Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28.834
Filter
Add more filters

Publication year range
1.
Cell ; 187(21): 5967-5980.e17, 2024 Oct 17.
Article in English | MEDLINE | ID: mdl-39276772

ABSTRACT

Protein aggregation causes a wide range of neurodegenerative diseases. Targeting and removing aggregates, but not the functional protein, is a considerable therapeutic challenge. Here, we describe a therapeutic strategy called "RING-Bait," which employs an aggregating protein sequence combined with an E3 ubiquitin ligase. RING-Bait is recruited into aggregates, whereupon clustering dimerizes the RING domain and activates its E3 function, resulting in the degradation of the aggregate complex. We exemplify this concept by demonstrating the specific degradation of tau aggregates while sparing soluble tau. Unlike immunotherapy, RING-Bait is effective against both seeded and cell-autonomous aggregation. RING-Bait removed tau aggregates seeded from Alzheimer's disease (AD) and progressive supranuclear palsy (PSP) brain extracts and was also effective in primary neurons. We used a brain-penetrant adeno-associated virus (AAV) to treat P301S tau transgenic mice, reducing tau pathology and improving motor function. A RING-Bait strategy could be applied to other neurodegenerative proteinopathies by replacing the Bait sequence to match the target aggregate.


Subject(s)
Alzheimer Disease , Mice, Transgenic , Neurons , tau Proteins , tau Proteins/metabolism , tau Proteins/chemistry , Animals , Humans , Mice , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/therapy , Neurons/metabolism , Brain/metabolism , Brain/pathology , Supranuclear Palsy, Progressive/metabolism , Protein Aggregation, Pathological/metabolism , Ubiquitin-Protein Ligases/metabolism , Dependovirus/metabolism , Dependovirus/genetics , Female , HEK293 Cells , Male , Protein Aggregates , Motor Activity
2.
Cell ; 186(9): 1817-1818, 2023 04 27.
Article in English | MEDLINE | ID: mdl-37116466

ABSTRACT

Proper regulation of protein degradation is essential for cell physiology. In the current issue of Cell, Baek et al. elucidated how a large class of ubiquitin ligase, known as CRL, is assembled and disassembled through a key regulator, CAND1.


Subject(s)
Transcription Factors , Ubiquitin-Protein Ligases , Cullin Proteins/metabolism , Proteolysis , Transcription Factors/metabolism , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism
3.
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
4.
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
5.
Cell ; 186(19): 4172-4188.e18, 2023 09 14.
Article in English | MEDLINE | ID: mdl-37633267

ABSTRACT

Selective clearance of organelles, including endoplasmic reticulum (ER) and mitochondria, by autophagy plays an important role in cell health. Here, we describe a developmentally programmed selective ER clearance by autophagy. We show that Parkinson's disease-associated PINK1, as well as Atl, Rtnl1, and Trp1 receptors, regulate ER clearance by autophagy. The E3 ubiquitin ligase Parkin functions downstream of PINK1 and is required for mitochondrial clearance while having the opposite function in ER clearance. By contrast, Keap1 and the E3 ubiquitin ligase Cullin3 function downstream of PINK1 to regulate ER clearance by influencing Rtnl1 and Atl. PINK1 regulates a change in Keap1 localization and Keap1-dependent ubiquitylation of the ER-phagy receptor Rtnl1 to facilitate ER clearance. Thus, PINK1 regulates the selective clearance of ER and mitochondria by influencing the balance of Keap1- and Parkin-dependent ubiquitylation of substrates that determine which organelle is removed by autophagy.


Subject(s)
Endoplasmic Reticulum , NF-E2-Related Factor 2 , Endoplasmic Reticulum/metabolism , Kelch-Like ECH-Associated Protein 1 , Protein Kinases , Ubiquitin-Protein Ligases , Drosophila melanogaster , Animals
6.
Cell ; 186(18): 3903-3920.e21, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37557169

ABSTRACT

Immune-checkpoint blockade has revolutionized cancer treatment, but some cancers, such as acute myeloid leukemia (AML), do not respond or develop resistance. A potential mode of resistance is immune evasion of T cell immunity involving aberrant major histocompatibility complex class I (MHC-I) antigen presentation (AP). To map such mechanisms of resistance, we identified key MHC-I regulators using specific peptide-MHC-I-guided CRISPR-Cas9 screens in AML. The top-ranked negative regulators were surface protein sushi domain containing 6 (SUSD6), transmembrane protein 127 (TMEM127), and the E3 ubiquitin ligase WWP2. SUSD6 is abundantly expressed in AML and multiple solid cancers, and its ablation enhanced MHC-I AP and reduced tumor growth in a CD8+ T cell-dependent manner. Mechanistically, SUSD6 forms a trimolecular complex with TMEM127 and MHC-I, which recruits WWP2 for MHC-I ubiquitination and lysosomal degradation. Together with the SUSD6/TMEM127/WWP2 gene signature, which negatively correlates with cancer survival, our findings define a membrane-associated MHC-I inhibitory axis as a potential therapeutic target for both leukemia and solid cancers.


Subject(s)
Histocompatibility Antigens Class I , Neoplasms , Tumor Escape , Humans , Antigen Presentation , CD8-Positive T-Lymphocytes , Histocompatibility Antigens Class I/metabolism , HLA Antigens , Neoplasms/immunology , Ubiquitin-Protein Ligases/genetics
7.
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
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 ; 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
10.
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
11.
Nat Immunol ; 25(5): 764-777, 2024 May.
Article in English | MEDLINE | ID: mdl-38609546

ABSTRACT

The linear ubiquitin assembly complex (LUBAC) consists of HOIP, HOIL-1 and SHARPIN and is essential for proper immune responses. Individuals with HOIP and HOIL-1 deficiencies present with severe immunodeficiency, autoinflammation and glycogen storage disease. In mice, the loss of Sharpin leads to severe dermatitis due to excessive keratinocyte cell death. Here, we report two individuals with SHARPIN deficiency who manifest autoinflammatory symptoms but unexpectedly no dermatological problems. Fibroblasts and B cells from these individuals showed attenuated canonical NF-κB responses and a propensity for cell death mediated by TNF superfamily members. Both SHARPIN-deficient and HOIP-deficient individuals showed a substantial reduction of secondary lymphoid germinal center B cell development. Treatment of one SHARPIN-deficient individual with anti-TNF therapies led to complete clinical and transcriptomic resolution of autoinflammation. These findings underscore the critical function of the LUBAC as a gatekeeper for cell death-mediated immune dysregulation in humans.


Subject(s)
Immunologic Deficiency Syndromes , Nerve Tissue Proteins , Ubiquitins , Humans , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Female , Male , NF-kappa B/metabolism , Ubiquitin-Protein Ligases/genetics , Inflammation/immunology , Inflammation/genetics , B-Lymphocytes/immunology , Loss of Function Mutation , Fibroblasts/metabolism , Fibroblasts/immunology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Animals , Mice , Alleles
12.
Nat Rev Mol Cell Biol ; 25(9): 740-757, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38684868

ABSTRACT

Targeted protein degradation refers to the use of small molecules to induce the selective degradation of proteins. In its most common form, this degradation is achieved through ligand-mediated neo-interactions between ubiquitin E3 ligases - the principal waste disposal machines of a cell - and the protein targets of interest, resulting in ubiquitylation and subsequent proteasomal degradation. Notable advances have been made in biological and mechanistic understanding of serendipitously discovered degraders. This improved understanding and novel chemistry has not only provided clinical proof of concept for targeted protein degradation but has also led to rapid growth of the field, with dozens of investigational drugs in active clinical trials. Two distinct classes of protein degradation therapeutics are being widely explored: bifunctional PROTACs and molecular glue degraders, both of which have their unique advantages and challenges. Here, we review the current landscape of targeted protein degradation approaches and how they have parallels in biological processes. We also outline the ongoing clinical exploration of novel degraders and provide some perspectives on the directions the field might take.


Subject(s)
Proteasome Endopeptidase Complex , Proteolysis , Ubiquitin-Protein Ligases , Ubiquitination , Humans , Ubiquitin-Protein Ligases/metabolism , Proteasome Endopeptidase Complex/metabolism , Animals , Proteins/metabolism
13.
Cell ; 184(9): 2412-2429.e16, 2021 04 29.
Article in English | MEDLINE | ID: mdl-33852913

ABSTRACT

Cellular versatility depends on accurate trafficking of diverse proteins to their organellar destinations. For the secretory pathway (followed by approximately 30% of all proteins), the physical nature of the vessel conducting the first portage (endoplasmic reticulum [ER] to Golgi apparatus) is unclear. We provide a dynamic 3D view of early secretory compartments in mammalian cells with isotropic resolution and precise protein localization using whole-cell, focused ion beam scanning electron microscopy with cryo-structured illumination microscopy and live-cell synchronized cargo release approaches. Rather than vesicles alone, the ER spawns an elaborate, interwoven tubular network of contiguous lipid bilayers (ER exit site) for protein export. This receptacle is capable of extending microns along microtubules while still connected to the ER by a thin neck. COPII localizes to this neck region and dynamically regulates cargo entry from the ER, while COPI acts more distally, escorting the detached, accelerating tubular entity on its way to joining the Golgi apparatus through microtubule-directed movement.


Subject(s)
COP-Coated Vesicles/metabolism , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Microtubules/metabolism , Ubiquitin-Protein Ligases/metabolism , Biological Transport, Active , HeLa Cells , Humans , Protein Transport
14.
Cell ; 184(4): 1081-1097.e19, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33606978

ABSTRACT

Mutations in DNA damage response (DDR) genes endanger genome integrity and predispose to cancer and genetic disorders. Here, using CRISPR-dependent cytosine base editing screens, we identify > 2,000 sgRNAs that generate nucleotide variants in 86 DDR genes, resulting in altered cellular fitness upon DNA damage. Among those variants, we discover loss- and gain-of-function mutants in the Tudor domain of the DDR regulator 53BP1 that define a non-canonical surface required for binding the deubiquitinase USP28. Moreover, we characterize variants of the TRAIP ubiquitin ligase that define a domain, whose loss renders cells resistant to topoisomerase I inhibition. Finally, we identify mutations in the ATM kinase with opposing genome stability phenotypes and loss-of-function mutations in the CHK2 kinase previously categorized as variants of uncertain significance for breast cancer. We anticipate that this resource will enable the discovery of additional DDR gene functions and expedite studies of DDR variants in human disease.


Subject(s)
DNA Damage , Gene Editing , Genetic Testing , Amino Acid Sequence , Ataxia Telangiectasia Mutated Proteins/metabolism , Base Sequence , CRISPR-Cas Systems/genetics , Camptothecin/pharmacology , Cell Line , DNA Damage/genetics , DNA Repair/genetics , Female , Humans , Mutation/genetics , Phenotype , Protein Binding , Protein Domains , RNA, Guide, Kinetoplastida/genetics , Topoisomerase Inhibitors/pharmacology , Tumor Suppressor p53-Binding Protein 1/chemistry , Tumor Suppressor p53-Binding Protein 1/genetics , Ubiquitin Thiolesterase/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
15.
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
16.
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
17.
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
18.
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
19.
Nat Rev Mol Cell Biol ; 24(4): 273-287, 2023 04.
Article in English | MEDLINE | ID: mdl-36284179

ABSTRACT

Our understanding of the ubiquitin code has greatly evolved from conventional E1, E2 and E3 enzymes that modify Lys residues on specific substrates with a single type of ubiquitin chain to more complex processes that regulate and mediate ubiquitylation. In this Review, we discuss recently discovered endogenous mechanisms and unprecedented pathways by which pathogens rewrite the ubiquitin code to promote infection. These processes include unconventional ubiquitin modifications involving ester linkages with proteins, lipids and sugars, or ubiquitylation through a phosphoribosyl bridge involving Arg42 of ubiquitin. We also introduce the enzymatic pathways that write and reverse these modifications, such as the papain-like proteases of severe acute respiratory syndrome coronavirus (SARS-CoV) and SARS-CoV-2. Furthermore, structural studies have revealed that the ultimate functions of ubiquitin are mediated not simply by straightforward recognition by ubiquitin-binding domains. Instead, elaborate multivalent interactions between ubiquitylated targets or ubiquitin chains and their readers (for example, the proteasome, the MLL1 complex or DOT1L) can elicit conformational changes that regulate protein degradation or transcription. The newly discovered mechanisms provide opportunities for innovative therapeutic interventions for diseases such as cancer and infectious diseases.


Subject(s)
COVID-19 , Ubiquitin , Humans , Ubiquitin/metabolism , SARS-CoV-2 , Ubiquitination , Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Proteasome Endopeptidase Complex/metabolism
20.
Cell ; 181(1): 102-114, 2020 04 02.
Article in English | MEDLINE | ID: mdl-31955850

ABSTRACT

New biological tools provide new techniques to probe fundamental biological processes. Here we describe the burgeoning field of proteolysis-targeting chimeras (PROTACs), which are capable of modulating protein concentrations at a post-translational level by co-opting the ubiquitin-proteasome system. We describe the PROTAC technology and its application to drug discovery and provide examples where PROTACs have enabled novel biological insights. Furthermore, we provide a workflow for PROTAC development and use and discuss the benefits and issues associated with PROTACs. Finally, we compare PROTAC-mediated protein-level modulation with other technologies, such as RNAi and genome editing.


Subject(s)
Drug Discovery , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Ubiquitin-Protein Ligases/metabolism , Humans
SELECTION OF CITATIONS
SEARCH DETAIL