Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 181.751
Filter
Add more filters

Publication year range
1.
Annu Rev Biochem ; 93(1): 233-259, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38621235

ABSTRACT

Peroxisomes are organelles that play a central role in lipid metabolism and cellular redox homeostasis. The import of peroxisomal matrix proteins by peroxisomal targeting signal (PTS) receptors is an ATP-dependent mechanism. However, the energy-dependent steps do not occur early during the binding of the receptor-cargo complex to the membrane but late, because they are linked to the peroxisomal export complex for the release of the unloaded receptor. The first ATP-demanding step is the cysteine-dependent monoubiquitination of the PTS receptors, which is required for recognition by the AAA+ peroxins. They execute the second ATP-dependent step by extracting the ubiqitinated PTS receptors from the membrane for release back to the cytosol. After deubiquitination, the PTS receptors regain import competence and can facilitate further rounds of cargo import. Here, we give a general overview and discuss recent data regarding the ATP-dependent steps in peroxisome protein import.


Subject(s)
Adenosine Triphosphate , Peroxisomes , Protein Transport , Ubiquitination , Peroxisomes/metabolism , Adenosine Triphosphate/metabolism , Humans , Animals , Peroxisome-Targeting Signal 1 Receptor/metabolism , Peroxisome-Targeting Signal 1 Receptor/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Peroxisomal Targeting Signals , Peroxins/metabolism , Peroxins/genetics , Membrane Proteins
2.
Cell ; 187(5): 1109-1126.e21, 2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38382525

ABSTRACT

Oocytes are among the longest-lived cells in the body and need to preserve their cytoplasm to support proper embryonic development. Protein aggregation is a major threat for intracellular homeostasis in long-lived cells. How oocytes cope with protein aggregation during their extended life is unknown. Here, we find that mouse oocytes accumulate protein aggregates in specialized compartments that we named endolysosomal vesicular assemblies (ELVAs). Combining live-cell imaging, electron microscopy, and proteomics, we found that ELVAs are non-membrane-bound compartments composed of endolysosomes, autophagosomes, and proteasomes held together by a protein matrix formed by RUFY1. Functional assays revealed that in immature oocytes, ELVAs sequester aggregated proteins, including TDP-43, and degrade them upon oocyte maturation. Inhibiting degradative activity in ELVAs leads to the accumulation of protein aggregates in the embryo and is detrimental for embryo survival. Thus, ELVAs represent a strategy to safeguard protein homeostasis in long-lived cells.


Subject(s)
Cytoplasmic Vesicles , Oocytes , Protein Aggregates , Animals , Female , Mice , Autophagosomes , Cytoplasmic Vesicles/metabolism , Lysosomes/metabolism , Oocytes/cytology , Oocytes/metabolism , Proteasome Endopeptidase Complex , Proteolysis
3.
Cell ; 187(14): 3619-3637.e27, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38851188

ABSTRACT

Mitochondrial dynamics play a critical role in cell fate decisions and in controlling mtDNA levels and distribution. However, the molecular mechanisms linking mitochondrial membrane remodeling and quality control to mtDNA copy number (CN) regulation remain elusive. Here, we demonstrate that the inner mitochondrial membrane (IMM) protein mitochondrial fission process 1 (MTFP1) negatively regulates IMM fusion. Moreover, manipulation of mitochondrial fusion through the regulation of MTFP1 levels results in mtDNA CN modulation. Mechanistically, we found that MTFP1 inhibits mitochondrial fusion to isolate and exclude damaged IMM subdomains from the rest of the network. Subsequently, peripheral fission ensures their segregation into small MTFP1-enriched mitochondria (SMEM) that are targeted for degradation in an autophagic-dependent manner. Remarkably, MTFP1-dependent IMM quality control is essential for basal nucleoid recycling and therefore to maintain adequate mtDNA levels within the cell.


Subject(s)
DNA, Mitochondrial , Mitochondria , Mitochondrial Dynamics , Mitochondrial Membranes , Mitochondrial Proteins , DNA, Mitochondrial/metabolism , DNA, Mitochondrial/genetics , Mitochondrial Proteins/metabolism , Humans , Mitochondrial Membranes/metabolism , Mitochondria/metabolism , Animals , HeLa Cells , Mice , Membrane Proteins/metabolism , Membrane Proteins/genetics , Autophagy
4.
Cell ; 186(16): 3443-3459.e24, 2023 08 03.
Article in English | MEDLINE | ID: mdl-37480851

ABSTRACT

Cells contain numerous abundant molecular machines assembled from multiple subunits. Imbalances in subunit production and failed assembly generate orphan subunits that are eliminated by poorly defined pathways. Here, we determined how orphan subunits of the cytosolic chaperonin CCT are recognized. Several unassembled CCT subunits recruited the E3 ubiquitin ligase HERC2 using ZNRD2 as an adaptor. Both factors were necessary for orphan CCT subunit degradation in cells, sufficient for CCT subunit ubiquitination with purified factors, and necessary for optimal cell fitness. Domain mapping and structure prediction defined the molecular features of a minimal HERC2-ZNRD2-CCT module. The structural model, whose key elements were validated in cells using point mutants, shows why ZNRD2 selectively recognizes multiple orphaned CCT subunits without engaging assembled CCT. Our findings reveal how failures during CCT assembly are monitored and provide a paradigm for the molecular recognition of orphan subunits, the largest source of quality control substrates in cells.


Subject(s)
Chaperonin Containing TCP-1 , Ubiquitin-Protein Ligases , Chaperonin Containing TCP-1/chemistry , Ubiquitin-Protein Ligases/genetics , Humans
5.
Cell ; 186(16): 3460-3475.e23, 2023 08 03.
Article in English | MEDLINE | ID: mdl-37478862

ABSTRACT

All eukaryotes require intricate protein networks to translate developmental signals into accurate cell fate decisions. Mutations that disturb interactions between network components often result in disease, but how the composition and dynamics of complex networks are established remains poorly understood. Here, we identify the E3 ligase UBR5 as a signaling hub that helps degrade unpaired subunits of multiple transcriptional regulators that act within a network centered on the c-Myc oncoprotein. Biochemical and structural analyses show that UBR5 binds motifs that only become available upon complex dissociation. By rapidly turning over unpaired transcription factor subunits, UBR5 establishes dynamic interactions between transcriptional regulators that allow cells to effectively execute gene expression while remaining receptive to environmental signals. We conclude that orphan quality control plays an essential role in establishing dynamic protein networks, which may explain the conserved need for protein degradation during transcription and offers opportunities to modulate gene expression in disease.


Subject(s)
Transcription Factors , Ubiquitin-Protein Ligases , Humans , Gene Expression , HEK293 Cells , HeLa Cells , Mutation , Signal Transduction , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism
6.
Cell ; 186(10): 2044-2061, 2023 05 11.
Article in English | MEDLINE | ID: mdl-37172561

ABSTRACT

Phenotypic sex-based differences exist for many complex traits. In other cases, phenotypes may be similar, but underlying biology may vary. Thus, sex-aware genetic analyses are becoming increasingly important for understanding the mechanisms driving these differences. To this end, we provide a guide outlining the current best practices for testing various models of sex-dependent genetic effects in complex traits and disease conditions, noting that this is an evolving field. Insights from sex-aware analyses will not only teach us about the biology of complex traits but also aid in achieving the goals of precision medicine and health equity for all.


Subject(s)
Models, Genetic , Sex Characteristics , Animals , Female , Male , Multifactorial Inheritance , Phenotype , Quality Control , Genome-Wide Association Study , Guidelines as Topic , Gene-Environment Interaction , Humans
7.
Cell ; 186(10): 2176-2192.e22, 2023 05 11.
Article in English | MEDLINE | ID: mdl-37137307

ABSTRACT

The ClpC1:ClpP1P2 protease is a core component of the proteostasis system in mycobacteria. To improve the efficacy of antitubercular agents targeting the Clp protease, we characterized the mechanism of the antibiotics cyclomarin A and ecumicin. Quantitative proteomics revealed that the antibiotics cause massive proteome imbalances, including upregulation of two unannotated yet conserved stress response factors, ClpC2 and ClpC3. These proteins likely protect the Clp protease from excessive amounts of misfolded proteins or from cyclomarin A, which we show to mimic damaged proteins. To overcome the Clp security system, we developed a BacPROTAC that induces degradation of ClpC1 together with its ClpC2 caretaker. The dual Clp degrader, built from linked cyclomarin A heads, was highly efficient in killing pathogenic Mycobacterium tuberculosis, with >100-fold increased potency over the parent antibiotic. Together, our data reveal Clp scavenger proteins as important proteostasis safeguards and highlight the potential of BacPROTACs as future antibiotics.


Subject(s)
Antitubercular Agents , Mycobacterium tuberculosis , Antitubercular Agents/pharmacology , Bacterial Proteins/metabolism , Endopeptidase Clp/metabolism , Heat-Shock Proteins/metabolism , Mycobacterium tuberculosis/drug effects , Proteostasis
8.
Cell ; 186(2): 346-362.e17, 2023 01 19.
Article in English | MEDLINE | ID: mdl-36638793

ABSTRACT

Ribosomes frequently stall during mRNA translation, resulting in the context-dependent activation of quality control pathways to maintain proteostasis. However, surveillance mechanisms that specifically respond to stalled ribosomes with an occluded A site have not been identified. We discovered that the elongation factor-1α (eEF1A) inhibitor, ternatin-4, triggers the ubiquitination and degradation of eEF1A on stalled ribosomes. Using a chemical genetic approach, we unveiled a signaling network comprising two E3 ligases, RNF14 and RNF25, which are required for eEF1A degradation. Quantitative proteomics revealed the RNF14 and RNF25-dependent ubiquitination of eEF1A and a discrete set of ribosomal proteins. The ribosome collision sensor GCN1 plays an essential role by engaging RNF14, which directly ubiquitinates eEF1A. The site-specific, RNF25-dependent ubiquitination of the ribosomal protein RPS27A/eS31 provides a second essential signaling input. Our findings illuminate a ubiquitin signaling network that monitors the ribosomal A site and promotes the degradation of stalled translation factors, including eEF1A and the termination factor eRF1.


Subject(s)
RNA-Binding Proteins , Trans-Activators , Carrier Proteins/metabolism , Peptide Elongation Factors/genetics , Protein Biosynthesis , Ribosomal Proteins/metabolism , Ribosomes/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Humans , HeLa Cells , HEK293 Cells , RNA-Binding Proteins/metabolism , Trans-Activators/metabolism , Peptide Elongation Factor 1/metabolism
9.
Annu Rev Biochem ; 90: 631-658, 2021 06 20.
Article in English | MEDLINE | ID: mdl-33823651

ABSTRACT

Collagen is the most abundant protein in mammals. A unique feature of collagen is its triple-helical structure formed by the Gly-Xaa-Yaa repeats. Three single chains of procollagen make a trimer, and the triple-helical structure is then folded in the endoplasmic reticulum (ER). This unique structure is essential for collagen's functions in vivo, including imparting bone strength, allowing signal transduction, and forming basement membranes. The triple-helical structure of procollagen is stabilized by posttranslational modifications and intermolecular interactions, but collagen is labile even at normal body temperature. Heat shock protein 47 (Hsp47) is a collagen-specific molecular chaperone residing in the ER that plays a pivotal role in collagen biosynthesis and quality control of procollagen in the ER. Mutations that affect the triple-helical structure or result in loss of Hsp47 activity cause the destabilization of procollagen, which is then degraded by autophagy. In this review, we present the current state of the field regarding quality control of procollagen.


Subject(s)
Collagen/chemistry , Fibrosis/metabolism , HSP47 Heat-Shock Proteins/metabolism , Procollagen/chemistry , Procollagen/metabolism , Animals , Collagen/metabolism , Endoplasmic Reticulum/metabolism , Fibrosis/genetics , HSP47 Heat-Shock Proteins/chemistry , HSP47 Heat-Shock Proteins/genetics , Humans , Hydroxylation , Molecular Chaperones/metabolism , Proline/chemistry , Proline/metabolism , Protein Conformation , Protein Folding , Protein Processing, Post-Translational
10.
Cell ; 184(11): 2896-2910.e13, 2021 05 27.
Article in English | MEDLINE | ID: mdl-34048705

ABSTRACT

Damaged mitochondria need to be cleared to maintain the quality of the mitochondrial pool. Here, we report mitocytosis, a migrasome-mediated mitochondrial quality-control process. We found that, upon exposure to mild mitochondrial stresses, damaged mitochondria are transported into migrasomes and subsequently disposed of from migrating cells. Mechanistically, mitocytosis requires positioning of damaged mitochondria at the cell periphery, which occurs because damaged mitochondria avoid binding to inward motor proteins. Functionally, mitocytosis plays an important role in maintaining mitochondrial quality. Enhanced mitocytosis protects cells from mitochondrial stressor-induced loss of mitochondrial membrane potential (MMP) and mitochondrial respiration; conversely, blocking mitocytosis causes loss of MMP and mitochondrial respiration under normal conditions. Physiologically, we demonstrate that mitocytosis is required for maintaining MMP and viability in neutrophils in vivo. We propose that mitocytosis is an important mitochondrial quality-control process in migrating cells, which couples mitochondrial homeostasis with cell migration.


Subject(s)
Membrane Potential, Mitochondrial/physiology , Mitochondria/metabolism , Animals , Biological Transport , Cell Line , Cell Movement/physiology , Cytoplasm/metabolism , Exocytosis/physiology , Female , Homeostasis , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission/methods , Mitochondria/physiology , Mitochondrial Membranes/metabolism , Organelles/metabolism
11.
Cell ; 184(13): 3542-3558.e16, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34051138

ABSTRACT

Structural variations (SVs) and gene copy number variations (gCNVs) have contributed to crop evolution, domestication, and improvement. Here, we assembled 31 high-quality genomes of genetically diverse rice accessions. Coupling with two existing assemblies, we developed pan-genome-scale genomic resources including a graph-based genome, providing access to rice genomic variations. Specifically, we discovered 171,072 SVs and 25,549 gCNVs and used an Oryza glaberrima assembly to infer the derived states of SVs in the Oryza sativa population. Our analyses of SV formation mechanisms, impacts on gene expression, and distributions among subpopulations illustrate the utility of these resources for understanding how SVs and gCNVs shaped rice environmental adaptation and domestication. Our graph-based genome enabled genome-wide association study (GWAS)-based identification of phenotype-associated genetic variations undetectable when using only SNPs and a single reference assembly. Our work provides rich population-scale resources paired with easy-to-access tools to facilitate rice breeding as well as plant functional genomics and evolutionary biology research.


Subject(s)
Ecotype , Genetic Variation , Genome, Plant , Oryza/genetics , Adaptation, Physiological/genetics , Agriculture , Domestication , Gene Expression Profiling , Gene Expression Regulation, Plant , Genes, Plant , Genomic Structural Variation , Molecular Sequence Annotation , Phenotype
12.
Cell ; 184(14): 3626-3642.e14, 2021 07 08.
Article in English | MEDLINE | ID: mdl-34186018

ABSTRACT

All cells fold their genomes, including bacterial cells, where the chromosome is compacted into a domain-organized meshwork called the nucleoid. How compaction and domain organization arise is not fully understood. Here, we describe a method to estimate the average mesh size of the nucleoid in Escherichia coli. Using nucleoid mesh size and DNA concentration estimates, we find that the cytoplasm behaves as a poor solvent for the chromosome when the cell is considered as a simple semidilute polymer solution. Monte Carlo simulations suggest that a poor solvent leads to chromosome compaction and DNA density heterogeneity (i.e., domain formation) at physiological DNA concentration. Fluorescence microscopy reveals that the heterogeneous DNA density negatively correlates with ribosome density within the nucleoid, consistent with cryoelectron tomography data. Drug experiments, together with past observations, suggest the hypothesis that RNAs contribute to the poor solvent effects, connecting chromosome compaction and domain formation to transcription and intracellular organization.


Subject(s)
Chromosomes, Bacterial/chemistry , Escherichia coli/metabolism , Nucleic Acid Conformation , Solvents/chemistry , Transcription, Genetic , Aminoglycosides/pharmacology , Computer Simulation , DNA, Bacterial/chemistry , Diffusion , Escherichia coli/drug effects , Green Fluorescent Proteins/metabolism , Particle Size , RNA, Bacterial/metabolism , Ribosomes/metabolism , Ribosomes/ultrastructure , Transcription, Genetic/drug effects
13.
Annu Rev Cell Dev Biol ; 38: 241-262, 2022 10 06.
Article in English | MEDLINE | ID: mdl-35587265

ABSTRACT

While cellular proteins were initially thought to be stable, research over the last decades has firmly established that intracellular protein degradation is an active and highly regulated process: Lysosomal, proteasomal, and mitochondrial degradation systems were identified and found to be involved in a staggering number of biological functions. Here, we provide a global overview of the diverse roles of cellular protein degradation using seven categories: homeostasis, regulation, quality control, stoichiometry control, proteome remodeling, immune surveillance, and baseline turnover. Using selected examples, we outline how proteins are degraded and why this is functionally relevant.


Subject(s)
Autophagy , Proteome , Autophagy/genetics , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Proteome/metabolism , Ubiquitination
14.
Annu Rev Biochem ; 89: 501-528, 2020 06 20.
Article in English | MEDLINE | ID: mdl-32075415

ABSTRACT

Mitochondria are essential metabolic hubs that dynamically adapt to physiological demands. More than 40 proteases residing in different compartments of mitochondria, termed mitoproteases, preserve mitochondrial proteostasis and are emerging as central regulators of mitochondrial plasticity. These multifaceted enzymes limit the accumulation of short-lived, regulatory proteins within mitochondria, modulate the activity of mitochondrial proteins by protein processing, and mediate the degradation of damaged proteins. Various signaling cascades coordinate the activity of mitoproteases to preserve mitochondrial homeostasis and ensure cell survival. Loss of mitoproteases severely impairs the functional integrity of mitochondria, is associated with aging, and causes pleiotropic diseases. Understanding the dual function of mitoproteases as regulatory and quality control enzymes will help unravel the role of mitochondrial plasticity in aging and disease.


Subject(s)
Aging/genetics , Mitochondria/genetics , Mitochondrial Proteins/chemistry , Neoplasms/genetics , Neurodegenerative Diseases/genetics , Peptide Hydrolases/chemistry , Aging/metabolism , Animals , Apoptosis/genetics , Gene Expression Regulation , Homeostasis/genetics , Humans , Lipid Metabolism/genetics , Mitochondria/enzymology , Mitochondrial Dynamics/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mitophagy/genetics , Neoplasms/enzymology , Neoplasms/pathology , Neurodegenerative Diseases/enzymology , Neurodegenerative Diseases/pathology , Peptide Hydrolases/genetics , Peptide Hydrolases/metabolism , Phospholipids/metabolism , Proteolysis , Proteostasis/genetics
15.
Annu Rev Biochem ; 89: 21-43, 2020 06 20.
Article in English | MEDLINE | ID: mdl-32569520

ABSTRACT

My coworkers and I have used animal viruses and their interaction with host cells to investigate cellular processes difficult to study by other means. This approach has allowed us to branch out in many directions, including membrane protein characterization, endocytosis, secretion, protein folding, quality control, and glycobiology. At the same time, our aim has been to employ cell biological approaches to expand the fundamental understanding of animal viruses and their pathogenic lifestyles. We have studied mechanisms of host cell entry and the uncoating of incoming viruses as well as the synthesis, folding, maturation, and intracellular movement of viral proteins and molecular assemblies. I have had the privilege to work in institutions in four different countries. The early years in Finland (the University of Helsinki) were followed by 6 years in Germany (European Molecular Biology Laboratory), 16 years in the United States (Yale School of Medicine), and 16 years in Switzerland (ETH Zurich).


Subject(s)
Calnexin/genetics , Calreticulin/genetics , Host-Pathogen Interactions/genetics , Influenza A virus/genetics , Picornaviridae/genetics , Viral Proteins/genetics , Virology/history , Animals , Calnexin/chemistry , Calnexin/metabolism , Calreticulin/chemistry , Calreticulin/metabolism , Cell Line , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/virology , Endosomes/metabolism , Endosomes/virology , Gene Expression Regulation , History, 20th Century , History, 21st Century , Humans , Influenza A virus/metabolism , Picornaviridae/metabolism , Protein Folding , Semliki forest virus/genetics , Semliki forest virus/metabolism , Vesiculovirus/genetics , Vesiculovirus/metabolism , Viral Proteins/chemistry , Viral Proteins/metabolism , Virus Internalization
16.
Annu Rev Biochem ; 89: 529-555, 2020 06 20.
Article in English | MEDLINE | ID: mdl-32097570

ABSTRACT

Protein folding in the cell is mediated by an extensive network of >1,000 chaperones, quality control factors, and trafficking mechanisms collectively termed the proteostasis network. While the components and organization of this network are generally well established, our understanding of how protein-folding problems are identified, how the network components integrate to successfully address challenges, and what types of biophysical issues each proteostasis network component is capable of addressing remains immature. We describe a chemical biology-informed framework for studying cellular proteostasis that relies on selection of interesting protein-folding problems and precise researcher control of proteostasis network composition and activities. By combining these methods with multifaceted strategies to monitor protein folding, degradation, trafficking, and aggregation in cells, researchers continue to rapidly generate new insights into cellular proteostasis.


Subject(s)
Molecular Chaperones/genetics , Molecular Probe Techniques , Proteome/genetics , Proteostasis Deficiencies/genetics , Proteostasis/genetics , Animals , CRISPR-Cas Systems , Gene Expression Regulation , Half-Life , Heat-Shock Response/drug effects , Humans , Molecular Chaperones/metabolism , Protein Aggregates , Protein Engineering/methods , Protein Folding/drug effects , Protein Transport/drug effects , Proteome/chemistry , Proteome/metabolism , Proteostasis/drug effects , Proteostasis Deficiencies/metabolism , Proteostasis Deficiencies/pathology , Signal Transduction , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/pharmacology , Unfolded Protein Response/drug effects
17.
Cell ; 180(1): 9-14, 2020 01 09.
Article in English | MEDLINE | ID: mdl-31951522

ABSTRACT

This commentary introduces a new clinical trial construct, the Master Observational Trial (MOT), which hybridizes the power of molecularly based master interventional protocols with the breadth of real-world data. The MOT provides a clinical venue to allow molecular medicine to rapidly advance, answers questions that traditional interventional trials generally do not address, and seamlessly integrates with interventional trials in both diagnostic and therapeutic arenas. The result is a more comprehensive data collection ecosystem in precision medicine.


Subject(s)
Observational Studies as Topic/methods , Precision Medicine/methods , Research Design/standards , Big Data , Clinical Trial Protocols as Topic , Humans , Molecular Targeted Therapy/methods , Molecular Targeted Therapy/trends , Observational Studies as Topic/standards
18.
Cell ; 178(1): 76-90.e22, 2019 06 27.
Article in English | MEDLINE | ID: mdl-31155236

ABSTRACT

In ribosome-associated quality control (RQC), Rqc2/NEMF closely supports the E3 ligase Ltn1/listerin in promoting ubiquitylation and degradation of aberrant nascent-chains obstructing large (60S) ribosomal subunits-products of ribosome stalling during translation. However, while Ltn1 is eukaryote-specific, Rqc2 homologs are also found in bacteria and archaea; whether prokaryotic Rqc2 has an RQC-related function has remained unknown. Here, we show that, as in eukaryotes, a bacterial Rqc2 homolog (RqcH) recognizes obstructed 50S subunits and promotes nascent-chain proteolysis. Unexpectedly, RqcH marks nascent-chains for degradation in a direct manner, by appending C-terminal poly-alanine tails that act as degrons recognized by the ClpXP protease. Furthermore, RqcH acts redundantly with tmRNA/ssrA and protects cells against translational and environmental stresses. Our results uncover a proteolytic-tagging mechanism with implications toward the function of related modifications in eukaryotes and suggest that RQC was already active in the last universal common ancestor (LUCA) to help cope with incomplete translation.


Subject(s)
Alanine/metabolism , Bacillus subtilis/metabolism , Prokaryotic Cells/metabolism , Proteolysis , Ribosome Subunits, Large, Bacterial/metabolism , Eukaryotic Cells/metabolism , Protein Biosynthesis , RNA, Messenger/metabolism , RNA, Transfer/metabolism , RNA-Binding Proteins/metabolism , Ribosome Subunits, Large, Eukaryotic/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
19.
Cell ; 177(3): 737-750.e15, 2019 04 18.
Article in English | MEDLINE | ID: mdl-31002798

ABSTRACT

The proteasome mediates selective protein degradation and is dynamically regulated in response to proteotoxic challenges. SKN-1A/Nrf1, an endoplasmic reticulum (ER)-associated transcription factor that undergoes N-linked glycosylation, serves as a sensor of proteasome dysfunction and triggers compensatory upregulation of proteasome subunit genes. Here, we show that the PNG-1/NGLY1 peptide:N-glycanase edits the sequence of SKN-1A protein by converting particular N-glycosylated asparagine residues to aspartic acid. Genetically introducing aspartates at these N-glycosylation sites bypasses the requirement for PNG-1/NGLY1, showing that protein sequence editing rather than deglycosylation is key to SKN-1A function. This pathway is required to maintain sufficient proteasome expression and activity, and SKN-1A hyperactivation confers resistance to the proteotoxicity of human amyloid beta peptide. Deglycosylation-dependent protein sequence editing explains how ER-associated and cytosolic isoforms of SKN-1 perform distinct cytoprotective functions corresponding to those of mammalian Nrf1 and Nrf2. Thus, we uncover an unexpected mechanism by which N-linked glycosylation regulates protein function and proteostasis.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , DNA-Binding Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Asparagine/metabolism , Bortezomib/pharmacology , CRISPR-Cas Systems/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Endoplasmic Reticulum/metabolism , Gene Editing , Gene Expression Regulation/drug effects , Oxidative Stress , Proteasome Endopeptidase Complex/genetics , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Sequence Alignment , Transcription Factors/chemistry , Transcription Factors/genetics
20.
Annu Rev Biochem ; 87: 725-749, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29925261

ABSTRACT

Nuclear proteins participate in diverse cellular processes, many of which are essential for cell survival and viability. To maintain optimal nuclear physiology, the cell employs the ubiquitin-proteasome system to eliminate damaged and misfolded proteins in the nucleus that could otherwise harm the cell. In this review, we highlight the current knowledge about the major ubiquitin-protein ligases involved in protein quality control degradation (PQCD) in the nucleus and how they orchestrate their functions to eliminate misfolded proteins in different nuclear subcompartments. Many human disorders are causally linked to protein misfolding in the nucleus, hence we discuss major concepts that still need to be clarified to better understand the basis of the nuclear misfolded proteins' toxic effects. Additionally, we touch upon potential strategies for manipulating nuclear PQCD pathways to ameliorate diseases associated with protein misfolding and aggregation in the nucleus.


Subject(s)
Cell Nucleus/metabolism , Nuclear Proteins/metabolism , Proteolysis , Aging/metabolism , Humans , Metabolic Networks and Pathways , Models, Biological , Neoplasms/metabolism , Nuclear Envelope/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Aggregation, Pathological/metabolism , Protein Biosynthesis , Protein Folding , Proteostasis Deficiencies/metabolism , Saccharomyces cerevisiae/metabolism , Stress, Physiological , Substrate Specificity , Ubiquitin-Protein Ligases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL