains of the BXD mouse genetic reference population on two diverse diets. We discovered dozens of transcript, protein, and metabolite QTLs, several of which linked to metabolic phenotypes. Most prominently, Dhtkd1 was identified as a primary regulator of 2-aminoadipate, explaining variance in fasted glucose and diabetes status in both mice and humans. These integrated molecular profiles also allowed further characterization of complex pathways, particularly the mitochondrial unfolded protein response (UPR(mt)). UPR(mt) shows strikingly variant responses at the transcript and protein level that are remarkably conserved among C. elegans, mice, and humans. Overall, these examples demonstrate the value of an integrated multilayered omics approach to characterize complex metabolic phenotypes.


Assuntos
Perfilação da Expressão Gênica , Fígado/química , Camundongos/metabolismo , Mitocôndrias/química , Proteoma/análise , Soro/química , Animais , Glucose/metabolismo , Humanos , Cetona Oxirredutases/metabolismo , Fígado/citologia , Fígado/metabolismo , Camundongos/classificação , Camundongos/genética , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Mitocôndrias/metabolismo , Locos de Características Quantitativas , Soro/metabolismo , Resposta a Proteínas não Dobradas
6.
Nature ; 620(7976): 1101-1108, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37612504

RESUMO

Distinct morphologies of the mitochondrial network support divergent metabolic and regulatory processes that determine cell function and fate1-3. The mechanochemical GTPase optic atrophy 1 (OPA1) influences the architecture of cristae and catalyses the fusion of the mitochondrial inner membrane4,5. Despite its fundamental importance, the molecular mechanisms by which OPA1 modulates mitochondrial morphology are unclear. Here, using a combination of cellular and structural analyses, we illuminate the molecular mechanisms that are key to OPA1-dependent membrane remodelling and fusion. Human OPA1 embeds itself into cardiolipin-containing membranes through a lipid-binding paddle domain. A conserved loop within the paddle domain inserts deeply into the bilayer, further stabilizing the interactions with cardiolipin-enriched membranes. OPA1 dimerization through the paddle domain promotes the helical assembly of a flexible OPA1 lattice on the membrane, which drives mitochondrial fusion in cells. Moreover, the membrane-bending OPA1 oligomer undergoes conformational changes that pull the membrane-inserting loop out of the outer leaflet and contribute to the mechanics of membrane remodelling. Our findings provide a structural framework for understanding how human OPA1 shapes mitochondrial morphology and show us how human disease mutations compromise OPA1 functions.


Assuntos
GTP Fosfo-Hidrolases , Fusão de Membrana , Mitocôndrias , Membranas Mitocondriais , Humanos , Biocatálise , Cardiolipinas/química , Cardiolipinas/metabolismo , GTP Fosfo-Hidrolases/genética , GTP Fosfo-Hidrolases/metabolismo , Mitocôndrias/química , Mitocôndrias/metabolismo , Membranas Mitocondriais/química , Membranas Mitocondriais/enzimologia , Membranas Mitocondriais/metabolismo , Mutação , Domínios Proteicos , Multimerização Proteica , Dinâmica Mitocondrial
7.
Nature ; 618(7963): 188-192, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37165187

RESUMO

The endoplasmic reticulum and mitochondria are main hubs of eukaryotic membrane biogenesis that rely on lipid exchange via membrane contact sites1-3, but the underpinning mechanisms remain poorly understood. In yeast, tethering and lipid transfer between the two organelles is mediated by the endoplasmic reticulum-mitochondria encounter structure (ERMES), a four-subunit complex of unresolved stoichiometry and architecture4-6. Here we determined the molecular organization of ERMES within Saccharomyces cerevisiae cells using integrative structural biology by combining quantitative live imaging, cryo-correlative microscopy, subtomogram averaging and molecular modelling. We found that ERMES assembles into approximately 25 discrete bridge-like complexes distributed irregularly across a contact site. Each bridge consists of three synaptotagmin-like mitochondrial lipid binding protein domains oriented in a zig-zag arrangement. Our molecular model of ERMES reveals a pathway for lipids. These findings resolve the in situ supramolecular architecture of a major inter-organelle lipid transfer machinery and provide a basis for the mechanistic understanding of lipid fluxes in eukaryotic cells.


Assuntos
Retículo Endoplasmático , Mitocôndrias , Saccharomyces cerevisiae , Retículo Endoplasmático/química , Retículo Endoplasmático/metabolismo , Lipídeos , Mitocôndrias/química , Mitocôndrias/metabolismo , Membranas Mitocondriais/metabolismo , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/citologia , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Modelos Moleculares , Sinaptotagminas/química , Sinaptotagminas/metabolismo
8.
Nature ; 621(7979): 620-626, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37344598

RESUMO

Mitochondria import nearly all of their approximately 1,000-2,000 constituent proteins from the cytosol across their double-membrane envelope1-5. Genetic and biochemical studies have shown that the conserved protein translocase, termed the TIM23 complex, mediates import of presequence-containing proteins (preproteins) into the mitochondrial matrix and inner membrane. Among about ten different subunits of the TIM23 complex, the essential multipass membrane protein Tim23, together with the evolutionarily related protein Tim17, has long been postulated to form a protein-conducting channel6-11. However, the mechanism by which these subunits form a translocation path in the membrane and enable the import process remains unclear due to a lack of structural information. Here we determined the cryo-electron microscopy structure of the core TIM23 complex (heterotrimeric Tim17-Tim23-Tim44) from Saccharomyces cerevisiae. Contrary to the prevailing model, Tim23 and Tim17 themselves do not form a water-filled channel, but instead have separate, lipid-exposed concave cavities that face in opposite directions. Our structural and biochemical analyses show that the cavity of Tim17, but not Tim23, forms the protein translocation path, whereas Tim23 probably has a structural role. The results further suggest that, during translocation of substrate polypeptides, the nonessential subunit Mgr2 seals the lateral opening of the Tim17 cavity to facilitate the translocation process. We propose a new model for the TIM23-mediated protein import and sorting mechanism, a central pathway in mitochondrial biogenesis.


Assuntos
Mitocôndrias , Proteínas do Complexo de Importação de Proteína Precursora Mitocondrial , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Microscopia Crioeletrônica , Proteínas do Complexo de Importação de Proteína Precursora Mitocondrial/química , Proteínas do Complexo de Importação de Proteína Precursora Mitocondrial/metabolismo , Proteínas do Complexo de Importação de Proteína Precursora Mitocondrial/ultraestrutura , Transporte Proteico , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/citologia , Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/ultraestrutura , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/ultraestrutura , Mitocôndrias/química , Mitocôndrias/metabolismo , Mitocôndrias/ultraestrutura
9.
Nature ; 615(7954): 934-938, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36949187

RESUMO

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.


Assuntos
Microscopia Crioeletrônica , Complexo III da Cadeia de Transporte de Elétrons , Complexo II de Transporte de Elétrons , Complexo IV da Cadeia de Transporte de Elétrons , Complexo I de Transporte de Elétrons , Mitocôndrias , Membranas Mitocondriais , Transporte de Elétrons , Complexo III da Cadeia de Transporte de Elétrons/química , Complexo III da Cadeia de Transporte de Elétrons/metabolismo , Complexo III da Cadeia de Transporte de Elétrons/ultraestrutura , Complexo IV da Cadeia de Transporte de Elétrons/química , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Complexo IV da Cadeia de Transporte de Elétrons/ultraestrutura , Mitocôndrias/química , Mitocôndrias/enzimologia , Mitocôndrias/metabolismo , Mitocôndrias/ultraestrutura , Membranas Mitocondriais/química , Membranas Mitocondriais/enzimologia , Membranas Mitocondriais/metabolismo , Membranas Mitocondriais/ultraestrutura , Complexo II de Transporte de Elétrons/química , Complexo II de Transporte de Elétrons/metabolismo , Complexo II de Transporte de Elétrons/ultraestrutura , Complexo I de Transporte de Elétrons/química , Complexo I de Transporte de Elétrons/metabolismo , Complexo I de Transporte de Elétrons/ultraestrutura , Multimerização Proteica , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Simulação de Dinâmica Molecular , Sítios de Ligação , Evolução Molecular
10.
Nat Rev Mol Cell Biol ; 17(2): 69-82, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26627931

RESUMO

The endoplasmic reticulum (ER) is the largest organelle in the cell, and its functions have been studied for decades. The past several years have provided novel insights into the existence of distinct domains between the ER and other organelles, known as membrane contact sites (MCSs). At these contact sites, organelle membranes are closely apposed and tethered, but do not fuse. Here, various protein complexes can work in concert to perform specialized functions such as binding, sensing and transferring molecules, as well as engaging in organelle biogenesis and dynamics. This Review describes the structure and functions of MCSs, primarily focusing on contacts of the ER with mitochondria and endosomes.


Assuntos
Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/ultraestrutura , Células Eucarióticas/metabolismo , Proteínas de Membrana/metabolismo , Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/ultraestrutura , Animais , Transporte Biológico , Cálcio/metabolismo , Membrana Celular/química , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Retículo Endoplasmático/química , Endossomos/química , Endossomos/metabolismo , Endossomos/ultraestrutura , Células Eucarióticas/ultraestrutura , Regulação da Expressão Gênica , Humanos , Metabolismo dos Lipídeos , Proteínas de Membrana/química , Proteínas de Membrana/genética , Mitocôndrias/química , Mitocôndrias/metabolismo , Mitocôndrias/ultraestrutura , Membranas Mitocondriais/química , Membranas Mitocondriais/metabolismo , Membranas Mitocondriais/ultraestrutura , Biogênese de Organelas , Saccharomyces cerevisiae/genética
11.
Cell ; 155(1): 160-71, 2013 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-24055366

RESUMO

Respiratory chain complexes assemble into functional quaternary structures called supercomplexes (RCS) within the folds of the inner mitochondrial membrane, or cristae. Here, we investigate the relationship between respiratory function and mitochondrial ultrastructure and provide evidence that cristae shape determines the assembly and stability of RCS and hence mitochondrial respiratory efficiency. Genetic and apoptotic manipulations of cristae structure affect assembly and activity of RCS in vitro and in vivo, independently of changes to mitochondrial protein synthesis or apoptotic outer mitochondrial membrane permeabilization. We demonstrate that, accordingly, the efficiency of mitochondria-dependent cell growth depends on cristae shape. Thus, RCS assembly emerges as a link between membrane morphology and function.


Assuntos
Respiração Celular , Transporte de Elétrons , Membranas Mitocondriais/fisiologia , Sequência de Aminoácidos , Animais , Apoptose , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/química , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/metabolismo , GTP Fosfo-Hidrolases/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/química , Mitocôndrias/fisiologia , Membranas Mitocondriais/química , Membranas Mitocondriais/ultraestrutura , Dados de Sequência Molecular , Complexos Multiproteicos/metabolismo , Alinhamento de Sequência
12.
Nature ; 606(7914): 603-608, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35676484

RESUMO

Mitoribosomes are essential for the synthesis and maintenance of bioenergetic proteins. Here we use cryo-electron microscopy to determine a series of the small mitoribosomal subunit (SSU) intermediates in complex with auxiliary factors, revealing a sequential assembly mechanism. The methyltransferase TFB1M binds to partially unfolded rRNA h45 that is promoted by RBFA, while the mRNA channel is blocked. This enables binding of METTL15 that promotes further rRNA maturation and a large conformational change of RBFA. The new conformation allows initiation factor mtIF3 to already occupy the subunit interface during the assembly. Finally, the mitochondria-specific ribosomal protein mS37 (ref. 1) outcompetes RBFA to complete the assembly with the SSU-mS37-mtIF3 complex2 that proceeds towards mtIF2 binding and translation initiation. Our results explain how the action of step-specific factors modulate the dynamic assembly of the SSU, and adaptation of a unique protein, mS37, links the assembly to initiation to establish the catalytic human mitoribosome.


Assuntos
Ribossomos Mitocondriais , Subunidades Ribossômicas Menores , Humanos , Microscopia Crioeletrônica , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Fatores de Iniciação em Eucariotos/química , Fatores de Iniciação em Eucariotos/metabolismo , Mitocôndrias/química , Mitocôndrias/metabolismo , Proteínas Mitocondriais/química , Proteínas Mitocondriais/metabolismo , Ribossomos Mitocondriais/química , Ribossomos Mitocondriais/metabolismo , Ribossomos Mitocondriais/ultraestrutura , Proteínas Ribossômicas/química , Proteínas Ribossômicas/metabolismo , Subunidades Ribossômicas Menores/química , Subunidades Ribossômicas Menores/metabolismo , Subunidades Ribossômicas Menores/ultraestrutura , RNA Ribossômico/química , RNA Ribossômico/metabolismo , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/metabolismo , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo
13.
Annu Rev Biochem ; 81: 1-33, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22663076

RESUMO

Good fortune let me be an innocent child during World War II, a hopeful adolescent with encouraging parents during the years of German recovery, and a self-determined adult in a period of peace, freedom, and wealth. My luck continued as a scientist who could entirely follow his fancy. My mind was always set on understanding how things are made. At a certain point, I found myself confronted with the question of how mitochondria and organelles, which cannot be formed de novo, are put together. Intracellular transport of proteins, their translocation across the mitochondrial membranes, and their folding and assembly were the processes that fascinated me. Now, after some 30 years, we have wonderful insights, unimagined views of a complex and at the same time simple machinery and its workings. We have glimpses of how orderly processes are established in the cell to assemble from single molecules our beautiful mitochondria that every day make some 50 kg of ATP for each of us. At the same time, we have learned amazing lessons from the tinkering of evolution that developed mitochondria from bacteria.


Assuntos
Biologia Celular/história , Mitocôndrias/metabolismo , Animais , Alemanha , História do Século XX , Mitocôndrias/química , Mitocôndrias/genética , Proteínas Mitocondriais/metabolismo , Dobramento de Proteína , Transporte Proteico , Proteínas/metabolismo
14.
Nat Methods ; 21(4): 648-656, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38036853

RESUMO

Although StayGold is a bright and highly photostable fluorescent protein, its propensity for obligate dimer formation may hinder applications in molecular fusion and membrane targeting. To attain monovalent as well as bright and photostable labeling, we engineered tandem dimers of StayGold to promote dispersibility. On the basis of the crystal structure of this fluorescent protein, we disrupted the dimerization to generate a monomeric variant that offers improved photostability and brightness compared to StayGold. We applied the new monovalent StayGold tools to live-cell imaging experiments using spinning-disk laser-scanning confocal microscopy or structured illumination microscopy. We achieved cell-wide, high-spatiotemporal resolution and sustained imaging of dynamic subcellular events, including the targeting of endogenous condensin I to mitotic chromosomes, the movement of the Golgi apparatus and its membranous derivatives along microtubule networks, the distribution of cortical filamentous actin and the remolding of cristae membranes within mobile mitochondria.


Assuntos
Complexo de Golgi , Mitocôndrias , Mitocôndrias/química , Complexo de Golgi/metabolismo , Microtúbulos/metabolismo , Microscopia Confocal/métodos
15.
Cell ; 151(7): 1528-41, 2012 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-23260140

RESUMO

Mitochondrial respiratory-chain complexes assemble from subunits of dual genetic origin assisted by specialized assembly factors. Whereas core subunits are translated on mitochondrial ribosomes, others are imported after cytosolic translation. How imported subunits are ushered to assembly intermediates containing mitochondria-encoded subunits is unresolved. Here, we report a comprehensive dissection of early cytochrome c oxidase assembly intermediates containing proteins required for normal mitochondrial translation and reveal assembly factors promoting biogenesis of human respiratory-chain complexes. We find that TIM21, a subunit of the inner-membrane presequence translocase, is also present in the major assembly intermediates containing newly mitochondria-synthesized and imported respiratory-chain subunits, which we term MITRAC complexes. Human TIM21 is dispensable for protein import but required for integration of early-assembling, presequence-containing subunits into respiratory-chain intermediates. We establish an unexpected molecular link between the TIM23 transport machinery and assembly of respiratory-chain complexes that regulate mitochondrial protein synthesis in response to their assembly state.


Assuntos
Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Mitocôndrias/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Proteínas Mitocondriais/metabolismo , Ciclo-Oxigenase 1/genética , Ciclo-Oxigenase 1/metabolismo , Citosol/metabolismo , Humanos , Proteínas de Membrana/química , Proteínas de Membrana Transportadoras/química , Mitocôndrias/química , Mitocôndrias/genética , Proteínas do Complexo de Importação de Proteína Precursora Mitocondrial , Proteínas Mitocondriais/química , Biossíntese de Proteínas
16.
Nature ; 590(7844): 57-66, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33536648

RESUMO

Mitochondria form dynamic networks in the cell that are balanced by the flux of iterative fusion and fission events of the organelles. It is now appreciated that mitochondrial fission also represents an end-point event in a signalling axis that allows cells to sense and respond to external cues. The fission process is orchestrated by membrane-associated adaptors, influenced by organellar and cytoskeletal interactions and ultimately executed by the dynamin-like GTPase DRP1. Here we invoke the framework of the 'mitochondrial divisome', which is conceptually and operationally similar to the bacterial cell-division machinery. We review the functional and regulatory aspects of the mitochondrial divisome and, within this framework, parse the core from the accessory machinery. In so doing, we transition from a phenomenological to a mechanistic understanding of the fission process.


Assuntos
Mitocôndrias/química , Mitocôndrias/metabolismo , Dinâmica Mitocondrial/fisiologia , Animais , Evolução Biológica , Sinalização do Cálcio , Morte Celular , Doença , Dinaminas/química , Dinaminas/genética , Dinaminas/metabolismo , Saúde , Humanos , Mitocôndrias/patologia
17.
Nature ; 591(7851): 659-664, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33658713

RESUMO

Symmetric cell division requires the even partitioning of genetic information and cytoplasmic contents between daughter cells. Whereas the mechanisms coordinating the segregation of the genome are well known, the processes that ensure organelle segregation between daughter cells remain less well understood1. Here we identify multiple actin assemblies with distinct but complementary roles in mitochondrial organization and inheritance in mitosis. First, we find a dense meshwork of subcortical actin cables assembled throughout the mitotic cytoplasm. This network scaffolds the endoplasmic reticulum and organizes three-dimensional mitochondrial positioning to ensure the equal segregation of mitochondrial mass at cytokinesis. Second, we identify a dynamic wave of actin filaments reversibly assembling on the surface of mitochondria during mitosis. Mitochondria sampled by this wave are enveloped within actin clouds that can spontaneously break symmetry to form elongated comet tails. Mitochondrial comet tails promote randomly directed bursts of movement that shuffle mitochondrial position within the mother cell to randomize inheritance of healthy and damaged mitochondria between daughter cells. Thus, parallel mechanisms mediated by the actin cytoskeleton ensure both equal and random inheritance of mitochondria in symmetrically dividing cells.


Assuntos
Actinas/química , Actinas/metabolismo , Mitocôndrias/metabolismo , Mitose , Citoesqueleto de Actina/química , Citoesqueleto de Actina/metabolismo , Animais , Divisão Celular , Linhagem Celular , Citocinese , Retículo Endoplasmático/metabolismo , Hipocampo/citologia , Hipocampo/embriologia , Humanos , Mitocôndrias/química , Neurônios , Ratos
18.
Nature ; 595(7865): 120-124, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34079125

RESUMO

Compartmentalization is a defining characteristic of eukaryotic cells, and partitions distinct biochemical processes into discrete subcellular locations. Microscopy1 and biochemical fractionation coupled with mass spectrometry2-4 have defined the proteomes of a variety of different organelles, but many intracellular compartments have remained refractory to such approaches. Proximity-dependent biotinylation techniques such as BioID provide an alternative approach to define the composition of cellular compartments in living cells5-7. Here we present a BioID-based map of a human cell on the basis of 192 subcellular markers, and define the intracellular locations of 4,145 unique proteins in HEK293 cells. Our localization predictions exceed the specificity of previous approaches, and enabled the discovery of proteins at the interface between the mitochondrial outer membrane and the endoplasmic reticulum that are crucial for mitochondrial homeostasis. On the basis of this dataset, we created humancellmap.org as a community resource that provides online tools for localization analysis of user BioID data, and demonstrate how this resource can be used to understand BioID results better.


Assuntos
Biotinilação , Compartimento Celular , Transporte Proteico , Proteoma/análise , Proteoma/química , Células Cultivadas , Conjuntos de Dados como Assunto , Retículo Endoplasmático/química , Retículo Endoplasmático/metabolismo , Células HEK293 , Células HeLa , Homeostase , Humanos , Espectrometria de Massas , Mitocôndrias/química , Mitocôndrias/metabolismo , Organelas/química , Organelas/metabolismo , Proteoma/metabolismo , Reprodutibilidade dos Testes
19.
Nature ; 590(7844): 163-169, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33408415

RESUMO

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.


Assuntos
Microscopia Crioeletrônica , Mitocôndrias/química , Mitocôndrias/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/citologia , Saccharomyces cerevisiae/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Proteínas de Membrana/ultraestrutura , Mitocôndrias/ultraestrutura , Proteínas Mitocondriais/química , Proteínas Mitocondriais/metabolismo , Proteínas Mitocondriais/ultraestrutura , Modelos Moleculares , Complexos Multiproteicos/química , Complexos Multiproteicos/metabolismo , Multimerização Proteica , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Saccharomyces cerevisiae/ultraestrutura , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/ultraestrutura
20.
Mol Cell ; 74(2): 347-362.e6, 2019 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-30853401

RESUMO

Selective autophagy recycles damaged organelles and clears intracellular pathogens to prevent their aberrant accumulation. How ULK1 kinase is targeted and activated during selective autophagic events remains to be elucidated. In this study, we used chemically inducible dimerization (CID) assays in tandem with CRISPR KO lines to systematically analyze the molecular basis of selective autophagosome biogenesis. We demonstrate that ectopic placement of NDP52 on mitochondria or peroxisomes is sufficient to initiate selective autophagy by focally localizing and activating the ULK1 complex. The capability of NDP52 to induce mitophagy is dependent on its interaction with the FIP200/ULK1 complex, which is facilitated by TBK1. Ectopically tethering ULK1 to cargo bypasses the requirement for autophagy receptors and TBK1. Focal activation of ULK1 occurs independently of AMPK and mTOR. Our findings provide a parsimonious model of selective autophagy, which highlights the coordination of ULK1 complex localization by autophagy receptors and TBK1 as principal drivers of targeted autophagosome biogenesis.


Assuntos
Proteína Homóloga à Proteína-1 Relacionada à Autofagia/genética , Autofagia/genética , Proteínas Nucleares/genética , Proteínas Serina-Treonina Quinases/genética , Quinases Proteína-Quinases Ativadas por AMP , Proteínas Relacionadas à Autofagia , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Células HeLa , Humanos , Mitocôndrias/química , Mitocôndrias/genética , Complexos Multiproteicos/química , Complexos Multiproteicos/genética , Peroxissomos/química , Peroxissomos/genética , Fosforilação , Proteínas Quinases/genética , Multimerização Proteica , Proteínas Tirosina Quinases/química , Proteínas Tirosina Quinases/genética , Transdução de Sinais/genética , Serina-Treonina Quinases TOR/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA