Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 8.238
Filter
Add more filters

Publication year range
1.
Cell ; 187(11): 2875-2892.e21, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38626770

ABSTRACT

Ubiquitylation regulates most proteins and biological processes in a eukaryotic cell. However, the site-specific occupancy (stoichiometry) and turnover rate of ubiquitylation have not been quantified. Here we present an integrated picture of the global ubiquitylation site occupancy and half-life. Ubiquitylation site occupancy spans over four orders of magnitude, but the median ubiquitylation site occupancy is three orders of magnitude lower than that of phosphorylation. The occupancy, turnover rate, and regulation of sites by proteasome inhibitors are strongly interrelated, and these attributes distinguish sites involved in proteasomal degradation and cellular signaling. Sites in structured protein regions exhibit longer half-lives and stronger upregulation by proteasome inhibitors than sites in unstructured regions. Importantly, we discovered a surveillance mechanism that rapidly and site-indiscriminately deubiquitylates all ubiquitin-specific E1 and E2 enzymes, protecting them against accumulation of bystander ubiquitylation. The work provides a systems-scale, quantitative view of ubiquitylation properties and reveals general principles of ubiquitylation-dependent governance.


Subject(s)
Proteasome Endopeptidase Complex , Ubiquitination , Humans , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Proteolysis , Ubiquitin/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Animals , Mice , Cell Line
2.
Cell ; 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38942013

ABSTRACT

The ability of proteins and RNA to coalesce into phase-separated assemblies, such as the nucleolus and stress granules, is a basic principle in organizing membraneless cellular compartments. While the constituents of biomolecular condensates are generally well documented, the mechanisms underlying their formation under stress are only partially understood. Here, we show in yeast that covalent modification with the ubiquitin-like modifier Urm1 promotes the phase separation of a wide range of proteins. We find that the drop in cellular pH induced by stress triggers Urm1 self-association and its interaction with both target proteins and the Urm1-conjugating enzyme Uba4. Urmylation of stress-sensitive proteins promotes their deposition into stress granules and nuclear condensates. Yeast cells lacking Urm1 exhibit condensate defects that manifest in reduced stress resilience. We propose that Urm1 acts as a reversible molecular "adhesive" to drive protective phase separation of functionally critical proteins under cellular stress.

3.
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
4.
Cell ; 186(9): 1895-1911.e21, 2023 04 27.
Article in English | MEDLINE | ID: mdl-37028429

ABSTRACT

Cells respond to environmental cues by remodeling their inventories of multiprotein complexes. Cellular repertoires of SCF (SKP1-CUL1-F box protein) ubiquitin ligase complexes, which mediate much protein degradation, require CAND1 to distribute the limiting CUL1 subunit across the family of ∼70 different F box proteins. Yet, how a single factor coordinately assembles numerous distinct multiprotein complexes remains unknown. We obtained cryo-EM structures of CAND1-bound SCF complexes in multiple states and correlated mutational effects on structures, biochemistry, and cellular assays. The data suggest that CAND1 clasps idling catalytic domains of an inactive SCF, rolls around, and allosterically rocks and destabilizes the SCF. New SCF production proceeds in reverse, through SKP1-F box allosterically destabilizing CAND1. The CAND1-SCF conformational ensemble recycles CUL1 from inactive complexes, fueling mixing and matching of SCF parts for E3 activation in response to substrate availability. Our data reveal biogenesis of a predominant family of E3 ligases, and the molecular basis for systemwide multiprotein complex assembly.


Subject(s)
Cullin Proteins , F-Box Proteins , SKP Cullin F-Box Protein Ligases , Transcription Factors , Humans , Cullin Proteins/chemistry , Cullin Proteins/metabolism , F-Box Proteins/metabolism , Molecular Conformation , SKP Cullin F-Box Protein Ligases/chemistry , SKP Cullin F-Box Protein Ligases/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism
5.
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
6.
Annu Rev Biochem ; 91: 295-319, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35320687

ABSTRACT

Methods to direct the degradation of protein targets with proximity-inducing molecules that coopt the cellular degradation machinery are advancing in leaps and bounds, and diverse modalities are emerging. The most used and well-studied approach is to hijack E3 ligases of the ubiquitin-proteasome system. E3 ligases use specific molecular recognition to determine which proteins in the cell are ubiquitinated and degraded. This review focuses on the structural determinants of E3 ligase recruitment of natural substrates and neo-substrates obtained through monovalent molecular glues and bivalent proteolysis-targeting chimeras. We use structures to illustrate the different types of substrate recognition and assess the basis for neo-protein-protein interactions in ternary complex structures. The emerging structural and mechanistic complexity is reflective of the diverse physiological roles of protein ubiquitination. This molecular insight is also guiding the application of structure-based design approaches to the development of new and existing degraders as chemical tools and therapeutics.


Subject(s)
Ubiquitin-Protein Ligases , Ubiquitin , Proteins/metabolism , Proteolysis , Substrate Specificity , Ubiquitin/genetics , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
7.
Cell ; 185(21): 3913-3930.e19, 2022 10 13.
Article in English | MEDLINE | ID: mdl-36198316

ABSTRACT

Although women experience significantly higher tau burden and increased risk for Alzheimer's disease (AD) than men, the underlying mechanism for this vulnerability has not been explained. Here, we demonstrate through in vitro and in vivo models, as well as human AD brain tissue, that X-linked ubiquitin specific peptidase 11 (USP11) augments pathological tau aggregation via tau deubiquitination initiated at lysine-281. Removal of ubiquitin provides access for enzymatic tau acetylation at lysines 281 and 274. USP11 escapes complete X-inactivation, and female mice and people both exhibit higher USP11 levels than males. Genetic elimination of usp11 in a tauopathy mouse model preferentially protects females from acetylated tau accumulation, tau pathology, and cognitive impairment. USP11 levels also strongly associate positively with tau pathology in females but not males. Thus, inhibiting USP11-mediated tau deubiquitination may provide an effective therapeutic opportunity to protect women from increased vulnerability to AD and other tauopathies.


Subject(s)
Alzheimer Disease , Tauopathies , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Brain/metabolism , Brain/pathology , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Transgenic , Sex Characteristics , Tauopathies/genetics , Tauopathies/pathology , Thiolester Hydrolases/genetics , Ubiquitin-Specific Proteases , tau Proteins/genetics
8.
Annu Rev Biochem ; 90: 659-679, 2021 06 20.
Article in English | MEDLINE | ID: mdl-34153214

ABSTRACT

The polytopic, endoplasmic reticulum (ER) membrane protein 3-hydroxy-3-methylglutaryl coenzyme A (HMG CoA) reductase produces mevalonate, the key intermediate in the synthesis of cholesterol and many nonsterol isoprenoids including geranylgeranyl pyrophosphate (GGpp). Transcriptional, translational, and posttranslational feedback mechanisms converge on this reductase to ensure cells maintain a sufficient supply of essential nonsterol isoprenoids but avoid overaccumulation of cholesterol and other sterols. The focus of this review is mechanisms for the posttranslational regulation of HMG CoA reductase, which include sterol-accelerated ubiquitination and ER-associated degradation (ERAD) that is augmented by GGpp. We discuss how GGpp-induced ER-to-Golgi trafficking of the vitamin K2 synthetic enzyme UbiA prenyltransferase domain-containing protein-1 (UBIAD1) modulates HMG CoA reductase ERAD to balance the synthesis of sterol and nonsterol isoprenoids. We also summarize the characterization of genetically manipulated mice, which established that sterol-accelerated, UBIAD1-modulated ERAD plays a major role in regulation of HMG CoA reductase and cholesterol metabolism in vivo.


Subject(s)
Cholesterol/biosynthesis , Endoplasmic Reticulum-Associated Degradation/physiology , Hydroxymethylglutaryl CoA Reductases/metabolism , Animals , Dimethylallyltranstransferase/metabolism , Endoplasmic Reticulum-Associated Degradation/drug effects , Humans , Hydroxymethylglutaryl CoA Reductases/chemistry , Hydroxymethylglutaryl CoA Reductases/genetics , Mice , Polyisoprenyl Phosphates/metabolism , Protein Processing, Post-Translational , Sterols/metabolism , Terpenes/metabolism , Terpenes/pharmacology , Ubiquitination
9.
Annu Rev Biochem ; 90: 403-429, 2021 06 20.
Article in English | MEDLINE | ID: mdl-33823649

ABSTRACT

Cullin-RING ubiquitin ligases (CRLs) are dynamic modular platforms that regulate myriad biological processes through target-specific ubiquitylation. Our knowledge of this system emerged from the F-box hypothesis, posited a quarter century ago: Numerous interchangeable F-box proteins confer specific substrate recognition for a core CUL1-based RING E3 ubiquitin ligase. This paradigm has been expanded through the evolution of a superfamily of analogous modular CRLs, with five major families and over 200 different substrate-binding receptors in humans. Regulation is achieved by numerous factors organized in circuits that dynamically control CRL activation and substrate ubiquitylation. CRLs also serve as a vast landscape for developing small molecules that reshape interactions and promote targeted ubiquitylation-dependent turnover of proteins of interest. Here, we review molecular principles underlying CRL function, the role of allosteric and conformational mechanisms in controlling substrate timing and ubiquitylation, and how the dynamics of substrate receptor interchange drives the turnover of selected target proteins to promote cellular decision-making.


Subject(s)
Cullin Proteins/chemistry , Cullin Proteins/metabolism , F-Box Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , F-Box Proteins/chemistry , Feedback, Physiological , Host-Pathogen Interactions/physiology , Humans , NEDD8 Protein/metabolism , Plant Growth Regulators/metabolism , Protein Domains , Protein Processing, Post-Translational , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitination
10.
Cell ; 184(13): 3410-3425.e17, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34062120

ABSTRACT

To control viral infection, vertebrates rely on both inducible interferon responses and less well-characterized cell-intrinsic responses composed of "at the ready" antiviral effector proteins. Here, we show that E3 ubiquitin ligase TRIM7 is a cell-intrinsic antiviral effector that restricts multiple human enteroviruses by targeting viral 2BC, a membrane remodeling protein, for ubiquitination and proteasome-dependent degradation. Selective pressure exerted by TRIM7 results in emergence of a TRIM7-resistant coxsackievirus with a single point mutation in the viral 2C ATPase/helicase. In cultured cells, the mutation helps the virus evade TRIM7 but impairs optimal viral replication, and this correlates with a hyperactive and structurally plastic 2C ATPase. Unexpectedly, the TRIM7-resistant virus has a replication advantage in mice and causes lethal pancreatitis. These findings reveal a unique mechanism for targeting enterovirus replication and provide molecular insight into the benefits and trade-offs of viral evolution imposed by a host restriction factor.


Subject(s)
Enterovirus/physiology , Enterovirus/pathogenicity , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Virus Replication/physiology , Adenosine Triphosphatases/metabolism , Animals , Cell Line , Female , Humans , Inflammation/pathology , Mice, Inbred C57BL , Mutation/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Proteolysis , RNA, Viral/metabolism , Ubiquitin/metabolism , Viral Proteins/genetics
11.
Cell ; 184(21): 5357-5374.e22, 2021 10 14.
Article in English | MEDLINE | ID: mdl-34582788

ABSTRACT

Despite remarkable clinical efficacy of immune checkpoint blockade (ICB) in cancer treatment, ICB benefits for triple-negative breast cancer (TNBC) remain limited. Through pooled in vivo CRISPR knockout (KO) screens in syngeneic TNBC mouse models, we found that deletion of the E3 ubiquitin ligase Cop1 in cancer cells decreases secretion of macrophage-associated chemokines, reduces tumor macrophage infiltration, enhances anti-tumor immunity, and strengthens ICB response. Transcriptomics, epigenomics, and proteomics analyses revealed that Cop1 functions through proteasomal degradation of the C/ebpδ protein. The Cop1 substrate Trib2 functions as a scaffold linking Cop1 and C/ebpδ, which leads to polyubiquitination of C/ebpδ. In addition, deletion of the E3 ubiquitin ligase Cop1 in cancer cells stabilizes C/ebpδ to suppress expression of macrophage chemoattractant genes. Our integrated approach implicates Cop1 as a target for improving cancer immunotherapy efficacy in TNBC by regulating chemokine secretion and macrophage infiltration in the tumor microenvironment.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Immunotherapy , Macrophages/enzymology , Neoplasms/immunology , Neoplasms/therapy , Nuclear Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , CCAAT-Enhancer-Binding Protein-delta/metabolism , CRISPR-Associated Protein 9/metabolism , Cell Line, Tumor , Chemokines/metabolism , Chemotaxis , Disease Models, Animal , Gene Library , Humans , Immune Evasion , Mice, Inbred BALB C , Mice, Inbred C57BL , Proteolysis , Substrate Specificity , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/therapy
12.
Cell ; 184(21): 5375-5390.e16, 2021 10 14.
Article in English | MEDLINE | ID: mdl-34562363

ABSTRACT

Although oxidative phosphorylation is best known for producing ATP, it also yields reactive oxygen species (ROS) as invariant byproducts. Depletion of ROS below their physiological levels, a phenomenon known as reductive stress, impedes cellular signaling and has been linked to cancer, diabetes, and cardiomyopathy. Cells alleviate reductive stress by ubiquitylating and degrading the mitochondrial gatekeeper FNIP1, yet it is unknown how the responsible E3 ligase CUL2FEM1B can bind its target based on redox state and how this is adjusted to changing cellular environments. Here, we show that CUL2FEM1B relies on zinc as a molecular glue to selectively recruit reduced FNIP1 during reductive stress. FNIP1 ubiquitylation is gated by pseudosubstrate inhibitors of the BEX family, which prevent premature FNIP1 degradation to protect cells from unwarranted ROS accumulation. FEM1B gain-of-function mutation and BEX deletion elicit similar developmental syndromes, showing that the zinc-dependent reductive stress response must be tightly regulated to maintain cellular and organismal homeostasis.


Subject(s)
Stress, Physiological , Amino Acids/chemistry , Animals , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Cell Line , Female , Humans , Ions , Mice , Mutant Proteins/metabolism , Mutation/genetics , Protein Binding/drug effects , Protein Stability/drug effects , Reactive Oxygen Species/metabolism , Stress, Physiological/drug effects , Structure-Activity Relationship , Substrate Specificity/drug effects , Ubiquitin-Protein Ligase Complexes/chemistry , Ubiquitin-Protein Ligase Complexes/metabolism , Ubiquitination/drug effects , Zinc/pharmacology
13.
Cell ; 184(12): 3178-3191.e18, 2021 06 10.
Article in English | MEDLINE | ID: mdl-34022140

ABSTRACT

Gasdermin B (GSDMB) belongs to a large family of pore-forming cytolysins that execute inflammatory cell death programs. While genetic studies have linked GSDMB polymorphisms to human disease, its function in the immunological response to pathogens remains poorly understood. Here, we report a dynamic host-pathogen conflict between GSDMB and the IpaH7.8 effector protein secreted by enteroinvasive Shigella flexneri. We show that IpaH7.8 ubiquitinates and targets GSDMB for 26S proteasome destruction. This virulence strategy protects Shigella from the bacteriocidic activity of natural killer cells by suppressing granzyme-A-mediated activation of GSDMB. In contrast to the canonical function of most gasdermin family members, GSDMB does not inhibit Shigella by lysing host cells. Rather, it exhibits direct microbiocidal activity through recognition of phospholipids found on Gram-negative bacterial membranes. These findings place GSDMB as a central executioner of intracellular bacterial killing and reveal a mechanism employed by pathogens to counteract this host defense system.


Subject(s)
Biomarkers, Tumor/metabolism , Host-Pathogen Interactions , Killer Cells, Natural/immunology , Neoplasm Proteins/metabolism , Pore Forming Cytotoxic Proteins/metabolism , Shigella flexneri/physiology , Ubiquitination , Animals , Bacterial Proteins/metabolism , Cardiolipins/metabolism , Cell Line , Cell Membrane/metabolism , Female , Granzymes/metabolism , Humans , Lipid A/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Microbial Viability , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Proteolysis , Substrate Specificity
14.
Cell ; 184(20): 5201-5214.e12, 2021 09 30.
Article in English | MEDLINE | ID: mdl-34536345

ABSTRACT

Certain obligate parasites induce complex and substantial phenotypic changes in their hosts in ways that favor their transmission to other trophic levels. However, the mechanisms underlying these changes remain largely unknown. Here we demonstrate how SAP05 protein effectors from insect-vectored plant pathogenic phytoplasmas take control of several plant developmental processes. These effectors simultaneously prolong the host lifespan and induce witches' broom-like proliferations of leaf and sterile shoots, organs colonized by phytoplasmas and vectors. SAP05 acts by mediating the concurrent degradation of SPL and GATA developmental regulators via a process that relies on hijacking the plant ubiquitin receptor RPN10 independent of substrate ubiquitination. RPN10 is highly conserved among eukaryotes, but SAP05 does not bind insect vector RPN10. A two-amino-acid substitution within plant RPN10 generates a functional variant that is resistant to SAP05 activities. Therefore, one effector protein enables obligate parasitic phytoplasmas to induce a plethora of developmental phenotypes in their hosts.


Subject(s)
Arabidopsis/growth & development , Arabidopsis/parasitology , Host-Parasite Interactions/physiology , Parasites/physiology , Proteolysis , Ubiquitins/metabolism , Amino Acid Sequence , Animals , Arabidopsis/genetics , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/metabolism , Genetic Engineering , Humans , Insecta/physiology , Models, Biological , Phenotype , Photoperiod , Phylogeny , Phytoplasma/physiology , Plant Development , Plant Shoots/growth & development , Plants, Genetically Modified , Proteasome Endopeptidase Complex/metabolism , Protein Stability , Reproduction , Nicotiana , Transcription Factors/metabolism , Transcription, Genetic
15.
Cell ; 184(21): 5391-5404.e17, 2021 10 14.
Article in English | MEDLINE | ID: mdl-34597584

ABSTRACT

Plant immunity is activated upon pathogen perception and often affects growth and yield when it is constitutively active. How plants fine-tune immune homeostasis in their natural habitats remains elusive. Here, we discover a conserved immune suppression network in cereals that orchestrates immune homeostasis, centering on a Ca2+-sensor, RESISTANCE OF RICE TO DISEASES1 (ROD1). ROD1 promotes reactive oxygen species (ROS) scavenging by stimulating catalase activity, and its protein stability is regulated by ubiquitination. ROD1 disruption confers resistance to multiple pathogens, whereas a natural ROD1 allele prevalent in indica rice with agroecology-specific distribution enhances resistance without yield penalty. The fungal effector AvrPiz-t structurally mimics ROD1 and activates the same ROS-scavenging cascade to suppress host immunity and promote virulence. We thus reveal a molecular framework adopted by both host and pathogen that integrates Ca2+ sensing and ROS homeostasis to suppress plant immunity, suggesting a principle for breeding disease-resistant, high-yield crops.


Subject(s)
Calcium/metabolism , Free Radical Scavengers/metabolism , Fungal Proteins/metabolism , Oryza/immunology , Plant Immunity , Plant Proteins/metabolism , Reactive Oxygen Species/metabolism , CRISPR-Cas Systems/genetics , Cell Membrane/metabolism , Disease Resistance/genetics , Models, Biological , Oryza/genetics , Plant Diseases/immunology , Plant Proteins/genetics , Protein Binding , Protein Stability , Reproduction , Species Specificity , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Zea mays/immunology
16.
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
17.
Annu Rev Biochem ; 89: 417-442, 2020 06 20.
Article in English | MEDLINE | ID: mdl-32569528

ABSTRACT

Stalled protein synthesis produces defective nascent chains that can harm cells. In response, cells degrade these nascent chains via a process called ribosome-associated quality control (RQC). Here, we review the irregularities in the translation process that cause ribosomes to stall as well as how cells use RQC to detect stalled ribosomes, ubiquitylate their tethered nascent chains, and deliver the ubiquitylated nascent chains to the proteasome. We additionally summarize how cells respond to RQC failure.


Subject(s)
Escherichia coli/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Biosynthesis , Protein Processing, Post-Translational , Ribosomes/genetics , Escherichia coli/metabolism , Humans , Models, Molecular , Poly A/chemistry , Poly A/genetics , Poly A/metabolism , Proteasome Endopeptidase Complex/genetics , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Proteolysis , RNA Splicing , RNA Stability , Ribosomes/metabolism , Ribosomes/ultrastructure , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
18.
Cell ; 183(6): 1714-1731.e10, 2020 12 10.
Article in English | MEDLINE | ID: mdl-33275901

ABSTRACT

Targeted protein degradation (TPD) refers to the use of small molecules to induce ubiquitin-dependent degradation of proteins. TPD is of interest in drug development, as it can address previously inaccessible targets. However, degrader discovery and optimization remains an inefficient process due to a lack of understanding of the relative importance of the key molecular events required to induce target degradation. Here, we use chemo-proteomics to annotate the degradable kinome. Our expansive dataset provides chemical leads for ∼200 kinases and demonstrates that the current practice of starting from the highest potency binder is an ineffective method for discovering active compounds. We develop multitargeted degraders to answer fundamental questions about the ubiquitin proteasome system, uncovering that kinase degradation is p97 dependent. This work will not only fuel kinase degrader discovery, but also provides a blueprint for evaluating targeted degradation across entire gene families to accelerate understanding of TPD beyond the kinome.


Subject(s)
Protein Kinases/metabolism , Proteolysis , Proteome/metabolism , Adult , Cell Line , Databases, Protein , Female , Humans , Male , Middle Aged , Proteasome Endopeptidase Complex/metabolism , Protein Kinases/genetics , Proteomics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ubiquitin-Protein Ligases/metabolism , Young Adult
19.
Cell ; 180(6): 1245-1261.e21, 2020 03 19.
Article in English | MEDLINE | ID: mdl-32142654

ABSTRACT

In response to transcription-blocking DNA damage, cells orchestrate a multi-pronged reaction, involving transcription-coupled DNA repair, degradation of RNA polymerase II (RNAPII), and genome-wide transcription shutdown. Here, we provide insight into how these responses are connected by the finding that ubiquitylation of RNAPII itself, at a single lysine (RPB1 K1268), is the focal point for DNA-damage-response coordination. K1268 ubiquitylation affects DNA repair and signals RNAPII degradation, essential for surviving genotoxic insult. RNAPII degradation results in a shutdown of transcriptional initiation, in the absence of which cells display dramatic transcriptome alterations. Additionally, regulation of RNAPII stability is central to transcription recovery-persistent RNAPII depletion underlies the failure of this process in Cockayne syndrome B cells. These data expose regulation of global RNAPII levels as integral to the cellular DNA-damage response and open the intriguing possibility that RNAPII pool size generally affects cell-specific transcription programs in genome instability disorders and even normal cells.


Subject(s)
DNA Damage , RNA Polymerase II/metabolism , DNA Repair , HEK293 Cells , Humans , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Transcription, Genetic , Ubiquitination , Ultraviolet Rays
20.
Cell ; 183(1): 46-61.e21, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32941802

ABSTRACT

Metazoan organisms rely on conserved stress response pathways to alleviate adverse conditions and preserve cellular integrity. Stress responses are particularly important in stem cells that provide lifetime support for tissue formation and repair, but how these protective systems are integrated into developmental programs is poorly understood. Here we used myoblast differentiation to identify the E3 ligase CUL2FEM1B and its substrate FNIP1 as core components of the reductive stress response. Reductive stress, as caused by prolonged antioxidant signaling or mitochondrial inactivity, reverts the oxidation of invariant Cys residues in FNIP1 and allows CUL2FEM1B to recognize its target. The ensuing proteasomal degradation of FNIP1 restores mitochondrial activity to preserve redox homeostasis and stem cell integrity. The reductive stress response is therefore built around a ubiquitin-dependent rheostat that tunes mitochondrial activity to redox needs and implicates metabolic control in coordination of stress and developmental signaling.


Subject(s)
Carrier Proteins/metabolism , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Antioxidants/metabolism , Carrier Proteins/genetics , Cell Differentiation , HEK293 Cells , Homeostasis , Humans , Kelch-Like ECH-Associated Protein 1/metabolism , Mitochondria , Muscle Development/physiology , Myoblasts/metabolism , NF-E2-Related Factor 2/metabolism , Signal Transduction , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL