Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters











Publication year range
1.
Life Sci Alliance ; 7(2)2024 02.
Article in English | MEDLINE | ID: mdl-37984987

ABSTRACT

Mitochondria are essential organelles whose dysfunction causes human pathologies that often manifest in a tissue-specific manner. Accordingly, mitochondrial fitness depends on versatile proteomes specialized to meet diverse tissue-specific requirements. Increasing evidence suggests that phosphorylation may play an important role in regulating tissue-specific mitochondrial functions and pathophysiology. Building on recent advances in mass spectrometry (MS)-based proteomics, we here quantitatively profile mitochondrial tissue proteomes along with their matching phosphoproteomes. We isolated mitochondria from mouse heart, skeletal muscle, brown adipose tissue, kidney, liver, brain, and spleen by differential centrifugation followed by separation on Percoll gradients and performed high-resolution MS analysis of the proteomes and phosphoproteomes. This in-depth map substantially quantifies known and predicted mitochondrial proteins and provides a resource of core and tissue-specific mitochondrial proteins (mitophos.de). Predicting kinase substrate associations for different mitochondrial compartments indicates tissue-specific regulation at the phosphoproteome level. Illustrating the functional value of our resource, we reproduce mitochondrial phosphorylation events on dynamin-related protein 1 responsible for its mitochondrial recruitment and fission initiation and describe phosphorylation clusters on MIGA2 linked to mitochondrial fusion.


Subject(s)
Mitochondria , Proteome , Mice , Animals , Humans , Proteome/metabolism , Mitochondria/metabolism , Phosphorylation , Mass Spectrometry , Mitochondrial Proteins/metabolism
2.
Biol Chem ; 403(8-9): 779-805, 2022 07 26.
Article in English | MEDLINE | ID: mdl-35355496

ABSTRACT

Mitochondria are central hubs for cellular metabolism, coordinating a variety of metabolic reactions crucial for human health. Mitochondria provide most of the cellular energy via their oxidative phosphorylation (OXPHOS) system, which requires the coordinated expression of genes encoded by both the nuclear (nDNA) and mitochondrial genomes (mtDNA). Transcription of mtDNA is not only essential for the biogenesis of the OXPHOS system, but also generates RNA primers necessary to initiate mtDNA replication. Like the prokaryotic system, mitochondria have no membrane-based compartmentalization to separate the different steps of mtDNA maintenance and expression and depend entirely on nDNA-encoded factors imported into the organelle. Our understanding of mitochondrial transcription in mammalian cells has largely progressed, but the mechanisms regulating mtDNA gene expression are still poorly understood despite their profound importance for human disease. Here, we review mechanisms of mitochondrial gene expression with a focus on the recent findings in the field of mammalian mtDNA transcription and disease phenotypes caused by defects in proteins involved in this process.


Subject(s)
DNA, Mitochondrial , Genome, Mitochondrial , Animals , DNA, Mitochondrial/genetics , Humans , Mammals/genetics , Mammals/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Oxidative Phosphorylation , Transcription, Genetic
3.
Biochim Biophys Acta Mol Cell Res ; 1867(5): 118661, 2020 05.
Article in English | MEDLINE | ID: mdl-31987792

ABSTRACT

Artemisinin and its derivatives kill malaria parasites and inhibit the proliferation of cancer cells. In both processes, heme was shown to play a key role in artemisinin bioactivation. We found that artemisinin and clinical artemisinin derivatives are able to compensate for a mutation in the yeast Bcs1 protein, a key chaperon involved in biogenesis of the mitochondrial respiratory complex III. The equivalent Bcs1 variant causes an encephalopathy in human by affecting complex III assembly. We show that artemisinin derivatives decrease the content of mitochondrial cytochromes and disturb the maturation of the complex III cytochrome c1. This last effect is likely responsible for the compensation by decreasing the detrimental over-accumulation of the inactive pre-complex III observed in the bcs1 mutant. We further show that a fluorescent dihydroartemisinin probe rapidly accumulates in the mitochondrial network and targets cytochromes c and c1 in yeast, human cells and isolated mitochondria. In vitro this probe interacts with purified cytochrome c only under reducing conditions and we detect cytochrome c-dihydroartemisinin covalent adducts by mass spectrometry analyses. We propose that reduced mitochondrial c-type cytochromes act as both targets and mediators of artemisinin bioactivation in yeast and human cells.


Subject(s)
Artemisinins/pharmacology , Cytochromes c/metabolism , Mitochondria/metabolism , Saccharomyces cerevisiae/metabolism , ATPases Associated with Diverse Cellular Activities/genetics , Artemisinins/chemistry , Down-Regulation , Electron Transport Complex III/genetics , HEK293 Cells , Humans , Mitochondria/drug effects , Mitochondrial Proteins/genetics , Molecular Chaperones/genetics , Mutation , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
4.
PLoS Genet ; 15(6): e1008085, 2019 06.
Article in English | MEDLINE | ID: mdl-31170154

ABSTRACT

Mitochondrial dynamics is an essential physiological process controlling mitochondrial content mixing and mobility to ensure proper function and localization of mitochondria at intracellular sites of high-energy demand. Intriguingly, for yet unknown reasons, severe impairment of mitochondrial fusion drastically affects mtDNA copy number. To decipher the link between mitochondrial dynamics and mtDNA maintenance, we studied mouse embryonic fibroblasts (MEFs) and mouse cardiomyocytes with disruption of mitochondrial fusion. Super-resolution microscopy revealed that loss of outer mitochondrial membrane (OMM) fusion, but not inner mitochondrial membrane (IMM) fusion, leads to nucleoid clustering. Remarkably, fluorescence in situ hybridization (FISH), bromouridine labeling in MEFs and assessment of mitochondrial transcription in tissue homogenates revealed that abolished OMM fusion does not affect transcription. Furthermore, the profound mtDNA depletion in mouse hearts lacking OMM fusion is not caused by defective integrity or increased mutagenesis of mtDNA, but instead we show that mitochondrial fusion is necessary to maintain the stoichiometry of the protein components of the mtDNA replisome. OMM fusion is necessary for proliferating MEFs to recover from mtDNA depletion and for the marked increase of mtDNA copy number during postnatal heart development. Our findings thus link OMM fusion to replication and distribution of mtDNA.


Subject(s)
DNA, Mitochondrial/genetics , Mitochondria, Heart/genetics , Mitochondrial Dynamics/genetics , Mitochondrial Proteins/genetics , Animals , DNA Copy Number Variations/genetics , DNA Replication/genetics , Fibroblasts , Humans , In Situ Hybridization, Fluorescence , Membrane Fusion/genetics , Mice , Mitochondria, Heart/metabolism , Mitochondrial Membranes/metabolism , Mutagenesis , Myocytes, Cardiac/metabolism , Transcription, Genetic
5.
EMBO Rep ; 20(6)2019 06.
Article in English | MEDLINE | ID: mdl-31036713

ABSTRACT

Regulation of replication and expression of mitochondrial DNA (mtDNA) is essential for cellular energy conversion via oxidative phosphorylation. The mitochondrial transcription elongation factor (TEFM) has been proposed to regulate the switch between transcription termination for replication primer formation and processive, near genome-length transcription for mtDNA gene expression. Here, we report that Tefm is essential for mouse embryogenesis and that levels of promoter-distal mitochondrial transcripts are drastically reduced in conditional Tefm-knockout hearts. In contrast, the promoter-proximal transcripts are much increased in Tefm knockout mice, but they mostly terminate before the region where the switch from transcription to replication occurs, and consequently, de novo mtDNA replication is profoundly reduced. Unexpectedly, deep sequencing of RNA from Tefm knockouts revealed accumulation of unprocessed transcripts in addition to defective transcription elongation. Furthermore, a proximity-labeling (BioID) assay showed that TEFM interacts with multiple RNA processing factors. Our data demonstrate that TEFM acts as a general transcription elongation factor, necessary for both gene transcription and replication primer formation, and loss of TEFM affects RNA processing in mammalian mitochondria.


Subject(s)
Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , RNA Processing, Post-Transcriptional , Transcription Elongation, Genetic , Transcription Factors/metabolism , Animals , DNA, Mitochondrial , Embryonic Development/genetics , Gene Deletion , Gene Expression Regulation , Genetic Loci , Heterozygote , Mice , Mice, Knockout , Mitochondria/ultrastructure , Phenotype , Promoter Regions, Genetic
6.
Elife ; 62017 11 14.
Article in English | MEDLINE | ID: mdl-29132502

ABSTRACT

Dysfunction of the oxidative phosphorylation (OXPHOS) system is a major cause of human disease and the cellular consequences are highly complex. Here, we present comparative analyses of mitochondrial proteomes, cellular transcriptomes and targeted metabolomics of five knockout mouse strains deficient in essential factors required for mitochondrial DNA gene expression, leading to OXPHOS dysfunction. Moreover, we describe sequential protein changes during post-natal development and progressive OXPHOS dysfunction in time course analyses in control mice and a middle lifespan knockout, respectively. Very unexpectedly, we identify a new response pathway to OXPHOS dysfunction in which the intra-mitochondrial synthesis of coenzyme Q (ubiquinone, Q) and Q levels are profoundly decreased, pointing towards novel possibilities for therapy. Our extensive omics analyses provide a high-quality resource of altered gene expression patterns under severe OXPHOS deficiency comparing several mouse models, that will deepen our understanding, open avenues for research and provide an important reference for diagnosis and treatment.


Subject(s)
Ataxia/pathology , Gene Expression Profiling , Metabolome , Mitochondria/chemistry , Mitochondrial Diseases/pathology , Muscle Weakness/pathology , Proteome/analysis , Ubiquinone/deficiency , Animals , Mice, Knockout
7.
Sci Adv ; 2(8): e1600963, 2016 08.
Article in English | MEDLINE | ID: mdl-27532055

ABSTRACT

Mitochondria are vital in providing cellular energy via their oxidative phosphorylation system, which requires the coordinated expression of genes encoded by both the nuclear and mitochondrial genomes (mtDNA). Transcription of the circular mammalian mtDNA depends on a single mitochondrial RNA polymerase (POLRMT). Although the transcription initiation process is well understood, it is debated whether POLRMT also serves as the primase for the initiation of mtDNA replication. In the nucleus, the RNA polymerases needed for gene expression have no such role. Conditional knockout of Polrmt in the heart results in severe mitochondrial dysfunction causing dilated cardiomyopathy in young mice. We further studied the molecular consequences of different expression levels of POLRMT and found that POLRMT is essential for primer synthesis to initiate mtDNA replication in vivo. Furthermore, transcription initiation for primer formation has priority over gene expression. Surprisingly, mitochondrial transcription factor A (TFAM) exists in an mtDNA-free pool in the Polrmt knockout mice. TFAM levels remain unchanged despite strong mtDNA depletion, and TFAM is thus protected from degradation of the AAA(+) Lon protease in the absence of POLRMT. Last, we report that mitochondrial transcription elongation factor may compensate for a partial depletion of POLRMT in heterozygous Polrmt knockout mice, indicating a direct regulatory role of this factor in transcription. In conclusion, we present in vivo evidence that POLRMT has a key regulatory role in the replication of mammalian mtDNA and is part of a transcriptional mechanism that provides a switch between primer formation for mtDNA replication and mitochondrial gene expression.


Subject(s)
DNA, Mitochondrial/genetics , DNA-Binding Proteins/genetics , DNA-Directed RNA Polymerases/genetics , High Mobility Group Proteins/genetics , Transcription, Genetic , Animals , DNA Replication/genetics , Gene Expression Regulation , Genome, Mitochondrial , Mice
8.
Proc Natl Acad Sci U S A ; 112(36): 11288-93, 2015 Sep 08.
Article in English | MEDLINE | ID: mdl-26305956

ABSTRACT

Mammalian mitochondrial DNA (mtDNA) is packaged by mitochondrial transcription factor A (TFAM) into mitochondrial nucleoids that are of key importance in controlling the transmission and expression of mtDNA. Nucleoid ultrastructure is poorly defined, and therefore we used a combination of biochemistry, superresolution microscopy, and electron microscopy to show that mitochondrial nucleoids have an irregular ellipsoidal shape and typically contain a single copy of mtDNA. Rotary shadowing electron microscopy revealed that nucleoid formation in vitro is a multistep process initiated by TFAM aggregation and cross-strand binding. Superresolution microscopy of cultivated cells showed that increased mtDNA copy number increases nucleoid numbers without altering their sizes. Electron cryo-tomography visualized nucleoids at high resolution in isolated mammalian mitochondria and confirmed the sizes observed by superresolution microscopy of cell lines. We conclude that the fundamental organizational unit of the mitochondrial nucleoid is a single copy of mtDNA compacted by TFAM, and we suggest a packaging mechanism.


Subject(s)
DNA, Mitochondrial/metabolism , DNA-Binding Proteins/metabolism , High Mobility Group Proteins/metabolism , Mitochondria/metabolism , Nucleoproteins/metabolism , Animals , Cells, Cultured , Cryoelectron Microscopy , DNA, Mitochondrial/genetics , DNA, Mitochondrial/ultrastructure , DNA-Binding Proteins/genetics , DNA-Binding Proteins/ultrastructure , Electron Microscope Tomography , Genome, Mitochondrial/genetics , High Mobility Group Proteins/genetics , High Mobility Group Proteins/ultrastructure , Mice , Microscopy, Confocal , Mitochondria/genetics , Mitochondria/ultrastructure , Mutation , Nucleoproteins/genetics , Nucleoproteins/ultrastructure , Protein Binding
10.
Hum Mol Genet ; 22(10): 1983-93, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23393161

ABSTRACT

Replication of the mammalian mitochondrial DNA (mtDNA) is dependent on the minimal replisome, consisting of the heterotrimeric mtDNA polymerase (POLG), the hexameric DNA helicase TWINKLE and the tetrameric single-stranded DNA-binding protein (mtSSB). TWINKLE has been shown to unwind DNA during the replication process and many disease-causing mutations have been mapped to its gene. Patients carrying Twinkle mutations develop multiple deletions of mtDNA, deficient respiratory chain function and neuromuscular symptoms. Despite its importance in human disease, it has been unclear whether TWINKLE is the only replicative DNA helicase in mammalian mitochondria. Furthermore, a substantial portion of mtDNA replication events is prematurely terminated at the end of mitochondrial control region (D-loop) and it is unknown whether TWINKLE also has a role in this abortive replication. Here, we present a conditional mouse knockout for Twinkle and demonstrate that TWINKLE is essential for mouse embryonic development and thus is the only replicative DNA helicase in mammalian mitochondria. Conditional knockout of Twinkle results in severe and rapid mtDNA depletion in heart and skeletal muscle. No replication intermediates or deleted mtDNA molecules are observed after Twinkle knockout, suggesting that TWINKLE once loaded is very processive. We also demonstrate that TWINKLE is essential for nascent H-strand synthesis in the D-loop, thus showing that there is no separate DNA helicase responsible for replication of this region. Our data thus suggest that the relative levels of abortive D-loop synthesis versus complete mtDNA replication are regulated and may provide a mechanism to control progression to complete mtDNA replication.


Subject(s)
DNA Helicases/metabolism , DNA Replication/physiology , DNA, Mitochondrial/biosynthesis , Mitochondrial Proteins/metabolism , Animals , DNA Helicases/genetics , DNA, Mitochondrial/genetics , Genetic Diseases, Inborn/enzymology , Genetic Diseases, Inborn/genetics , Humans , Mice , Mice, Knockout , Mitochondrial Proteins/genetics , Mutation , Neuromuscular Diseases/enzymology , Neuromuscular Diseases/genetics
11.
J Cell Biol ; 199(1): 137-50, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-23007649

ABSTRACT

Respiratory chain complexes in mitochondria are assembled from subunits derived from two genetic systems. For example, the bc(1) complex consists of nine nuclear encoded subunits and the mitochondrially encoded subunit cytochrome b. We recently showed that the Cbp3-Cbp6 complex has a dual function for biogenesis of cytochrome b: it is both required for efficient synthesis of cytochrome b and for protection of the newly synthesized protein from proteolysis. Here, we report that Cbp3-Cbp6 also coordinates cytochrome b synthesis with bc(1) complex assembly. We show that newly synthesized cytochrome b assembled through a series of four assembly intermediates. Blocking assembly at early and intermediate steps resulted in sequestration of Cbp3-Cbp6 in a cytochrome b-containing complex, thereby making Cbp3-Cbp6 unavailable for cytochrome b synthesis and thus reducing overall cytochrome b levels. This feedback loop regulates protein synthesis at the inner mitochondrial membrane by directly monitoring the efficiency of bc(1) complex assembly.


Subject(s)
Cytochrome b Group/biosynthesis , Membrane Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Molecular Chaperones/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Cytochrome b Group/metabolism , Membrane Proteins/genetics , Mitochondria/genetics , Mitochondrial Proteins/genetics , Models, Biological , Molecular Chaperones/genetics , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
12.
Mitochondrion ; 12(3): 381-90, 2012 May.
Article in English | MEDLINE | ID: mdl-22349564

ABSTRACT

Complexes III and IV of the mitochondrial respiratory chain contain a few key subunits encoded by the mitochondrial genome. In Saccharomyces cerevisiae, fifteen mRNA-specific translational activators control mitochondrial translation, of which five are conserved in Schizosaccharomyces pombe. These include homologs of Cbp3, Cbp6 and Mss51 that participate in translation and the post-translational steps leading to the assembly of respiratory complexes III and IV. In this study we show that in contrast to budding yeast, Cbp3, Cbp6 and Mss51 from S. pombe are not required for the translation of mitochondrial mRNAs, but fulfill post-translational functions, thus probably accounting for their conservation.


Subject(s)
Electron Transport Complex IV/biosynthesis , Mitochondrial Proteins/metabolism , Protein Biosynthesis , Saccharomyces cerevisiae/metabolism , Schizosaccharomyces/metabolism , Sequence Homology, Amino Acid , Amino Acid Sequence , Electron Transport Complex IV/genetics , Fungal Proteins/genetics , Fungal Proteins/metabolism , Gene Expression Regulation, Fungal , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Molecular Sequence Data , Organelle Biogenesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Mitochondrial , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Schizosaccharomyces/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
13.
Nucleic Acids Res ; 39(18): 8029-41, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21727087

ABSTRACT

Pentatricopeptide repeat (PPR) proteins are particularly numerous in plant mitochondria and chloroplasts, where they are involved in different steps of RNA metabolism, probably due to the repeated 35 amino acid PPR motifs that are thought to mediate interactions with RNA. In non-photosynthetic eukaryotes only a handful of PPR proteins exist, for example the human LRPPRC, which is involved in a mitochondrial disease. We have conducted a systematic study of the PPR proteins in the fission yeast Schizosaccharomyces pombe and identified, in addition to the mitochondrial RNA polymerase, eight proteins all of which localized to the mitochondria, and showed some association with the membrane. The absence of all but one of these PPR proteins leads to a respiratory deficiency and modified patterns of steady state mt-mRNAs or newly synthesized mitochondrial proteins. Some cause a general defect, whereas others affect specific mitochondrial RNAs, either coding or non-coding: cox1, cox2, cox3, 15S rRNA, atp9 or atp6, sometimes leading to secondary defects. Interestingly, the two possible homologs of LRPPRC, ppr4 and ppr5, play opposite roles in the expression of the cox1 mt-mRNA, ppr4 being the first mRNA-specific translational activator identified in S. pombe, whereas ppr5 appears to be a general negative regulator of mitochondrial translation.


Subject(s)
Gene Expression Regulation, Fungal , Genes, Mitochondrial , Mitochondria/genetics , Mitochondrial Proteins/physiology , Schizosaccharomyces pombe Proteins/physiology , Amino Acid Motifs , Genome, Fungal , Mitochondria/metabolism , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/genetics , Mutation , Phenotype , Protein Biosynthesis , RNA/metabolism , RNA Stability , RNA, Mitochondrial , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/chemistry , Schizosaccharomyces pombe Proteins/genetics , Sequence Homology, Amino Acid
14.
Nucleic Acids Res ; 36(18): 5787-99, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18782833

ABSTRACT

The molecular mechanism of human mitochondrial translation has yet to be fully described. We are particularly interested in understanding the process of translational termination and ribosome recycling in the mitochondrion. Several candidates have been implicated, for which subcellular localization and characterization have not been reported. Here, we show that the putative mitochondrial recycling factor, mtRRF, is indeed a mitochondrial protein. Expression of human mtRRF in fission yeast devoid of endogenous mitochondrial recycling factor suppresses the respiratory phenotype. Further, human mtRRF is able to associate with Escherichia coli ribosomes in vitro and can associate with mitoribosomes in vivo. Depletion of mtRRF in human cell lines is lethal, initially causing profound mitochondrial dysmorphism, aggregation of mitoribosomes, elevated mitochondrial superoxide production and eventual loss of OXPHOS complexes. Finally, mtRRF was shown to co-immunoprecipitate a large number of mitoribosomal proteins attached to other mitochondrial proteins, including putative members of the mitochondrial nucleoid.


Subject(s)
Mitochondria/genetics , Mitochondrial Proteins/physiology , Ribosomal Proteins/physiology , Amino Acid Sequence , Cell Death , Cell Line , Cell Proliferation , Cell Respiration , Cell Survival , Escherichia coli/genetics , HeLa Cells , Humans , Immunoprecipitation , Mitochondria/ultrastructure , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/genetics , Molecular Sequence Data , Protein Biosynthesis , Reactive Oxygen Species/metabolism , Ribosomal Proteins/antagonists & inhibitors , Ribosomal Proteins/genetics , Ribosomes/metabolism , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/genetics
15.
Mol Cell ; 27(5): 745-57, 2007 Sep 07.
Article in English | MEDLINE | ID: mdl-17803939

ABSTRACT

Human mitochondria contain their own genome, encoding 13 polypeptides that are synthesized within the organelle. The molecular processes that govern and facilitate this mitochondrial translation remain unclear. Many key factors have yet to be characterized-for example, those required for translation termination. All other systems have two classes of release factors that either promote codon-specific hydrolysis of peptidyl-tRNA (class I) or lack specificity but stimulate the dissociation of class I factors from the ribosome (class II). One human mitochondrial protein has been previously identified in silico as a putative member of the class I release factors. Although we could not confirm the function of this factor, we report the identification of a different mitochondrial protein, mtRF1a, that is capable in vitro and in vivo of terminating translation at UAA/UAG codons. Further, mtRF1a depletion in HeLa cells led to compromised growth in galactose and increased production of reactive oxygen species.


Subject(s)
Codon, Terminator , Mitochondrial Proteins/physiology , Peptide Termination Factors/physiology , Transcription Factors/physiology , Cell Proliferation , Gene Dosage , Genetic Complementation Test , HeLa Cells , Humans , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/ultrastructure , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Models, Genetic , Peptide Chain Termination, Translational , Peptide Termination Factors/genetics , Peptide Termination Factors/metabolism , Protein Biosynthesis/physiology , RNA/metabolism , RNA Interference , RNA, Mitochondrial , Reactive Oxygen Species/metabolism , Saccharomyces cerevisiae/genetics , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL