Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Cell Biol ; 217(4): 1369-1382, 2018 04 02.
Article in English | MEDLINE | ID: mdl-29382700

ABSTRACT

The biogenesis of mitochondria depends on the import of hundreds of preproteins. N-terminal matrix-targeting signals (MTSs) direct preproteins to the surface receptors Tom20, Tom22, and Tom70. In this study, we show that many preproteins contain additional internal MTS-like signals (iMTS-Ls) in their mature region that share the characteristic properties of presequences. These features allow the in silico prediction of iMTS-Ls. Using Atp1 as model substrate, we show that iMTS-Ls mediate the binding to Tom70 and have the potential to target the protein to mitochondria if they are presented at its N terminus. The import of preproteins with high iMTS-L content is significantly impaired in the absence of Tom70, whereas preproteins with low iMTS-L scores are less dependent on Tom70. We propose a stepping stone model according to which the Tom70-mediated interaction with internal binding sites improves the import competence of preproteins and increases the efficiency of their translocation into the mitochondrial matrix.


Subject(s)
Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Protein Interaction Domains and Motifs , Protein Precursors/metabolism , Protein Sorting Signals , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Chaperonin 60/chemistry , Chaperonin 60/genetics , Chaperonin 60/metabolism , Kinetics , Mitochondrial Membrane Transport Proteins/chemistry , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Precursor Protein Import Complex Proteins , Models, Biological , Protein Binding , Protein Precursors/chemistry , Protein Precursors/genetics , Protein Transport , Proteomics/methods , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics
2.
Mol Biol Cell ; 27(20): 3031-3039, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27582385

ABSTRACT

Whereas the structure and function of cytosolic ribosomes are well characterized, we only have a limited understanding of the mitochondrial translation apparatus. Using SILAC-based proteomic profiling, we identified 13 proteins that cofractionated with the mitochondrial ribosome, most of which play a role in translation or ribosomal biogenesis. One of these proteins is a homologue of the bacterial ribosome-silencing factor (Rsf). This protein is generated from the composite precursor protein Atp25 upon internal cleavage by the matrix processing peptidase MPP, and in this respect, it differs from all other characterized mitochondrial proteins of baker's yeast. We observed that cytosolic expression of Rsf, but not of noncleaved Atp25 protein, is toxic. Our results suggest that eukaryotic cells face the challenge of avoiding negative interference from the biogenesis of their two distinct translation machineries.


Subject(s)
Mitochondria/metabolism , Mitochondrial Ribosomes/metabolism , Amino Acid Sequence , Gene Expression Profiling/methods , Metalloendopeptidases/metabolism , Mitochondrial Proteins , Mitochondrial Ribosomes/physiology , Protein Biosynthesis/physiology , Protein Precursors/metabolism , Proteomics/methods , Ribosomal Proteins/metabolism , Ribosomes/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/metabolism , Mitochondrial Processing Peptidase
3.
Mol Biol Cell ; 26(13): 2385-401, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25926683

ABSTRACT

Members of the twin Cx9C protein family constitute the largest group of proteins in the intermembrane space (IMS) of mitochondria. Despite their conserved nature and their essential role in the biogenesis of the respiratory chain, the molecular function of twin Cx9C proteins is largely unknown. We performed a SILAC-based quantitative proteomic analysis to identify interaction partners of the conserved twin Cx9C protein Cox19. We found that Cox19 interacts in a dynamic manner with Cox11, a copper transfer protein that facilitates metalation of the Cu(B) center of subunit 1 of cytochrome c oxidase. The interaction with Cox11 is critical for the stable accumulation of Cox19 in mitochondria. Cox19 consists of a helical hairpin structure that forms a hydrophobic surface characterized by two highly conserved tyrosine-leucine dipeptides. These residues are essential for Cox19 function and its specific binding to a cysteine-containing sequence in Cox11. Our observations suggest that an oxidative modification of this cysteine residue of Cox11 stimulates Cox19 binding, pointing to a redox-regulated interplay of Cox19 and Cox11 that is critical for copper transfer in the IMS and thus for biogenesis of cytochrome c oxidase.


Subject(s)
Electron Transport Complex IV/biosynthesis , Membrane Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Molecular Chaperones/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Amino Acid Sequence , Carrier Proteins/metabolism , Electron Transport Complex IV/metabolism , Mitochondria/enzymology , Mitochondrial Membranes/enzymology , Mitochondrial Membranes/metabolism , Models, Molecular , Oxidation-Reduction , Protein Structure, Tertiary , Proteomics
4.
Nat Commun ; 6: 6019, 2015 Jan 22.
Article in English | MEDLINE | ID: mdl-25609543

ABSTRACT

Whereas the structure and function of cytosolic ribosomes have been studied in great detail, we know surprisingly little about the structural basis of mitochondrial protein synthesis. Here we used cryoelectron tomography and subtomogram analysis to visualize mitoribosomes in isolated yeast mitochondria, avoiding perturbations during ribosomal purification. Most mitoribosomes reside in immediate proximity to the inner mitochondrial membrane, in line with their specialization in the synthesis of hydrophobic membrane proteins. The subtomogram average of membrane-associated mitoribosomes reveals two distinct membrane contact sites, formed by the 21S rRNA expansion segment 96-ES1 and the inner membrane protein Mba1. On the basis of our data, we further hypothesize that Mba1 is not just a passive mitoribosome receptor on the inner membrane, but that it spatially aligns mitoribosomes with the membrane insertion machinery. This study reveals detailed insights into the supramolecular organization of the mitochondrial translation machinery and its association with the inner membrane in translation-competent mitochondria.


Subject(s)
Membrane Proteins/physiology , Mitochondria/metabolism , Mitochondrial Proteins/physiology , Protein Biosynthesis , Saccharomyces cerevisiae Proteins/physiology , Cryoelectron Microscopy , Cytosol/metabolism , Electron Transport Complex IV , Membrane Proteins/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/metabolism , Nuclear Proteins , RNA, Ribosomal/metabolism , Ribosomes/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Yeasts/physiology
5.
Nucleic Acids Res ; 42(13): 8500-15, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24948607

ABSTRACT

MPV17 is a mitochondrial protein of unknown function, and mutations in MPV17 are associated with mitochondrial deoxyribonucleic acid (DNA) maintenance disorders. Here we investigated its most similar relative, MPV17L2, which is also annotated as a mitochondrial protein. Mitochondrial fractionation analyses demonstrate MPV17L2 is an integral inner membrane protein, like MPV17. However, unlike MPV17, MPV17L2 is dependent on mitochondrial DNA, as it is absent from ρ(0) cells, and co-sediments on sucrose gradients with the large subunit of the mitochondrial ribosome and the monosome. Gene silencing of MPV17L2 results in marked decreases in the monosome and both subunits of the mitochondrial ribosome, leading to impaired protein synthesis in the mitochondria. Depletion of MPV17L2 also induces mitochondrial DNA aggregation. The DNA and ribosome phenotypes are linked, as in the absence of MPV17L2 proteins of the small subunit of the mitochondrial ribosome are trapped in the enlarged nucleoids, in contrast to a component of the large subunit. These findings suggest MPV17L2 contributes to the biogenesis of the mitochondrial ribosome, uniting the two subunits to create the translationally competent monosome, and provide evidence that assembly of the small subunit of the mitochondrial ribosome occurs at the nucleoid.


Subject(s)
Membrane Proteins/physiology , Mitochondria/genetics , Mitochondrial Proteins/physiology , Ribosomes/metabolism , Gene Silencing , HEK293 Cells , HeLa Cells , Humans , Membrane Proteins/classification , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondria/chemistry , Mitochondrial Proteins/classification , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mitochondrial Swelling , Protein Biosynthesis , Ribosome Subunits, Large, Eukaryotic/chemistry
6.
Biochem Cell Biol ; 92(6): 489-98, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24943357

ABSTRACT

Mitochondrial ribosomes of baker's yeast contain at least 78 protein subunits. All but one of these proteins are nuclear-encoded, synthesized on cytosolic ribosomes, and imported into the matrix for biogenesis. The import of matrix proteins typically relies on N-terminal mitochondrial targeting sequences that form positively charged amphipathic helices. Interestingly, the N-terminal regions of many ribosomal proteins do not closely match the characteristics of matrix targeting sequences, suggesting that the import processes of these proteins might deviate to some extent from the general import route. So far, the biogenesis of only two ribosomal proteins, Mrpl32 and Mrp10, was studied experimentally and indeed showed surprising differences to the import of other preproteins. In this review article we summarize the current knowledge on the transport of proteins into the mitochondrial matrix, and thereby specifically focus on proteins of the mitochondrial ribosome.


Subject(s)
Mitochondria/metabolism , Ribosomal Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Mitochondria/genetics , Protein Transport/physiology , Ribosomal Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
7.
Dev Cell ; 28(1): 30-42, 2014 Jan 13.
Article in English | MEDLINE | ID: mdl-24360785

ABSTRACT

Most mitochondrial proteins are synthesized in the cytosol and directed into the organelle; matrix proteins contain presequences that guide them through translocases in contact sites of the outer and inner membrane. In contrast, the import of many intermembrane space proteins depends on cysteine residues and the oxidoreductase Mia40. Here, we show that both import machineries can cooperate in the biogenesis of matrix proteins. Mrp10, a conserved protein of the mitochondrial ribosome, interacts with Mia40 during passage into the matrix. Mrp10 contains an unconventional proline-rich matrix-targeting sequence that renders import intermediates accessible to Mia40. Although oxidation of Mrp10 is not essential for its function in mitochondrial translation, the disulfide bonds prevent proteolytic degradation of Mrp10 and thereby counteract instability of the mitochondrial genome. The unconventional import pathway of Mrp10 is presumably part of a quality-control circle that connects mitochondrial ribosome biogenesis to the functionality of the mitochondrial disulfide relay.


Subject(s)
Disulfides/metabolism , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membranes/metabolism , Ribosomal Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Disulfides/chemistry , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Precursor Protein Import Complex Proteins , Oxidation-Reduction , Protein Stability , Protein Transport , Ribosomal Proteins/chemistry , Ribosomal Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics
8.
Biochim Biophys Acta ; 1833(2): 286-94, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22450032

ABSTRACT

Mitochondria contain their own genome which codes for a small number of proteins. Most mitochondrial translation products are part of the membrane-embedded reaction centers of the respiratory chain complexes. In the yeast Saccharomyces cerevisiae, the expression of these proteins is regulated by translational activators that bind mitochondrial mRNAs, in most cases to their 5'-untranslated regions, and each mitochondrial mRNA appears to have its own translational activator(s). Recent studies showed that these translational activators can be part of feedback control loops which only permit translation if the downstream assembly of nascent translation products can occur. In several cases, the accumulation of a non-assembled protein prevents further synthesis of this protein but not translation in general. These control loops prevent the synthesis of potentially harmful assembly intermediates of the reaction centers of mitochondrial enzymes. Since such regulatory feedback loops only work if translation occurs in the compartment in which the complexes of the respiratory chain are assembled, these control mechanisms require the presence of a translation machinery in mitochondria. This might explain why eukaryotic cells maintained DNA in mitochondria during the last two billion years of evolution. This review gives an overview of the mitochondrial translation system and summarizes the current knowledge on translational activators and their role in the regulation of mitochondrial protein synthesis. This article is part of a Special Issue entitled: Protein import and quality control in mitochondria and plastids.


Subject(s)
Fungal Proteins/biosynthesis , Mitochondria/metabolism , Mitochondrial Proteins/biosynthesis , Protein Biosynthesis , Yeasts/metabolism , Fungal Proteins/genetics , Mitochondrial Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/biosynthesis , Saccharomyces cerevisiae Proteins/genetics , Yeasts/genetics
9.
FEBS J ; 280(3): 904-15, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23198851

ABSTRACT

The biogenesis of hydrophobic membrane proteins involves their cotranslational membrane integration in order to prevent their unproductive aggregation. In the cytosol of bacteria and eukaryotes, membrane targeting of ribosomes that synthesize membrane proteins is achieved by signal recognition particles (SRPs) and their cognate membrane-bound receptors. As is evident from the genomes of fully sequenced eukaryotes, mitochondria generally lack an SRP system. Instead, mitochondrial ribosomes are physically associated with the protein insertion machinery in the inner membrane. Accordingly, deletion of ribosome-binding sites on the Oxa1 insertase and the Mba1 ribosome receptor in yeast leads to severe defects in cotranslational protein insertion and results in respiration-deficient mutants. In this study, we expressed mitochondria-targeted versions of the bacterial SRP protein Ffh and its receptor FtsY in these yeast mutants. Interestingly, Ffh was found to bind to the large subunit of mitochondrial ribosomes, and could relieve, to some degree, the defect of these insertion mutants. Although FtsY could also bind to mitochondrial membranes, it did not improve membrane protein biogenesis in this strain, presumably because of its inability to interact with Ffh. Hence, mitochondrial ribosomes are still able to interact physically and functionally with the bacterial SRP system. Our observations are consistent with a model according to which the protein insertion system in mitochondria evolved in three steps. The loss of genes for hydrophilic polypeptides (step 1) allowed the development of ribosome-binding sites on membrane proteins (step 2), which finally made the existence of an SRP-mediated system dispensable (step 3).


Subject(s)
Electron Transport Complex IV/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Nuclear Proteins/metabolism , Signal Recognition Particle/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Blotting, Western , Electron Transport Complex IV/genetics , Electrophoresis, Polyacrylamide Gel , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondria/genetics , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/genetics , Models, Genetic , Mutation , Nuclear Proteins/genetics , Protein Binding , Protein Biosynthesis/genetics , Protein Transport , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Ribosomes/genetics , Ribosomes/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Signal Recognition Particle/genetics
10.
J Biol Chem ; 287(41): 34484-93, 2012 Oct 05.
Article in English | MEDLINE | ID: mdl-22904327

ABSTRACT

The terminal enzyme of the respiratory chain, cytochrome c oxidase, consists of a hydrophobic reaction center formed by three mitochondrially encoded subunits with which 9-10 nuclear encoded subunits are associated. The three core subunits are synthesized on mitochondrial ribosomes and inserted into the inner membrane in a co-translational reaction facilitated by the Oxa1 insertase. Oxa1 consists of an N-terminal insertase domain and a C-terminal ribosome-binding region. Mutants lacking the C-terminal region show specific defects in co-translational insertion, suggesting that the close contact of the ribosome with the insertase promotes co-translational insertion of nascent chains. In this study, we inserted flexible linkers of 100 or 200 amino acid residues between the insertase domain and ribosome-binding region of Oxa1 of Saccharomyces cerevisiae. In the absence of the ribosome receptor Mba1, these linkers caused a length-dependent decrease in mitochondrial respiratory activity caused by diminished levels of cytochrome c oxidase. Interestingly, considerable amounts of mitochondrial translation products were still integrated into the inner membrane in these linker mutants. However, they showed severe defects in later stages of the biogenesis process, presumably during assembly into functional complexes. Our observations suggest that the close proximity of Oxa1 to ribosomes is not only used to improve membrane insertion but is also critical for the productive assembly of the subunits of the cytochrome c oxidase. This points to a role for Oxa1 in the spatial coordination of the ribosome with assembly factors that are critical for enzyme biogenesis.


Subject(s)
Electron Transport Complex IV/biosynthesis , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Nuclear Proteins/metabolism , Ribosomes/metabolism , Saccharomyces cerevisiae/metabolism , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondria/genetics , Mitochondrial Proteins/genetics , Mutation , Nuclear Proteins/genetics , Protein Structure, Tertiary , Ribosomes/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
11.
Biochim Biophys Acta ; 1788(9): 1841-50, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19560440

ABSTRACT

The present study explores the impact of the molecular size on the permeation of low-molecular-weight polyethylene glycols (PEG200-1500) through the plasma membrane of Jurkat cells under iso- and hypotonic conditions. To this end, we analyzed the cell volume responses to PEG-substituted solutions of different osmolalities (100-300 mOsm) using video microscopy. In parallel experiments, the osmotically induced changes in the membrane capacitance and cytosolic conductivity were measured by electrorotation (ROT). Upon moderate swelling in slightly hypotonic solutions (200 mOsm), the lymphocyte membrane remained impermeable to PEG300-1500, which allowed the cells to accomplish regulatory volume decrease (RVD). During RVD, lymphocytes released intracellular electrolytes through the swelling-activated pathways, as proved by a decrease of the cytosolic conductivity measured by electrorotation. RVD also occurred in strongly hypotonic solutions (100 mOsm) of PEG600-1500, whereas 100 mOsm solutions of PEG300-400 inhibited RVD in Jurkat cells. These findings suggest that extensive hypotonic swelling rendered the cell membrane highly permeable to PEG300-400, but not to PEG600-1500. The swelling-activated channels conducting PEG300-400 were inserted into the plasma membrane from cytosolic vesicles via swelling-mediated exocytosis, as suggested by an increase of the whole cell capacitance. Using the hydrodynamic radii R(h) of PEGs (determined by viscosimetry), the observed size-selectivity of membrane permeation yielded an estimate of approximately 0.74 nm for the cut-off radius of the swelling-activated channel for organic osmolytes. Unlike PEG300-1500, the smallest PEG (PEG200, R(h)=0.5 nm) permeated the lymphocyte membrane under isotonic conditions thus leading to a continuous isotonic swelling. The results are of interest for biotechnology and biomedicine, where PEGs are widely used for cryopreservation of cells and tissues.


Subject(s)
Cell Membrane Permeability/drug effects , Ion Channels/physiology , Lymphocytes/physiology , Cell Size/drug effects , Electrophysiology , Humans , Jurkat Cells , Lymphocytes/cytology , Osmolar Concentration , Polyethylene Glycols/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...