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










Publication year range
1.
Cell ; 167(2): 471-483.e10, 2016 Oct 06.
Article in English | MEDLINE | ID: mdl-27693358

ABSTRACT

Mitochondrial ribosomes translate membrane integral core subunits of the oxidative phosphorylation system encoded by mtDNA. These translation products associate with nuclear-encoded, imported proteins to form enzyme complexes that produce ATP. Here, we show that human mitochondrial ribosomes display translational plasticity to cope with the supply of imported nuclear-encoded subunits. Ribosomes expressing mitochondrial-encoded COX1 mRNA selectively engage with cytochrome c oxidase assembly factors in the inner membrane. Assembly defects of the cytochrome c oxidase arrest mitochondrial translation in a ribosome nascent chain complex with a partially membrane-inserted COX1 translation product. This complex represents a primed state of the translation product that can be retrieved for assembly. These findings establish a mammalian translational plasticity pathway in mitochondria that enables adaptation of mitochondrial protein synthesis to the influx of nuclear-encoded subunits.


Subject(s)
Cyclooxygenase 1/metabolism , Electron Transport Complex IV/metabolism , Membrane Proteins/metabolism , Mitochondria/enzymology , Mitochondrial Proteins/metabolism , Active Transport, Cell Nucleus , Cell Line, Tumor , Cyclooxygenase 1/biosynthesis , Cyclooxygenase 1/genetics , DNA, Mitochondrial/genetics , Electron Transport Complex IV/biosynthesis , Electron Transport Complex IV/genetics , HEK293 Cells , Humans , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mitochondrial Proteins/biosynthesis , Mitochondrial Proteins/genetics , Oxidative Phosphorylation , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Mitochondrial , Ribosomes/metabolism
2.
Mol Cell Biol ; 36(22): 2782-2793, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27550809

ABSTRACT

The three conserved core subunits of the cytochrome c oxidase are encoded by mitochondria in close to all eukaryotes. The Cox2 subunit spans the inner membrane twice, exposing the N and C termini to the intermembrane space. For this, the N terminus is exported cotranslationally by Oxa1 and subsequently undergoes proteolytic maturation in Saccharomyces cerevisiae Little is known about the translocation of the C terminus, but Cox18 has been identified to be a critical protein in this process. Here we find that the scaffold protein Cox20, which promotes processing of Cox2, is in complex with the ribosome receptor Mba1 and translating mitochondrial ribosomes in a Cox2-dependent manner. The Mba1-Cox20 complex accumulates when export of the C terminus of Cox2 is blocked by the loss of the Cox18 protein. While Cox20 engages with Cox18, Mba1 is no longer present at this stage. Our analyses indicate that Cox20 associates with nascent Cox2 and Mba1 to promote Cox2 maturation cotranslationally. We suggest that Mba1 stabilizes the Cox20-ribosome complex and supports the handover of Cox2 to the Cox18 tail export machinery.

3.
Mol Biol Cell ; 27(10): 1570-80, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27030670

ABSTRACT

The mitochondrial cytochrome c oxidase assembles in the inner membrane from subunits of dual genetic origin. The assembly process of the enzyme is initiated by membrane insertion of the mitochondria-encoded Cox1 subunit. During complex maturation, transient assembly intermediates, consisting of structural subunits and specialized chaperone-like assembly factors, are formed. In addition, cofactors such as heme and copper have to be inserted into the nascent complex. To regulate the assembly process, the availability of Cox1 is under control of a regulatory feedback cycle in which translation of COX1 mRNA is stalled when assembly intermediates of Cox1 accumulate through inactivation of the translational activator Mss51. Here we isolate a cytochrome c oxidase assembly intermediate in preparatory scale from coa1Δ mutant cells, using Mss51 as bait. We demonstrate that at this stage of assembly, the complex has not yet incorporated the heme a cofactors. Using quantitative mass spectrometry, we define the protein composition of the assembly intermediate and unexpectedly identify the putative methyltransferase Oms1 as a constituent. Our analyses show that Oms1 participates in cytochrome c oxidase assembly by stabilizing newly synthesized Cox1.


Subject(s)
Electron Transport Complex IV/metabolism , Methyltransferases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Cytochromes c/metabolism , Electron Transport Complex IV/genetics , Gene Expression Regulation, Fungal , Membrane Proteins/metabolism , Methyltransferases/genetics , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Molecular Chaperones/metabolism , Protein Biosynthesis , RNA, Messenger/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Transcription Factors/metabolism
4.
Cell Rep ; 12(10): 1644-55, 2015 Sep 08.
Article in English | MEDLINE | ID: mdl-26321642

ABSTRACT

Cytochrome c oxidase, the terminal enzyme of the respiratory chain, is assembled from mitochondria- and nuclear-encoded subunits. The MITRAC complex represents the central assembly intermediate during this process as it receives imported subunits and regulates mitochondrial translation of COX1 mRNA. The molecular processes that promote and regulate the progression of assembly downstream of MITRAC are still unknown. Here, we identify MITRAC7 as a constituent of a late form of MITRAC and as a COX1-specific chaperone. MITRAC7 is required for cytochrome c oxidase biogenesis. Surprisingly, loss of MITRAC7 or an increase in its amount causes selective cytochrome c oxidase deficiency in human cells. We demonstrate that increased MITRAC7 levels stabilize and trap COX1 in MITRAC, blocking progression in the assembly process. In contrast, MITRAC7 deficiency leads to turnover of newly synthesized COX1. Accordingly, MITRAC7 affects the biogenesis pathway by stabilizing newly synthesized COX1 in assembly intermediates, concomitantly preventing turnover.


Subject(s)
Electron Transport Complex IV/metabolism , Membrane Proteins/physiology , Mitochondrial Proteins/physiology , Molecular Chaperones/physiology , Amino Acid Sequence , Cell Line, Tumor , Enzyme Stability , HEK293 Cells , Humans , Membrane Proteins/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/metabolism , Molecular Sequence Data , Protein Multimerization , Protein Subunits/metabolism , Protein Transport
5.
Cell Metab ; 21(6): 823-33, 2015 Jun 02.
Article in English | MEDLINE | ID: mdl-25959673

ABSTRACT

Three mitochondria-encoded subunits form the catalytic core of cytochrome c oxidase, the terminal enzyme of the respiratory chain. COX1 and COX2 contain heme and copper redox centers, which are integrated during assembly of the enzyme. Defects in this process lead to an enzyme deficiency and manifest as mitochondrial disorders in humans. Here we demonstrate that COA6 is specifically required for COX2 biogenesis. Absence of COA6 leads to fast turnover of newly synthesized COX2 and a concomitant reduction in cytochrome c oxidase levels. COA6 interacts transiently with the copper-containing catalytic domain of newly synthesized COX2. Interestingly, similar to the copper metallochaperone SCO2, loss of COA6 causes cardiomyopathy in humans. We show that COA6 and SCO2 interact and that corresponding pathogenic mutations in each protein affect complex formation. Our analyses define COA6 as a constituent of the mitochondrial copper relay system, linking defects in COX2 metallation to cardiac cytochrome c oxidase deficiency.


Subject(s)
Cardiomyopathies/metabolism , Carrier Proteins/metabolism , Electron Transport Complex IV/metabolism , Mitochondrial Diseases/metabolism , Mitochondrial Proteins/metabolism , Cardiomyopathies/genetics , Carrier Proteins/genetics , Electron Transport Complex IV/genetics , HEK293 Cells , Humans , Mitochondrial Diseases/genetics , Mitochondrial Proteins/genetics , Molecular Chaperones , Saccharomyces cerevisiae
6.
FEBS Lett ; 588(17): 2985-92, 2014 Aug 25.
Article in English | MEDLINE | ID: mdl-24928273

ABSTRACT

The mitochondrial respiratory chain is essential for the conversion of energy derived from the oxidation of metabolites into the membrane potential, which drives the synthesis of ATP. The electron transporting complexes bc1 complex and the cytochrome c oxidase assemble into large supercomplexes, allowing efficient energy transduction. Currently, we have only limited information about what determines the structure of the supercomplex. Here, we characterize Aim24 in baker's yeast as a protein, which is integrated in the mitochondrial inner membrane and is required for the structural integrity of the supercomplex. Deletion of AIM24 strongly affects activity of the respiratory chain and induces a growth defect on non-fermentable medium. Our data indicate that Aim24 has a function in stabilizing the respiratory chain supercomplexes.


Subject(s)
Electron Transport Chain Complex Proteins/chemistry , Electron Transport Chain Complex Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/cytology , Amino Acid Sequence , Culture Media , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Molecular Sequence Data , Protein Stability , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry
7.
Mol Cell Biol ; 33(20): 4128-37, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23979592

ABSTRACT

Cox1, the core subunit of the cytochrome c oxidase, receives two heme a cofactors during assembly of the 13-subunit enzyme complex. However, at which step of the assembly process and how heme is inserted into Cox1 have remained an enigma. Shy1, the yeast SURF1 homolog, has been implicated in heme transfer to Cox1, whereas the heme a synthase, Cox15, catalyzes the final step of heme a synthesis. Here we performed a comprehensive analysis of cytochrome c oxidase assembly intermediates containing Shy1. Our analyses suggest that Cox15 displays a role in cytochrome c oxidase assembly, which is independent of its functions as the heme a synthase. Cox15 forms protein complexes with Shy1 and also associates with Cox1-containing complexes independently of Shy1 function. These findings indicate that Shy1 does not serve as a mobile heme carrier between the heme a synthase and maturing Cox1 but rather cooperates with Cox15 for heme transfer and insertion in early assembly intermediates of cytochrome c oxidase.


Subject(s)
Electron Transport Complex IV/genetics , Gene Expression Regulation, Fungal , Membrane Proteins/genetics , Mitochondrial Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Binding Sites , Electron Transport Complex IV/metabolism , Heme/metabolism , Membrane Proteins/metabolism , Mitochondrial Proteins/metabolism , Protein Binding , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Signal Transduction
8.
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
9.
Hum Mol Genet ; 20(12): 2379-93, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21470975

ABSTRACT

Defects in mitochondrial energy metabolism lead to severe human disorders, mainly affecting tissues especially dependent on oxidative phosphorylation, such as muscle and brain. Leigh Syndrome describes a severe encephalomyopathy in infancy, frequently caused by mutations in SURF1. SURF1, termed Shy1 in Saccharomyces cerevisiae, is a conserved assembly factor for the terminal enzyme of the respiratory chain, cytochrome c oxidase. Although the molecular function of SURF1/Shy1 is still enigmatic, loss of function leads to cytochrome c oxidase deficiency and reduced expression of the central subunit Cox1 in yeast. Here, we provide insights into the molecular mechanisms leading to disease through missense mutations in codons of the most conserved amino acids in SURF1. Mutations affecting G(124) do not compromise import of the SURF1 precursor protein but lead to fast turnover of the mature protein within the mitochondria. Interestingly, an Y(274)D exchange neither affects stability nor localization of the protein. Instead, SURF1(Y274D) accumulates in a 200 kDa cytochrome c oxidase assembly intermediate. Using yeast as a model, we demonstrate that the corresponding Shy1(Y344D) is able to overcome the stage where cytochrome c oxidase assembly links to the feedback regulation of mitochondrial Cox1 expression. However, Shy1(Y344D) impairs the assembly at later steps, most apparent at low temperature and exhibits a dominant-negative phenotype upon overexpression. Thus, exchanging the conserved tyrosine (Y(344)) with aspartate in yeast uncouples translational regulation of Cox1 from cytochrome c oxidase assembly and provides evidence for the dual functionality of Shy1.


Subject(s)
Electron Transport Complex IV/genetics , Energy Metabolism/genetics , Gene Expression Regulation, Fungal/physiology , Membrane Proteins/genetics , Mitochondrial Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/physiology , Amino Acid Substitution/genetics , Blotting, Western , Cell Line , Cloning, Molecular , Electron Transport Complex IV/metabolism , Electrophoresis, Polyacrylamide Gel , Gene Expression Regulation, Fungal/genetics , Humans , Immunoprecipitation , Mutation, Missense/genetics , Plasmids/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sequence Analysis, DNA
10.
ACS Chem Biol ; 6(4): 314-9, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21194240

ABSTRACT

To discover chemical probes to further under-stand the function of individual DNA polymerases, we established a generally applicable high-throughput screening. By applying this technique we discovered three novel inhibitor classes of human DNA polymerase λ (DNA Pol λ), a key enzyme to maintain the genetic integrity of the genome. The rhodanines, classified as an excellent drug scaffold, were found to be the most potent inhibitors for DNA Pol λ. Importantly, they are up to 10 times less active against the highly similar DNA polymerase ß. We investigated basic structure activity relationships. Furthermore, the rhodanines showed pharmacological activity in two human cancer cell lines. So the here reported small molecules could serve as useful DNA Pol λ probes and might serve as starting point to develop novel therapeutic agents.


Subject(s)
DNA Polymerase beta/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays/methods , Rhodanine/pharmacology , Antineoplastic Agents/pharmacology , Cell Survival , DNA Polymerase beta/genetics , DNA Polymerase beta/metabolism , Female , HeLa Cells , Hep G2 Cells , Humans , Inhibitory Concentration 50 , Models, Molecular , Neoplasms/drug therapy , Neoplasms/enzymology , Quantitative Structure-Activity Relationship , Rhodanine/analogs & derivatives
11.
Stem Cells ; 26(2): 455-64, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18055443

ABSTRACT

Evolutionary theory predicts that cellular maintenance, stress defense, and DNA repair mechanisms should be most active in germ line cells, including embryonic stem cells that can differentiate into germ line cells, whereas it would be energetically unfavorable to keep these up in mortal somatic cells. We tested this hypothesis by examining telomere maintenance, oxidative stress generation, and genes involved in antioxidant defense and DNA repair during spontaneous differentiation of two human embryonic stem cell lines. Telomerase activity was quickly downregulated during differentiation, probably due to deacetylation of histones H3 and H4 at the hTERT promoter and deacetylation of histone H3 at hTR promoter. Telomere length decreased accordingly. Mitochondrial superoxide production and cellular levels of reactive oxygen species increased as result of increased mitochondrial biogenesis. The expression of major antioxidant genes was downregulated despite this increased oxidative stress. DNA damage levels increased during differentiation, whereas expression of genes involved in different types of DNA repair decreased. These results confirm earlier data obtained during mouse embryonic stem cell differentiation and are in accordance with evolutionary predictions.


Subject(s)
Cell Differentiation/genetics , Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Antioxidants/metabolism , Cell Line , DNA Damage , DNA Repair , Down-Regulation , Humans , Mitochondria/metabolism , Oxidative Stress , Reactive Oxygen Species , Telomere/genetics , Telomere/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...