Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
1.
Annu Rev Biochem ; 87: 697-724, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29652515

ABSTRACT

As the endpoint for the ubiquitin-proteasome system, the 26S proteasome is the principal proteolytic machine responsible for regulated protein degradation in eukaryotic cells. The proteasome's cellular functions range from general protein homeostasis and stress response to the control of vital processes such as cell division and signal transduction. To reliably process all the proteins presented to it in the complex cellular environment, the proteasome must combine high promiscuity with exceptional substrate selectivity. Recent structural and biochemical studies have shed new light on the many steps involved in proteasomal substrate processing, including recognition, deubiquitination, and ATP-driven translocation and unfolding. In addition, these studies revealed a complex conformational landscape that ensures proper substrate selection before the proteasome commits to processive degradation. These advances in our understanding of the proteasome's intricate machinery set the stage for future studies on how the proteasome functions as a major regulator of the eukaryotic proteome.


Subject(s)
Proteasome Endopeptidase Complex/chemistry , Proteasome Endopeptidase Complex/metabolism , ATPases Associated with Diverse Cellular Activities/chemistry , ATPases Associated with Diverse Cellular Activities/metabolism , Deubiquitinating Enzymes/chemistry , Deubiquitinating Enzymes/metabolism , Humans , Models, Biological , Models, Molecular , Molecular Motor Proteins/chemistry , Molecular Motor Proteins/metabolism , Protein Conformation , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Substrate Specificity , Ubiquitin/chemistry , Ubiquitin/metabolism
2.
Annu Rev Biochem ; 86: 159-192, 2017 06 20.
Article in English | MEDLINE | ID: mdl-28498721

ABSTRACT

Protein ubiquitination is one of the most powerful posttranslational modifications of proteins, as it regulates a plethora of cellular processes in distinct manners. Simple monoubiquitination events coexist with more complex forms of polyubiquitination, the latter featuring many different chain architectures. Ubiquitin can be subjected to further posttranslational modifications (e.g., phosphorylation and acetylation) and can also be part of mixed polymers with ubiquitin-like modifiers such as SUMO (small ubiquitin-related modifier) or NEDD8 (neural precursor cell expressed, developmentally downregulated 8). Together, cellular ubiquitination events form a sophisticated and versatile ubiquitin code. Deubiquitinases (DUBs) reverse ubiquitin signals with equally high sophistication. In this review, we conceptualize the many layers of specificity that DUBs encompass to control the ubiquitin code and discuss examples in which DUB specificity has been understood at the molecular level. We further discuss the many mechanisms of DUB regulation with a focus on those that modulate catalytic activity. Our review provides a framework to tackle lingering questions in DUB biology.


Subject(s)
Deubiquitinating Enzymes/metabolism , Eukaryotic Cells/metabolism , Protein Processing, Post-Translational , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Ubiquitins/metabolism , Acetylation , Allosteric Regulation , Deubiquitinating Enzymes/chemistry , Deubiquitinating Enzymes/genetics , Humans , Models, Molecular , NEDD8 Protein , Phosphorylation , Protein Binding , Protein Conformation , Proteolysis , Substrate Specificity , Sumoylation , Ubiquitin/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination , Ubiquitins/genetics
3.
Mol Cell ; 74(3): 421-435.e10, 2019 05 02.
Article in English | MEDLINE | ID: mdl-30926243

ABSTRACT

Deubiquitinases have emerged as promising drug targets for cancer therapy. The two DUBs USP25 and USP28 share high similarity but vary in their cellular functions. USP28 is known for its tumor-promoting role, whereas USP25 is a regulator of the innate immune system and, recently, a role in tumorigenesis was proposed. We solved the structures of the catalytic domains of both proteins and established substantial differences in their activities. While USP28 is a constitutively active dimer, USP25 presents an auto-inhibited tetramer. Our data indicate that the activation of USP25 is not achieved through substrate or ubiquitin binding. USP25 cancer-associated mutations lead to activation in vitro and in vivo, thereby providing a functional link between auto-inhibition and the cancer-promoting role of the enzyme. Our work led to the identification of significant differences between USP25 and USP28 and provided the molecular basis for the development of new and highly specific anti-cancer drugs.


Subject(s)
Carcinogenesis/genetics , Neoplasms/genetics , Ubiquitin Thiolesterase/genetics , Amino Acid Sequence/genetics , Catalytic Domain/genetics , Deubiquitinating Enzymes/chemistry , Deubiquitinating Enzymes/genetics , Humans , Mutation/genetics , Neoplasms/drug therapy , Protein Binding/genetics , Protein Conformation , Protein Multimerization/genetics , Ubiquitin/genetics , Ubiquitin Thiolesterase/chemistry
4.
Mol Cell ; 74(3): 436-451.e7, 2019 05 02.
Article in English | MEDLINE | ID: mdl-30926242

ABSTRACT

The evolutionarily related deubiquitinating enzymes (DUBs) USP25 and USP28 comprise an identical overall domain architecture but are functionally non-redundant: USP28 stabilizes c-MYC and other nuclear proteins, and USP25 regulates inflammatory TRAF signaling. We here compare molecular features of USP25 and USP28. Active enzymes form distinctively shaped dimers, with a dimerizing insertion spatially separating independently active catalytic domains. In USP25, but not USP28, two dimers can form an autoinhibited tetramer, where a USP25-specific, conserved insertion sequence blocks ubiquitin binding. In full-length enzymes, a C-terminal domain with a previously unknown fold has no impact on oligomerization, but N-terminal regions affect the dimer-tetramer equilibrium in vitro. We confirm oligomeric states of USP25 and USP28 in cells and show that modulating oligomerization affects substrate stabilization in accordance with in vitro activity data. Our work highlights how regions outside of the catalytic domain enable a conceptually intriguing interplay of DUB oligomerization and activity.


Subject(s)
Inflammation/genetics , Protein Conformation , Ubiquitin Thiolesterase/genetics , Amino Acid Sequence/genetics , Catalytic Domain/genetics , Deubiquitinating Enzymes/chemistry , Deubiquitinating Enzymes/genetics , Humans , Inflammation/pathology , Mutation/genetics , Protein Binding/genetics , Protein Domains/genetics , Protein Multimerization/genetics , Proto-Oncogene Proteins c-myb/chemistry , Proto-Oncogene Proteins c-myb/genetics , Signal Transduction/genetics , Substrate Specificity , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/genetics , Ubiquitin/genetics , Ubiquitin Thiolesterase/chemistry
5.
Nature ; 587(7835): 657-662, 2020 11.
Article in English | MEDLINE | ID: mdl-32726803

ABSTRACT

The papain-like protease PLpro is an essential coronavirus enzyme that is required for processing viral polyproteins to generate a functional replicase complex and enable viral spread1,2. PLpro is also implicated in cleaving proteinaceous post-translational modifications on host proteins as an evasion mechanism against host antiviral immune responses3-5. Here we perform biochemical, structural and functional characterization of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) PLpro (SCoV2-PLpro) and outline differences with SARS-CoV PLpro (SCoV-PLpro) in regulation of host interferon and NF-κB pathways. SCoV2-PLpro and SCoV-PLpro share 83% sequence identity but exhibit different host substrate preferences; SCoV2-PLpro preferentially cleaves the ubiquitin-like interferon-stimulated gene 15 protein (ISG15), whereas SCoV-PLpro predominantly targets ubiquitin chains. The crystal structure of SCoV2-PLpro in complex with ISG15 reveals distinctive interactions with the amino-terminal ubiquitin-like domain of ISG15, highlighting the high affinity and specificity of these interactions. Furthermore, upon infection, SCoV2-PLpro contributes to the cleavage of ISG15 from interferon responsive factor 3 (IRF3) and attenuates type I interferon responses. Notably, inhibition of SCoV2-PLpro with GRL-0617 impairs the virus-induced cytopathogenic effect, maintains the antiviral interferon pathway and reduces viral replication in infected cells. These results highlight a potential dual therapeutic strategy in which targeting of SCoV2-PLpro can suppress SARS-CoV-2 infection and promote antiviral immunity.


Subject(s)
COVID-19/immunology , COVID-19/virology , Coronavirus Papain-Like Proteases/chemistry , Coronavirus Papain-Like Proteases/metabolism , Immunity, Innate , SARS-CoV-2/enzymology , SARS-CoV-2/immunology , Animals , Coronavirus Papain-Like Proteases/antagonists & inhibitors , Cytokines/chemistry , Cytokines/metabolism , Deubiquitinating Enzymes/antagonists & inhibitors , Deubiquitinating Enzymes/chemistry , Deubiquitinating Enzymes/metabolism , Humans , Interferon Regulatory Factor-3/metabolism , Interferons/immunology , Interferons/metabolism , Mice , Models, Molecular , Molecular Dynamics Simulation , NF-kappa B/immunology , NF-kappa B/metabolism , Protein Binding , SARS-CoV-2/drug effects , SARS-CoV-2/physiology , Ubiquitination , Ubiquitins/chemistry , Ubiquitins/metabolism , COVID-19 Drug Treatment
6.
Mol Cell ; 70(1): 150-164.e6, 2018 04 05.
Article in English | MEDLINE | ID: mdl-29576527

ABSTRACT

Deubiquitinating enzymes (DUBs) are important regulators of ubiquitin signaling. Here, we report the discovery of deubiquitinating activity in ZUFSP/C6orf113. High-resolution crystal structures of ZUFSP in complex with ubiquitin reveal several distinctive features of ubiquitin recognition and catalysis. Our analyses reveal that ZUFSP is a novel DUB with no homology to any known DUBs, leading us to classify ZUFSP as the seventh DUB family. Intriguingly, the minimal catalytic domain does not cleave polyubiquitin. We identify two ubiquitin binding domains in ZUFSP: a ZHA (ZUFSP helical arm) that binds to the distal ubiquitin and an atypical UBZ domain in ZUFSP that binds to polyubiquitin. Importantly, both domains are essential for ZUFSP to selectively cleave K63-linked polyubiquitin. We show that ZUFSP localizes to DNA lesions, where it plays an important role in genome stability pathways, functioning to prevent spontaneous DNA damage and also promote cellular survival in response to exogenous DNA damage.


Subject(s)
Cell Nucleus/enzymology , DNA Damage , Deubiquitinating Enzymes/metabolism , Genomic Instability , Polyubiquitin/metabolism , Binding Sites , Cell Survival , Deubiquitinating Enzymes/chemistry , Deubiquitinating Enzymes/genetics , HEK293 Cells , HeLa Cells , Humans , Jurkat Cells , Lysine , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Structure-Activity Relationship , Substrate Specificity , Ubiquitination
7.
J Biol Chem ; 300(6): 107315, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38663827

ABSTRACT

Lewy bodies (LB) are aberrant protein accumulations observed in the brain cells of individuals affected by Parkinson's disease (PD). A comprehensive analysis of LB proteome identified over a hundred proteins, many co-enriched with α-synuclein, a major constituent of LB. Within this context, OTUB1, a deubiquitinase detected in LB, exhibits amyloidogenic properties, yet the mechanisms underlying its aggregation remain elusive. In this study, we identify two critical sites in OTUB1-namely, positions 133 and 173-that significantly impact its amyloid aggregation. Substituting alanine at position 133 and lysine at position 173 enhances both thermodynamic and kinetic stability, effectively preventing amyloid aggregation. Remarkably, lysine at position 173 demonstrates the highest stability without compromising enzymatic activity. The increased stability and inhibition of amyloid aggregation are attributed mainly to the changes in the specific microenvironment at the hotspot. In our exploration of the in-vivo co-occurrence of α-synuclein and OTUB1 in LB, we observed a synergistic modulation of each other's aggregation. Collectively, our study unveils the molecular determinants influencing OTUB1 aggregation, shedding light on the role of specific residues in modulating aggregation kinetics and structural transition. These findings contribute valuable insights into the complex interplay of amino acid properties and protein aggregation, with potential implications for understanding broader aspects of protein folding and aggregation phenomena.


Subject(s)
alpha-Synuclein , Humans , alpha-Synuclein/metabolism , alpha-Synuclein/chemistry , alpha-Synuclein/genetics , Cysteine Endopeptidases/metabolism , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/chemistry , Deubiquitinating Enzymes/metabolism , Deubiquitinating Enzymes/chemistry , Protein Aggregates , Lewy Bodies/metabolism , Parkinson Disease/metabolism , Parkinson Disease/genetics , Amyloid/metabolism , Amyloid/chemistry , Protein Stability , Enzyme Stability , Kinetics
8.
Nature ; 570(7760): 194-199, 2019 06.
Article in English | MEDLINE | ID: mdl-31142841

ABSTRACT

Serine hydroxymethyltransferase 2 (SHMT2) regulates one-carbon transfer reactions that are essential for amino acid and nucleotide metabolism, and uses pyridoxal-5'-phosphate (PLP) as a cofactor. Apo SHMT2 exists as a dimer with unknown functions, whereas PLP binding stabilizes the active tetrameric state. SHMT2 also promotes inflammatory cytokine signalling by interacting with the deubiquitylating BRCC36 isopeptidase complex (BRISC), although it is unclear whether this function relates to metabolism. Here we present the cryo-electron microscopy structure of the human BRISC-SHMT2 complex at a resolution of 3.8 Å. BRISC is a U-shaped dimer of four subunits, and SHMT2 sterically blocks the BRCC36 active site and inhibits deubiquitylase activity. Only the inactive SHMT2 dimer-and not the active PLP-bound tetramer-binds and inhibits BRISC. Mutations in BRISC that disrupt SHMT2 binding impair type I interferon signalling in response to inflammatory stimuli. Intracellular levels of PLP regulate the interaction between BRISC and SHMT2, as well as inflammatory cytokine responses. These data reveal a mechanism in which metabolites regulate deubiquitylase activity and inflammatory signalling.


Subject(s)
Deubiquitinating Enzymes/metabolism , Glycine Hydroxymethyltransferase/metabolism , Interferon Type I/immunology , Multienzyme Complexes/immunology , Multienzyme Complexes/metabolism , Signal Transduction/immunology , Cryoelectron Microscopy , Deubiquitinating Enzymes/antagonists & inhibitors , Deubiquitinating Enzymes/chemistry , Deubiquitinating Enzymes/ultrastructure , Glycine Hydroxymethyltransferase/ultrastructure , HEK293 Cells , Humans , Inflammation/immunology , Models, Molecular , Multienzyme Complexes/chemistry , Multienzyme Complexes/genetics , Mutation , Protein Binding , Protein Multimerization , Protein Structure, Quaternary , Pyridoxal Phosphate/metabolism
9.
EMBO J ; 39(15): e105127, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32567101

ABSTRACT

Manipulation of host ubiquitin signaling is becoming an increasingly apparent evolutionary strategy among bacterial and viral pathogens. By removing host ubiquitin signals, for example, invading pathogens can inactivate immune response pathways and evade detection. The ovarian tumor (OTU) family of deubiquitinases regulates diverse ubiquitin signals in humans. Viral pathogens have also extensively co-opted the OTU fold to subvert host signaling, but the extent to which bacteria utilize the OTU fold was unknown. We have predicted and validated a set of OTU deubiquitinases encoded by several classes of pathogenic bacteria. Biochemical assays highlight the ubiquitin and polyubiquitin linkage specificities of these bacterial deubiquitinases. By determining the ubiquitin-bound structures of two examples, we demonstrate the novel strategies that have evolved to both thread an OTU fold and recognize a ubiquitin substrate. With these new examples, we perform the first cross-kingdom structural analysis of the OTU fold that highlights commonalities among distantly related OTU deubiquitinases.


Subject(s)
Bacterial Proteins , Deubiquitinating Enzymes , Legionella/enzymology , Protein Folding , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Deubiquitinating Enzymes/chemistry , Deubiquitinating Enzymes/genetics , Deubiquitinating Enzymes/metabolism , Legionella/genetics , Polyubiquitin/chemistry , Polyubiquitin/genetics , Polyubiquitin/metabolism , Substrate Specificity
10.
Mol Cell ; 62(4): 572-85, 2016 05 19.
Article in English | MEDLINE | ID: mdl-27203180

ABSTRACT

Deubiquitinating enzymes (DUBs) recognize and cleave linkage-specific polyubiquitin (polyUb) chains, but mechanisms underlying specificity remain elusive in many cases. The severe acute respiratory syndrome (SARS) coronavirus papain-like protease (PLpro) is a DUB that cleaves ISG15, a two-domain Ub-like protein, and Lys48-linked polyUb chains, releasing diUb(Lys48) products. To elucidate this specificity, we report the 2.85 Å crystal structure of SARS PLpro bound to a diUb(Lys48) activity-based probe. SARS PLpro binds diUb(Lys48) in an extended conformation via two contact sites, S1 and S2, which are proximal and distal to the active site, respectively. We show that specificity for polyUb(Lys48) chains is predicated on contacts in the S2 site and enhanced by an S1-S1' preference for a Lys48 linkage across the active site. In contrast, ISG15 specificity is dominated by contacts in the S1 site. Determinants revealed for polyUb(Lys48) specificity should prove useful in understanding PLpro deubiquitinating activities in coronavirus infections.


Subject(s)
Cysteine Endopeptidases/metabolism , Cytokines/metabolism , Deubiquitinating Enzymes/metabolism , Polyubiquitin/metabolism , Severe acute respiratory syndrome-related coronavirus/enzymology , Ubiquitins/metabolism , Viral Proteins/metabolism , Binding Sites , Coronavirus 3C Proteases , Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/genetics , Cytokines/chemistry , Deubiquitinating Enzymes/chemistry , HeLa Cells , Humans , Lysine , Models, Molecular , Mutation , Polyubiquitin/chemistry , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Severe acute respiratory syndrome-related coronavirus/genetics , Structure-Activity Relationship , Ubiquitination , Ubiquitins/chemistry , Viral Proteins/chemistry , Viral Proteins/genetics
11.
Mol Cell ; 63(1): 146-55, 2016 07 07.
Article in English | MEDLINE | ID: mdl-27292798

ABSTRACT

Deubiquitinating enzymes (DUBs) remove ubiquitin (Ub) from Ub-conjugated substrates to regulate the functional outcome of ubiquitylation. Here we report the discovery of a new family of DUBs, which we have named MINDY (motif interacting with Ub-containing novel DUB family). Found in all eukaryotes, MINDY-family DUBs are highly selective at cleaving K48-linked polyUb, a signal that targets proteins for degradation. We identify the catalytic activity to be encoded within a previously unannotated domain, the crystal structure of which reveals a distinct protein fold with no homology to any of the known DUBs. The crystal structure of MINDY-1 (also known as FAM63A) in complex with propargylated Ub reveals conformational changes that realign the active site for catalysis. MINDY-1 prefers cleaving long polyUb chains and works by trimming chains from the distal end. Collectively, our results reveal a new family of DUBs that may have specialized roles in regulating proteostasis.


Subject(s)
Deubiquitinating Enzymes/metabolism , Evolution, Molecular , Polyubiquitin/metabolism , Amino Acid Sequence , Catalytic Domain , Conserved Sequence , Deubiquitinating Enzymes/chemistry , Humans , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Models, Molecular , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Protein Folding , Structure-Activity Relationship , Substrate Specificity , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/chemistry , Ubiquitin Thiolesterase/metabolism , Ubiquitination
12.
J Biol Chem ; 298(10): 102414, 2022 10.
Article in English | MEDLINE | ID: mdl-36007613

ABSTRACT

Legionella pneumophila, a bacterial pathogen that causes a severe pneumonia known as Legionnaires' disease, extensively exploits the ubiquitin (Ub) pathway in the infected host cells through certain virulence effectors excreted by the Dot/Icm system. To date, several Dot/Icm effectors have been found to act as Ub ligases, and four effectors, including LotA, LotB, LotC, and Ceg7, have been identified as deubiquitinases (DUBs) from the ovarian tumor (OTU) domain family. LotA is unique among other OTU DUBs because it possesses two distinct DUB domains and exclusively exhibits catalytic activity against K6-linked diUb and polyUb chains. However, the structure of LotA and the molecular mechanism for the dual DUB activity remains elusive. In this study, we solved the structure of LotA in complex with proximally bound Ub and distal covalently bound Ub. Both Ub molecules are bound to the DUB1 domain and mimic a K6-linked diUb. Structural analysis reveals that the DUB1 domain utilizes a distinct mechanism for recognition of the K6-linked diUb within a large S1' binding site that is uncommon to OTU DUBs. Structural fold of the LotA DUB2 domain closely resembles LotB and LotC, similarly containing an extra α-helix lobe that has been demonstrated to play an important role in Ub binding. Collectively, our study uncovers the structural basis for the dual catalytic activity of the unique OTU family DUB LotA.


Subject(s)
Bacterial Proteins , Deubiquitinating Enzymes , Legionella pneumophila , Bacterial Proteins/chemistry , Deubiquitinating Enzymes/chemistry , Legionella pneumophila/enzymology , Ubiquitin/metabolism , Catalysis , Protein Domains , Protein Conformation, alpha-Helical
13.
Bioorg Med Chem Lett ; 94: 129458, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37634761

ABSTRACT

Malaria continues to be a major burden on global health, responsible for 619,000 deaths in 2021. The causative agent of malaria is the eukaryotic parasite Plasmodium. Resistance to artemisinin-based combination therapies (ACTs), the current first-line treatment for malaria, has emerged in Asia, South America, and more recently Africa, where >90% of all malaria-related deaths occur. This has necessitated the identification and investigation of novel parasite proteins and pathways as antimalarial targets, including components of the ubiquitin proteasome system. Here, we investigate Plasmodium falciparum deubiquitinase ubiquitin C-terminal hydrolase L3 (PfUCHL3) as one such target. We carried out a high-throughput screen with covalent fragments and identified seven scaffolds that selectively inhibit the plasmodial UCHL3, but not human UCHL3 or the closely related human UCHL1. After assessing toxicity in human cells, we identified four promising hits and demonstrated their efficacy against asexual P. falciparum blood stages and P. berghei sporozoite stages.


Subject(s)
Antimalarials , Deubiquitinating Enzymes , Folic Acid Antagonists , Antimalarials/pharmacology , Eukaryota , Plasmodium falciparum , Proteasome Endopeptidase Complex , Deubiquitinating Enzymes/antagonists & inhibitors , Deubiquitinating Enzymes/chemistry , Protozoan Proteins
14.
Proc Natl Acad Sci U S A ; 117(3): 1321-1329, 2020 01 21.
Article in English | MEDLINE | ID: mdl-31896586

ABSTRACT

Cysteine thiols of many cancer-associated proteins are attractive targets of anticancer agents. Herein, we unequivocally demonstrate a distinct thiol-targeting property of gold(III) mesoporphyrin IX dimethyl ester (AuMesoIX) and its anticancer activities. While the binding of cysteine thiols with metal complexes usually occurs via M-S bond formation, AuMesoIX is unique in that the meso-carbon atom of the porphyrin ring is activated by the gold(III) ion to undergo nucleophilic aromatic substitution with thiols. AuMesoIX was shown to modify reactive cysteine residues and inhibit the activities of anticancer protein targets including thioredoxin, peroxiredoxin, and deubiquitinases. Treatment of cancer cells with AuMesoIX resulted in the formation of gold-bound sulfur-rich protein aggregates, oxidative stress-mediated cytotoxicity, and accumulation of ubiquitinated proteins. Importantly, AuMesoIX exhibited effective antitumor activity in mice. Our study has uncovered a gold(III)-induced ligand scaffold reactivity for thiol targeting that can be exploited for anticancer applications.


Subject(s)
Antineoplastic Agents/chemistry , Cysteine/chemistry , Gold/chemistry , Mesoporphyrins/chemistry , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Deubiquitinating Enzymes/chemistry , Deubiquitinating Enzymes/metabolism , HCT116 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms, Experimental/drug therapy , Peroxiredoxins/chemistry , Peroxiredoxins/metabolism , Protein Binding , Thioredoxins/chemistry , Thioredoxins/metabolism , Tissue Distribution
15.
Nature ; 538(7625): 402-405, 2016 Oct 20.
Article in English | MEDLINE | ID: mdl-27732584

ABSTRACT

The post-translational modification of proteins with polyubiquitin regulates virtually all aspects of cell biology. Eight distinct chain linkage types co-exist in polyubiquitin and are independently regulated in cells. This 'ubiquitin code' determines the fate of the modified protein. Deubiquitinating enzymes of the ovarian tumour (OTU) family regulate cellular signalling by targeting distinct linkage types within polyubiquitin, and understanding their mechanisms of linkage specificity gives fundamental insights into the ubiquitin system. Here we reveal how the deubiquitinase Cezanne (also known as OTUD7B) specifically targets Lys11-linked polyubiquitin. Crystal structures of Cezanne alone and in complex with monoubiquitin and Lys11-linked diubiquitin, in combination with hydrogen-deuterium exchange mass spectrometry, enable us to reconstruct the enzymatic cycle in great detail. An intricate mechanism of ubiquitin-assisted conformational changes activates the enzyme, and while all chain types interact with the enzymatic S1 site, only Lys11-linked chains can bind productively across the active site and stimulate catalytic turnover. Our work highlights the plasticity of deubiquitinases and indicates that new conformational states can occur when a true substrate, such as diubiquitin, is bound at the active site.


Subject(s)
Deubiquitinating Enzymes/metabolism , Endopeptidases/metabolism , Lysine/metabolism , Polyubiquitin/metabolism , Biocatalysis , Catalytic Domain , Crystallography, X-Ray , Deubiquitinating Enzymes/chemistry , Deubiquitinating Enzymes/genetics , Deuterium Exchange Measurement , Endopeptidases/chemistry , Endopeptidases/genetics , Enzyme Activation , Humans , Mass Spectrometry , Models, Molecular , Protein Binding , Protein Conformation , Substrate Specificity , Ubiquitination , Ubiquitins/metabolism
16.
J Biol Chem ; 295(40): 13769-13783, 2020 10 02.
Article in English | MEDLINE | ID: mdl-32732284

ABSTRACT

Single-stranded, positive-sense RNA viruses assemble their replication complexes in infected cells from a multidomain replication polyprotein. This polyprotein usually contains at least one protease, the primary function of which is to process the polyprotein into mature proteins. Such proteases also may have other functions in the replication cycle. For instance, cysteine proteases (PRO) frequently double up as ubiquitin hydrolases (DUB), thus interfering with cellular processes critical for virus replication. We previously reported the crystal structures of such a PRO/DUB from Turnip yellow mosaic virus (TYMV) and of its complex with one of its PRO substrates. Here we report the crystal structure of TYMV PRO/DUB in complex with ubiquitin. We find that PRO/DUB recognizes ubiquitin in an unorthodox way: It interacts with the body of ubiquitin through a split recognition motif engaging both the major and the secondary recognition patches of ubiquitin (Ile44 patch and Ile36 patch, respectively, including Leu8, which is part of the two patches). However, the contacts are suboptimal on both sides. Introducing a single-point mutation in TYMV PRO/DUB aimed at improving ubiquitin-binding led to a much more active DUB. Comparison with other PRO/DUBs from other viral families, particularly coronaviruses, suggests that low DUB activities of viral PRO/DUBs may generally be fine-tuned features of interaction with host factors.


Subject(s)
Deubiquitinating Enzymes/chemistry , Peptide Hydrolases/chemistry , Tymovirus/enzymology , Ubiquitin/chemistry , Viral Proteins/chemistry , Crystallography, X-Ray , Deubiquitinating Enzymes/genetics , Peptide Hydrolases/genetics , Tymovirus/genetics , Ubiquitin/genetics , Viral Proteins/genetics
17.
J Biol Chem ; 295(6): 1646-1657, 2020 02 07.
Article in English | MEDLINE | ID: mdl-31907282

ABSTRACT

Legionella pneumophila is the causative agent of the lung malady Legionnaires' disease, it modulates host function to create a niche termed the Legionella-containing vacuole (LCV) that permits intracellular L. pneumophila replication. One important aspect of such modulation is the co-option of the host ubiquitin network with a panel of effector proteins. Here, using recombinantly expressed and purified proteins, analytic ultracentrifugation, structural analysis, and computational modeling, along with deubiquitinase (DUB), and bacterial infection assays, we found that the bacterial defective in organelle trafficking/intracellular multiplication effector Ceg23 is a member of the ovarian tumor (OTU) DUB family. We found that Ceg23 displays high specificity toward Lys-63-linked polyubiquitin chains and is localized on the LCV, where it removes ubiquitin moieties from proteins ubiquitinated by the Lys-63-chain type. Analysis of the crystal structure of a Ceg23 variant lacking two putative transmembrane domains at 2.80 Å resolution revealed that despite very limited homology to established members of the OTU family at the primary sequence level, Ceg23 harbors a catalytic motif resembling those associated with typical OTU-type DUBs. ceg23 deletion increased the association of Lys-63-linked polyubiquitin with the bacterial phagosome, indicating that Ceg23 regulates Lys-63-linked ubiquitin signaling on the LCV. In summary, our findings indicate that Ceg23 contributes to the regulation of the association of Lys-63 type polyubiquitin with the Legionella phagosome. Future identification of host substrates targeted by Ceg23 could clarify the roles of these polyubiquitin chains in the intracellular life cycle of L. pneumophila and Ceg23's role in bacterial virulence.


Subject(s)
Bacterial Proteins/metabolism , Deubiquitinating Enzymes/metabolism , Legionella pneumophila/metabolism , Legionnaires' Disease/microbiology , Polyubiquitin/metabolism , Bacterial Proteins/chemistry , Deubiquitinating Enzymes/chemistry , HEK293 Cells , HeLa Cells , Humans , Legionella pneumophila/chemistry , Legionnaires' Disease/metabolism , Lysine/metabolism , Phagosomes/metabolism , Protein Conformation , Substrate Specificity , Ubiquitination
18.
J Biol Chem ; 295(11): 3466-3484, 2020 03 13.
Article in English | MEDLINE | ID: mdl-32005664

ABSTRACT

Parkinson's disease (PD) is a multifactorial malady and the second most common neurodegenerative disorder, characterized by loss of dopaminergic neurons in the midbrain. A hallmark of PD pathology is the formation of intracellular protein inclusions, termed Lewy bodies (LBs). Recent MS studies have shown that OTU deubiquitinase ubiquitin aldehyde-binding 1 (OTUB1), a deubiquitinating enzyme of the OTU family, is enriched together with α-synuclein in LBs from individuals with PD and is also present in amyloid plaques associated with Alzheimer's disease. In the present study, using mammalian cell cultures and a PD mouse model, along with CD spectroscopy, atomic force microscopy, immunofluorescence-based imaging, and various biochemical assays, we demonstrate that after heat-induced protein aggregation, OTUB1 reacts strongly with both anti-A11 and anti-osteocalcin antibodies, detecting oligomeric, prefibrillar structures or fibrillar species of amyloidogenic proteins, respectively. Further, recombinant OTUB1 exhibited high thioflavin-T and Congo red binding and increased ß-sheet formation upon heat induction. The oligomeric OTUB1 aggregates were highly cytotoxic, characteristic of many amyloid proteins. OTUB1 formed inclusions in neuronal cells and co-localized with thioflavin S and with α-synuclein during rotenone-induced stress. It also co-localized with the disease-associated variant pS129-α-synuclein in rotenone-exposed mouse brains. Interestingly, OTUB1 aggregates were also associated with severe cytoskeleton damage, rapid internalization inside the neuronal cells, and mitochondrial damage, all of which contribute to neurotoxicity. In conclusion, the results of our study indicate that OTUB1 may contribute to LB pathology through its amyloidogenic properties.


Subject(s)
Amyloid/chemistry , Deubiquitinating Enzymes/toxicity , Neurotoxins/toxicity , Parkinson Disease/pathology , Protein Aggregates , Actin Cytoskeleton/metabolism , Animals , Apoptosis/drug effects , Cell Death/drug effects , Cell Line, Tumor , Computer Simulation , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Deubiquitinating Enzymes/chemistry , Disease Models, Animal , Endocytosis/drug effects , Male , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/pathology , Models, Biological , Nanostructures/chemistry , Neurons/drug effects , Neurons/metabolism , Oxidation-Reduction , Phosphoserine/metabolism , Protein Multimerization , Reactive Oxygen Species/metabolism , Rotenone , alpha-Synuclein/metabolism
19.
J Am Chem Soc ; 143(17): 6423-6433, 2021 05 05.
Article in English | MEDLINE | ID: mdl-33885283

ABSTRACT

Terminal unactivated alkynes are nowadays considered the golden standard for cysteine-reactive warheads in activity-based probes (ABPs) targeting cysteine deubiquitinating enzymes (DUBs). In this work, we study the versatility of the thiol-alkyne addition reaction in more depth. Contrary to previous findings with UCHL3, we now show that covalent adduct formation can progress with substituents on the terminal or internal alkyne position. Strikingly, acceptance of alkyne substituents is strictly DUB-specific as this is not conserved among members of the same subfamily. Covalent adduct formation with the catalytic cysteine residue was validated by gel analysis and mass spectrometry of intact ABP-treated USP16CDWT and catalytically inactive mutant USP16CDC205A. Bottom-up mass spectrometric analysis of the covalent adduct with a deuterated propargyl ABP provides mechanistic understanding of the in situ thiol-alkyne reaction, identifying the alkyne rather than an allenic intermediate as the reactive species. Furthermore, kinetic analysis revealed that introduction of (bulky/electron-donating) methyl substituents on the propargyl moiety decreases the rate of covalent adduct formation, thus providing a rational explanation for the commonly lower level of observed covalent adduct compared to unmodified alkynes. Altogether, our work extends the scope of possible propargyl derivatives in cysteine targeting ABPs from unmodified terminal alkynes to internal and substituted alkynes, which we anticipate will have great value in the development of ABPs with improved selectivity profiles.


Subject(s)
Alkynes/chemistry , Cysteine Proteases/chemistry , Pargyline/analogs & derivatives , Sulfhydryl Compounds/chemistry , Deubiquitinating Enzymes/chemistry , HEK293 Cells , Humans , Pargyline/chemistry , Propylamines/chemistry , Ubiquitin Thiolesterase/chemistry
20.
Org Biomol Chem ; 19(10): 2177-2181, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33630007

ABSTRACT

Eosin Y was assessed for its ability to induce a thiol-ene dependent protein-protein reaction in a metal-free, oxygen-tolerant, visible light mediated system. Protein-protein coupling efficiency under these mild conditions was comparable to previously reported UV-dependent conditions. The desired thiol-ene reaction was however limited within more complex biological systems.


Subject(s)
Cysteine Endopeptidases/chemistry , Deubiquitinating Enzymes/chemistry , Eosine Yellowish-(YS)/chemistry , Molecular Probes/chemistry , Alkenes/chemistry , Catalysis/radiation effects , Cysteine/chemistry , Eosine Yellowish-(YS)/radiation effects , HEK293 Cells , Humans , Light , Molecular Probes/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL