Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
1.
Mol Cell ; 77(1): 180-188.e9, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31630969

ABSTRACT

The mitochondrial proteome is built mainly by import of nuclear-encoded precursors, which are targeted mostly by cleavable presequences. Presequence processing upon import is essential for proteostasis and survival, but the consequences of dysfunctional protein maturation are unknown. We find that impaired presequence processing causes accumulation of precursors inside mitochondria that form aggregates, which escape degradation and unexpectedly do not cause cell death. Instead, cells survive via activation of a mitochondrial unfolded protein response (mtUPR)-like pathway that is triggered very early after precursor accumulation. In contrast to classical stress pathways, this immediate response maintains mitochondrial protein import, membrane potential, and translation through translocation of the nuclear HMG-box transcription factor Rox1 to mitochondria. Rox1 binds mtDNA and performs a TFAM-like function pivotal for transcription and translation. Induction of early mtUPR provides a reversible stress model to mechanistically dissect the initial steps in mtUPR pathways with the stressTFAM Rox1 as the first line of defense.


Subject(s)
Mitochondria/metabolism , Repressor Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/metabolism , Unfolded Protein Response/physiology , Cell Death/physiology , Cell Nucleus/metabolism , DNA, Mitochondrial/metabolism , Membrane Potentials/physiology , Protein Biosynthesis/physiology , Saccharomyces cerevisiae/metabolism , Transcription, Genetic/physiology
2.
PLoS Genet ; 17(7): e1009664, 2021 07.
Article in English | MEDLINE | ID: mdl-34214073

ABSTRACT

Mitochondrial defects can cause a variety of human diseases and protective mechanisms exist to maintain mitochondrial functionality. Imbalances in mitochondrial proteostasis trigger a transcriptional program, termed mitochondrial unfolded protein response (mtUPR). However, the temporal sequence of events in mtUPR is unclear and the consequences on mitochondrial protein import are controversial. Here, we have quantitatively analyzed all main import pathways into mitochondria after different time spans of mtUPR induction. Kinetic analyses reveal that protein import into all mitochondrial subcompartments strongly increases early upon mtUPR and that this is accompanied by rapid remodelling of the mitochondrial signature lipid cardiolipin. Genetic inactivation of cardiolipin synthesis precluded stimulation of protein import and compromised cellular fitness. At late stages of mtUPR upon sustained stress, mitochondrial protein import efficiency declined. Our work clarifies the enigma of protein import upon mtUPR and identifies sequential mtUPR stages, in which an early increase in protein biogenesis to restore mitochondrial proteostasis is followed by late stages characterized by a decrease in import capacity upon prolonged stress induction.


Subject(s)
Cardiolipins/metabolism , Protein Transport/physiology , Unfolded Protein Response/physiology , Cardiolipins/physiology , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/physiology , Mitochondrial Proteins/metabolism , Protein Biosynthesis , Protein Transport/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Unfolded Protein Response/genetics
3.
PLoS Genet ; 17(11): e1009911, 2021 11.
Article in English | MEDLINE | ID: mdl-34780474

ABSTRACT

The capacity of a cell to maintain proteostasis progressively declines during aging. Virtually all age-associated neurodegenerative disorders associated with aggregation of neurotoxic proteins are linked to defects in the cellular proteostasis network, including insufficient lysosomal hydrolysis. Here, we report that proteotoxicity in yeast and Drosophila models for Parkinson's disease can be prevented by increasing the bioavailability of Ca2+, which adjusts intracellular Ca2+ handling and boosts lysosomal proteolysis. Heterologous expression of human α-synuclein (αSyn), a protein critically linked to Parkinson's disease, selectively increases total cellular Ca2+ content, while the levels of manganese and iron remain unchanged. Disrupted Ca2+ homeostasis results in inhibition of the lysosomal protease cathepsin D and triggers premature cellular and organismal death. External administration of Ca2+ reduces αSyn oligomerization, stimulates cathepsin D activity and in consequence restores survival, which critically depends on the Ca2+/calmodulin-dependent phosphatase calcineurin. In flies, increasing the availability of Ca2+ discloses a neuroprotective role of αSyn upon manganese overload. In sum, we establish a molecular interplay between cathepsin D and calcineurin that can be activated by Ca2+ administration to counteract αSyn proteotoxicity.


Subject(s)
Calcineurin/genetics , Cathepsin D/genetics , Parkinson Disease/genetics , alpha-Synuclein/genetics , Aging/drug effects , Aging/genetics , Animals , Animals, Genetically Modified/genetics , Calcium/metabolism , Calcium/pharmacology , Cell Death/genetics , Drosophila melanogaster/genetics , Gene Expression Regulation/drug effects , Humans , Lysosomes/drug effects , Lysosomes/genetics , Neurons/drug effects , Parkinson Disease/metabolism , Parkinson Disease/pathology , Protein Aggregation, Pathological/drug therapy , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/pathology , Proteolysis/drug effects , Saccharomyces cerevisiae/genetics
4.
Proc Natl Acad Sci U S A ; 118(30)2021 07 27.
Article in English | MEDLINE | ID: mdl-34290138

ABSTRACT

Nuclear envelope budding (NEB) is a recently discovered alternative pathway for nucleocytoplasmic communication distinct from the movement of material through the nuclear pore complex. Through quantitative electron microscopy and tomography, we demonstrate how NEB is evolutionarily conserved from early protists to human cells. In the yeast Saccharomyces cerevisiae, NEB events occur with higher frequency during heat shock, upon exposure to arsenite or hydrogen peroxide, and when the proteasome is inhibited. Yeast cells treated with azetidine-2-carboxylic acid, a proline analog that induces protein misfolding, display the most dramatic increase in NEB, suggesting a causal link to protein quality control. This link was further supported by both localization of ubiquitin and Hsp104 to protein aggregates and NEB events, and the evolution of these structures during heat shock. We hypothesize that NEB is part of normal cellular physiology in a vast range of species and that in S. cerevisiae NEB comprises a stress response aiding the transport of protein aggregates across the nuclear envelope.


Subject(s)
Azetidinecarboxylic Acid/toxicity , Heat-Shock Response , Nuclear Envelope/physiology , Protein Folding , Proteostasis/drug effects , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/growth & development , Arsenites/toxicity , Hydrogen Peroxide/toxicity , Nuclear Envelope/drug effects , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae Proteins/drug effects , Saccharomyces cerevisiae Proteins/metabolism , Sodium Compounds/toxicity , Ubiquitin/metabolism , Ubiquitination
5.
EMBO Rep ; 21(12): e51015, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33016568

ABSTRACT

Respiratory chains are crucial for cellular energy conversion and consist of multi-subunit complexes that can assemble into supercomplexes. These structures have been intensively characterized in various organisms, but their physiological roles remain unclear. Here, we elucidate their function by leveraging a high-resolution structural model of yeast respiratory supercomplexes that allowed us to inhibit supercomplex formation by mutation of key residues in the interaction interface. Analyses of a mutant defective in supercomplex formation, which still contains fully functional individual complexes, show that the lack of supercomplex assembly delays the diffusion of cytochrome c between the separated complexes, thus reducing electron transfer efficiency. Consequently, competitive cellular fitness is severely reduced in the absence of supercomplex formation and can be restored by overexpression of cytochrome c. In sum, our results establish how respiratory supercomplexes increase the efficiency of cellular energy conversion, thereby providing an evolutionary advantage for aerobic organisms.


Subject(s)
Cytochromes c , Saccharomyces cerevisiae Proteins , Cytochromes c/genetics , Electron Transport , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
6.
EMBO Rep ; 20(10): e47865, 2019 10 04.
Article in English | MEDLINE | ID: mdl-31531937

ABSTRACT

The eukaryotic cell is morphologically and functionally organized as an interconnected network of organelles that responds to stress and aging. Organelles communicate via dedicated signal transduction pathways and the transfer of information in form of metabolites and energy levels. Recent data suggest that the communication between organellar proteostasis systems is a cornerstone of cellular stress responses in eukaryotic cells. Here, we discuss the integration of proteostasis and energy fluxes in the regulation of cellular stress and aging. We emphasize the molecular architecture of the regulatory transcriptional pathways that both sense and control metabolism and proteostasis. A special focus is placed on mechanistic insights gained from the model organism budding yeast in signaling from mitochondria to the nucleus and how this shapes cellular fitness.


Subject(s)
Mitochondria/metabolism , Proteostasis , Stress, Physiological , Animals , Cell Nucleus/metabolism , Humans , Protein Aggregates , Transcription, Genetic
7.
J Biol Chem ; 294(32): 12020-12039, 2019 08 09.
Article in English | MEDLINE | ID: mdl-31209110

ABSTRACT

Autophagy, a membrane-dependent catabolic process, ensures survival of aging cells and depends on the cellular energetic status. Acetyl-CoA carboxylase 1 (Acc1) connects central energy metabolism to lipid biosynthesis and is rate-limiting for the de novo synthesis of lipids. However, it is unclear how de novo lipogenesis and its metabolic consequences affect autophagic activity. Here, we show that in aging yeast, autophagy levels highly depend on the activity of Acc1. Constitutively active Acc1 (acc1S/A ) or a deletion of the Acc1 negative regulator, Snf1 (yeast AMPK), shows elevated autophagy levels, which can be reversed by the Acc1 inhibitor soraphen A. Vice versa, pharmacological inhibition of Acc1 drastically reduces cell survival and results in the accumulation of Atg8-positive structures at the vacuolar membrane, suggesting late defects in the autophagic cascade. As expected, acc1S/A cells exhibit a reduction in acetate/acetyl-CoA availability along with elevated cellular lipid content. However, concomitant administration of acetate fails to fully revert the increase in autophagy exerted by acc1S/A Instead, administration of oleate, while mimicking constitutively active Acc1 in WT cells, alleviates the vacuolar fusion defects induced by Acc1 inhibition. Our results argue for a largely lipid-dependent process of autophagy regulation downstream of Acc1. We present a versatile genetic model to investigate the complex relationship between acetate metabolism, lipid homeostasis, and autophagy and propose Acc1-dependent lipogenesis as a fundamental metabolic path downstream of Snf1 to maintain autophagy and survival during cellular aging.


Subject(s)
Acetyl-CoA Carboxylase/metabolism , Autophagy , Lipogenesis , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Acetates/metabolism , Acetyl-CoA Carboxylase/antagonists & inhibitors , Acetyl-CoA Carboxylase/genetics , Autophagy/drug effects , Macrolides/pharmacology , Mutagenesis, Site-Directed , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/genetics
8.
Hum Mol Genet ; 27(9): 1593-1607, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29474575

ABSTRACT

TDP-43 is a nuclear RNA-binding protein whose cytoplasmic accumulation is the pathological hallmark of amyotrophic lateral sclerosis (ALS). For a better understanding of this devastating disorder at the molecular level, it is important to identify cellular pathways involved in the clearance of detrimental TDP-43. Using a yeast model system, we systematically analyzed to which extent TDP-43-triggered cytotoxicity is modulated by conserved lysosomal clearance pathways. We observed that the lysosomal fusion machinery and the endolysosomal pathway, which are crucial for proper lysosomal function, were pivotal for survival of cells exposed to TDP-43. Interestingly, TDP-43 itself interfered with these critical TDP-43 clearance pathways. In contrast, autophagy played a complex role in this process. It contributed to the degradation of TDP-43 in the absence of endolysosomal pathway activity, but its induction also enhanced cell death. Thus, TDP-43 interfered with lysosomal function and its own degradation via lysosomal pathways, and triggered lethal autophagy. We propose that these effects critically contribute to cellular dysfunction in TDP-43 proteinopathies.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , DNA-Binding Proteins/metabolism , Lysosomes/metabolism , Autophagy/physiology
9.
Nucleic Acids Res ; 46(17): 9201-9219, 2018 09 28.
Article in English | MEDLINE | ID: mdl-30060171

ABSTRACT

The dissemination of multi-resistant bacteria represents an enormous burden on modern healthcare. Plasmid-borne conjugative transfer is the most prevalent mechanism, requiring a type IV secretion system that enables bacteria to spread beneficial traits, such as resistance to last-line antibiotics, among different genera. Inc18 plasmids, like the Gram-positive broad host-range plasmid pIP501, are substantially involved in propagation of vancomycin resistance from Enterococci to methicillin-resistant strains of Staphylococcus aureus. Here, we identified the small cytosolic protein TraN as a repressor of the pIP501-encoded conjugative transfer system, since deletion of traN resulted in upregulation of transfer factors, leading to highly enhanced conjugative transfer. Furthermore, we report the complex structure of TraN with DNA and define the exact sequence of its binding motif. Targeting this protein-DNA interaction might represent a novel therapeutic approach against the spreading of antibiotic resistances.


Subject(s)
Bacterial Outer Membrane Proteins/chemistry , Conjugation, Genetic , DNA, Bacterial/chemistry , Enterococcus faecalis/genetics , Escherichia coli Proteins/chemistry , Plasmids/chemistry , Type IV Secretion Systems/genetics , Amino Acid Sequence , Anti-Bacterial Agents/pharmacology , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Binding Sites , Crystallography, X-Ray , DNA, Bacterial/genetics , DNA, Bacterial/metabolism , Enterococcus faecalis/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gene Deletion , Kinetics , Models, Molecular , Nucleic Acid Conformation , Plasmids/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Sequence Alignment , Thermodynamics , Type IV Secretion Systems/metabolism , Vancomycin/pharmacology , Vancomycin Resistance/genetics
10.
EMBO J ; 32(23): 3041-54, 2013 Nov 27.
Article in English | MEDLINE | ID: mdl-24129513

ABSTRACT

Malfunctioning of the protein α-synuclein is critically involved in the demise of dopaminergic neurons relevant to Parkinson's disease. Nonetheless, the precise mechanisms explaining this pathogenic neuronal cell death remain elusive. Endonuclease G (EndoG) is a mitochondrially localized nuclease that triggers DNA degradation and cell death upon translocation from mitochondria to the nucleus. Here, we show that EndoG displays cytotoxic nuclear localization in dopaminergic neurons of human Parkinson-diseased patients, while EndoG depletion largely reduces α-synuclein-induced cell death in human neuroblastoma cells. Xenogenic expression of human α-synuclein in yeast cells triggers mitochondria-nuclear translocation of EndoG and EndoG-mediated DNA degradation through a mechanism that requires a functional kynurenine pathway and the permeability transition pore. In nematodes and flies, EndoG is essential for the α-synuclein-driven degeneration of dopaminergic neurons. Moreover, the locomotion and survival of α-synuclein-expressing flies is compromised, but reinstalled by parallel depletion of EndoG. In sum, we unravel a phylogenetically conserved pathway that involves EndoG as a critical downstream executor of α-synuclein cytotoxicity.


Subject(s)
Apoptosis , Endodeoxyribonucleases/metabolism , Neuroblastoma/pathology , Neurons/metabolism , Parkinson Disease/pathology , Substantia Nigra/pathology , alpha-Synuclein/metabolism , Aged , Animals , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , DNA Damage/genetics , Dopamine/pharmacology , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Endodeoxyribonucleases/genetics , Humans , Immunoblotting , Immunoenzyme Techniques , Mitochondria/metabolism , Mitochondria/pathology , Neuroblastoma/genetics , Neuroblastoma/metabolism , Neurons/cytology , Oxidative Stress , Parkinson Disease/genetics , Parkinson Disease/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Substantia Nigra/metabolism , Tumor Cells, Cultured , alpha-Synuclein/genetics
11.
Cell Tissue Res ; 367(1): 125-140, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27449929

ABSTRACT

Mitochondrial dysfunction is a common feature of many neurodegenerative diseases, including proteinopathies such as Alzheimer's or Parkinson's disease, which are characterized by the deposition of aggregated proteins in the form of insoluble fibrils or plaques. The distinct molecular processes that eventually result in mitochondrial dysfunction during neurodegeneration are well studied but still not fully understood. However, defects in mitochondrial fission and fusion, mitophagy, oxidative phosphorylation and mitochondrial bioenergetics have been linked to cellular demise. These processes are influenced by the lipid environment within mitochondrial membranes as, besides membrane structure and curvature, recruitment and activity of different proteins also largely depend on the respective lipid composition. Hence, the interaction of neurotoxic proteins with certain lipids and the modification of lipid composition in different cell compartments, in particular mitochondria, decisively impact cell death associated with neurodegeneration. Here, we discuss the relevance of mitochondrial lipids in the pathological alterations that result in neuronal demise, focussing on proteinopathies.


Subject(s)
Lipids/chemistry , Mitochondria/metabolism , Nerve Degeneration/metabolism , Animals , Humans , Mitochondrial Membranes/metabolism , Mitophagy , Models, Biological , Nerve Degeneration/pathology
12.
Plasmid ; 91: 9-18, 2017 05.
Article in English | MEDLINE | ID: mdl-28219792

ABSTRACT

Conjugative transfer plays a major role in the transmission of antibiotic resistance in bacteria. pIP501 is a Gram-positive conjugative model plasmid with the broadest transfer host-range known so far and is frequently found in Enterococcus faecalis and Enterococcus faecium clinical isolates. The pIP501 type IV secretion system is encoded by 15 transfer genes. In this work, we focus on the VirB1-like protein TraG, a modular peptidoglycan metabolizing enzyme, and the VirB8-homolog TraM, a potential member of the translocation channel. By providing full-length traG in trans, but not with a truncated variant, we achieved full recovery of wild type transfer efficiency in the traG-knockout mutant E. faecalis pIP501ΔtraG. With peptidoglycan digestion experiments and tandem mass spectrometry we could assign lytic transglycosylase and endopeptidase activity to TraG, with the CHAP domain alone displaying endopeptidase activity. We identified a novel interaction between TraG and TraM in a bacterial-2-hybrid assay. In addition we found that both proteins localize in focal spots at the E. faecalis cell membrane using immunostaining and fluorescence microscopy. Extracellular protease digestion to evaluate protein cell surface exposure revealed that correct membrane localization of TraM requires the transmembrane helix of TraG. Thus, we suggest an essential role for TraG in the assembly of the pIP501 type IV secretion system.


Subject(s)
Bacterial Proteins/genetics , Base Sequence , Carrier Proteins/genetics , Enterococcus faecalis/genetics , Gene Expression Regulation, Bacterial , Plasmids/chemistry , Sequence Deletion , Bacterial Proteins/metabolism , Binding Sites , Biological Transport , Carrier Proteins/metabolism , Cell Wall/metabolism , Cell Wall/ultrastructure , Conjugation, Genetic , Endopeptidases/genetics , Endopeptidases/metabolism , Enterococcus faecalis/metabolism , Enterococcus faecalis/ultrastructure , Peptidoglycan Glycosyltransferase/genetics , Peptidoglycan Glycosyltransferase/metabolism , Plasmids/metabolism , Protein Binding , Protein Domains , Type IV Secretion Systems/metabolism
13.
PLoS Genet ; 10(5): e1004347, 2014 May.
Article in English | MEDLINE | ID: mdl-24785424

ABSTRACT

Reduced supply of the amino acid methionine increases longevity across species through an as yet elusive mechanism. Here, we report that methionine restriction (MetR) extends yeast chronological lifespan in an autophagy-dependent manner. Single deletion of several genes essential for autophagy (ATG5, ATG7 or ATG8) fully abolished the longevity-enhancing capacity of MetR. While pharmacological or genetic inhibition of TOR1 increased lifespan in methionine-prototroph yeast, TOR1 suppression failed to extend the longevity of methionine-restricted yeast cells. Notably, vacuole-acidity was specifically enhanced by MetR, a phenotype that essentially required autophagy. Overexpression of vacuolar ATPase components (Vma1p or Vph2p) suffices to increase chronological lifespan of methionine-prototrophic yeast. In contrast, lifespan extension upon MetR was prevented by inhibition of vacuolar acidity upon disruption of the vacuolar ATPase. In conclusion, autophagy promotes lifespan extension upon MetR and requires the subsequent stimulation of vacuolar acidification, while it is epistatic to the equally autophagy-dependent anti-aging pathway triggered by TOR1 inhibition or deletion.


Subject(s)
Acids/metabolism , Autophagy , Longevity , Methionine/administration & dosage , Saccharomyces cerevisiae/physiology , Vacuoles/metabolism , Gene Deletion , Genes, Fungal , Hydrogen-Ion Concentration , Saccharomyces cerevisiae/immunology , Saccharomyces cerevisiae/metabolism
14.
EMBO J ; 30(14): 2779-92, 2011 Jun 14.
Article in English | MEDLINE | ID: mdl-21673659

ABSTRACT

Mitochondrial outer membrane permeabilization is a watershed event in the process of apoptosis, which is tightly regulated by a series of pro- and anti-apoptotic proteins belonging to the BCL-2 family, each characteristically possessing a BCL-2 homology domain 3 (BH3). Here, we identify a yeast protein (Ybh3p) that interacts with BCL-X(L) and harbours a functional BH3 domain. Upon lethal insult, Ybh3p translocates to mitochondria and triggers BH3 domain-dependent apoptosis. Ybh3p induces cell death and disruption of the mitochondrial transmembrane potential via the mitochondrial phosphate carrier Mir1p. Deletion of Mir1p and depletion of its human orthologue (SLC25A3/PHC) abolish stress-induced mitochondrial targeting of Ybh3p in yeast and that of BAX in human cells, respectively. Yeast cells lacking YBH3 display prolonged chronological and replicative lifespans and resistance to apoptosis induction. Thus, the yeast genome encodes a functional BH3 domain that induces cell death through phylogenetically conserved mechanisms.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Mitochondria/metabolism , Peptide Fragments/pharmacology , Proto-Oncogene Proteins/pharmacology , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Signal Transduction , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/genetics , Blotting, Western , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Cell Cycle , Flow Cytometry , Humans , Immunoprecipitation , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Membrane Potential, Mitochondrial , Mice , Mitochondria/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Molecular Sequence Data , Phosphate Transport Proteins/genetics , Phosphate Transport Proteins/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/genetics , Sequence Homology, Amino Acid , Tumor Cells, Cultured , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism , bcl-X Protein/genetics , bcl-X Protein/metabolism
15.
Proc Natl Acad Sci U S A ; 109(22): 8658-63, 2012 May 29.
Article in English | MEDLINE | ID: mdl-22586098

ABSTRACT

The large protein superfamily of NADPH oxidases (NOX enzymes) is found in members of all eukaryotic kingdoms: animals, plants, fungi, and protists. The physiological functions of these NOX enzymes range from defense to specialized oxidative biosynthesis and to signaling. In filamentous fungi, NOX enzymes are involved in signaling cell differentiation, in particular in the formation of fruiting bodies. On the basis of bioinformatics analysis, until now it was believed that the genomes of unicellular fungi like Saccharomyces cerevisiae and Schizosaccharomyces pombe do not harbor genes coding for NOX enzymes. Nevertheless, the genome of S. cerevisiae contains nine ORFs showing sequence similarity to the catalytic subunits of mammalian NOX enzymes, only some of which have been functionally assigned as ferric reductases involved in iron ion transport. Here we show that one of the nine ORFs (YGL160W, AIM14) encodes a genuine NADPH oxidase, which is located in the endoplasmic reticulum (ER) and produces superoxide in a NADPH-dependent fashion. We renamed this ORF YNO1 (yeast NADPH oxidase 1). Overexpression of YNO1 causes YCA1-dependent apoptosis, whereas deletion of the gene makes cells less sensitive to apoptotic stimuli. Several independent lines of evidence point to regulation of the actin cytoskeleton by reactive oxygen species (ROS) produced by Yno1p.


Subject(s)
Actins/metabolism , Apoptosis , NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Amino Acid Sequence , Caspases/genetics , Caspases/metabolism , Cytoskeleton/metabolism , Endoplasmic Reticulum/enzymology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Microscopy, Fluorescence , Microscopy, Phase-Contrast , Molecular Sequence Data , Mutation , NADPH Oxidases/classification , NADPH Oxidases/genetics , Open Reading Frames/genetics , Phylogeny , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Superoxides/metabolism
16.
Trends Biochem Sci ; 35(3): 135-44, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19926288

ABSTRACT

Neurodegeneration is characterized by the disease-specific loss of neuronal activity, culminating in the irreversible destruction of neurons. Neuronal cell death can proceed via distinct subroutines such as apoptosis and necrosis, but the underlying molecular mechanisms remain poorly understood. Saccharomyces cerevisiae is an established model for programmed cell death, characterized by distinct cell death pathways conserved from yeast to mammals. Recently, yeast models for several major classes of neurodegeneration, namely alpha-synucleinopathies, polyglutamine disorders, beta-amyloid diseases, tauopathies, and TDP-43 proteinopathies, have been established. Heterologous expression of the human proteins implicated in these disorders has unraveled important insights in their detrimental function, pointing to ways in which yeast might advance the mechanistic dissection of cell death pathways relevant for human neurodegeneration.


Subject(s)
Cell Death/physiology , Models, Biological , Neurodegenerative Diseases/metabolism , Saccharomyces cerevisiae/physiology , Animals , Humans , Necrosis/metabolism , Neurodegenerative Diseases/pathology , Saccharomyces cerevisiae Proteins/metabolism
17.
Yeast ; 31(8): 289-98, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24842537

ABSTRACT

The identification and characterization of the molecular determinants governing ageing represents the key to counteracting age-related diseases and eventually prolonging our health span. A large number of fundamental insights into the ageing process have been provided by research into the budding yeast Saccharomyces cerevisiae, which couples a wide array of technical advantages with a high degree of genetic, proteomic and mechanistic conservation. Indeed, this unicellular organism harbours regulatory pathways, such as those related to programmed cell death or nutrient signalling, that are crucial for ageing control and are reminiscent of other eukaryotes, including mammals. Here, we summarize and discuss three different paradigms of yeast ageing: replicative, chronological and colony ageing. We address their physiological relevance as well as the specific and common characteristics and regulators involved, providing an overview of the network underlying ageing in one of the most important eukaryotic model organisms.


Subject(s)
Microbial Viability , Saccharomyces cerevisiae/physiology , Gene Expression Regulation, Fungal , Gene Regulatory Networks , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development
18.
FEMS Yeast Res ; 14(1): 179-97, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24119111

ABSTRACT

Understanding lipid-induced malfunction represents a major challenge of today's biomedical research. The connection of lipids to cellular and organ dysfunction, cell death, and disease (often referred to as lipotoxicity) is more complex than the sole lipotoxic effects of excess free fatty acids and requires genetically tractable model systems for mechanistic investigation. We herein summarize recent advances in the field of lipid-induced toxicity that employ the established model system for cell death and aging research of budding yeast Saccharomyces cerevisiae. Studies in yeast have shed light on various aspects of lipotoxicity, including free fatty acid toxicity, sphingolipid-modulated cell death as well as the involvement of cardiolipin and lipid peroxidation in the mitochondrial pathways of apoptosis. Regimens used range from exogenously applied lipids, genetic modulation of lipolysis and triacylglyceride synthesis, variations in sphingolipid/ceramide metabolism as well as changes in peroxisome function by either genetic or pharmacological means. In future, the yeast model of programmed cell death will further contribute to the clarification of crucial questions of lipid-associated malfunction.


Subject(s)
Apoptosis , Lipid Metabolism , Lipids/toxicity , Saccharomyces cerevisiae/physiology , Models, Biological
19.
Sci Rep ; 14(1): 13653, 2024 06 13.
Article in English | MEDLINE | ID: mdl-38871812

ABSTRACT

Eukaryotic membranes are compartmentalized into distinct micro- and nanodomains that rearrange dynamically in response to external and internal cues. This lateral heterogeneity of the lipid bilayer and associated clustering of distinct membrane proteins contribute to the spatial organization of numerous cellular processes. Here, we show that membrane microdomains within the endoplasmic reticulum (ER) of yeast cells are reorganized during metabolic reprogramming and aging. Using biosensors with varying transmembrane domain length to map lipid bilayer thickness, we demonstrate that in young cells, microdomains of increased thickness mainly exist within the nuclear ER, while progressing cellular age drives the formation of numerous microdomains specifically in the cortical ER. Partitioning of biosensors with long transmembrane domains into these microdomains increased protein stability and prevented autophagic removal. In contrast, reporters with short transmembrane domains progressively accumulated at the membrane contact site between the nuclear ER and the vacuole, the so-called nucleus-vacuole junction (NVJ), and were subjected to turnover via selective microautophagy occurring specifically at these sites. Reporters with long transmembrane domains were excluded from the NVJ. Our data reveal age-dependent rearrangement of the lateral organization of the ER and establish transmembrane domain length as a determinant of membrane contact site localization and autophagic degradation.


Subject(s)
Autophagy , Cellular Senescence , Endoplasmic Reticulum , Membrane Microdomains , Saccharomyces cerevisiae , Endoplasmic Reticulum/metabolism , Saccharomyces cerevisiae/metabolism , Membrane Microdomains/metabolism , Cellular Senescence/physiology , Saccharomyces cerevisiae Proteins/metabolism , Vacuoles/metabolism , Membrane Proteins/metabolism
20.
Nat Commun ; 15(1): 315, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-38182580

ABSTRACT

The resilience of cellular proteostasis declines with age, which drives protein aggregation and compromises viability. The nucleus has emerged as a key quality control compartment that handles misfolded proteins produced by the cytosolic protein biosynthesis system. Here, we find that age-associated metabolic cues target the yeast protein disaggregase Hsp104 to the nucleus to maintain a functional nuclear proteome during quiescence. The switch to respiratory metabolism and the accompanying decrease in translation rates direct cytosolic Hsp104 to the nucleus to interact with latent translation initiation factor eIF2 and to suppress protein aggregation. Hindering Hsp104 from entering the nucleus in quiescent cells results in delayed re-entry into the cell cycle due to compromised resumption of protein synthesis. In sum, we report that cytosolic-nuclear partitioning of the Hsp104 disaggregase is a critical mechanism to protect the latent protein synthesis machinery during quiescence in yeast, ensuring the rapid restart of translation once nutrients are replenished.


Subject(s)
Heat-Shock Proteins , Protein Biosynthesis , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Cell Cycle , Cell Division , Cytosol , Protein Aggregates , Saccharomyces cerevisiae/metabolism , Heat-Shock Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL