Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 243
Filter
1.
Biochemistry ; 63(18): 2293-2299, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39189377

ABSTRACT

Knotted proteins are fascinating natural biomolecules whose backbones entangle themselves in a knot. Their particular knotted configurations provide them with a wide range of topological features. However, their folding/unfolding mechanisms, stability, and function are poorly understood. In the present work, native trapped ion mobility spectrometry-mass spectrometry (TIMS-MS) was used for characterizing structural features of two model knotted proteins: a Gordian 52 knot ubiquitin C-terminal hydrolase (UCH) and a Stevedore 61 knot (α-haloacid dehalogenase, DehI). Experimental results showed structural transitions of UCH and DehI as a function of solution composition (0-50% MeOH) and temperature (T ∼20-95 °C). An increase in the protein charge states and collision cross sections (∼2750-8750 Å2 and ∼3250-15,385 Å2 for UCH and DehI, respectively) with the solution organic content (OC) and temperature suggested a three-step unfolding pathway with at least four structural transitions. Results also showed that the integrity of the UCH knot core was more resistant to thermal unfolding when compared to DehI; however, both knot cores can be disrupted with the increase in the solution OC. Additional enzymatic digestion experiments using carboxypeptidase Y combined with molecular dynamics simulations showed that the knot core was preserved between Glu20 and Glu188 and Arg89 and His304 residues for UCH and DehI, respectively, where disruption of the knot core led to structural collapse followed by unfolding events. This work highlights the potential of solution OC and temperature studies combined with native TIMS-MS for the comprehensive characterization of knotted proteins to gain a better understanding of their structural transitions.


Subject(s)
Protein Conformation , Ubiquitin Thiolesterase , Ubiquitin Thiolesterase/chemistry , Ubiquitin Thiolesterase/metabolism , Protein Folding , Protein Unfolding , Models, Molecular , Humans , Ion Mobility Spectrometry/methods
2.
J Chem Inf Model ; 64(17): 6827-6837, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39045738

ABSTRACT

Knotted proteins are present in nature, but there is still an open issue regarding the existence of a universal role for these remarkable structures. To address this question, we used classical molecular dynamics (MD) simulations combined with in vitro experiments to investigate the role of the Gordian knot in the catalytic activity of UCH-L1. To create an unknotted form of UCH-L1, we modified its amino acid sequence by truncating several residues from its N-terminus. Remarkably, we find that deleting the first two N-terminal residues leads to a partial loss of enzyme activity with conservation of secondary structural content and knotted topological state. This happens because the integrity of the N-terminus is critical to ensure the correct alignment of the catalytic triad. However, the removal of five residues from the N-terminus, which significantly disrupts the native structure and the topological state, leads to a complete loss of enzymatic activity. Overall, our findings indicate that UCH-L1's catalytic activity depends critically on the integrity of the N-terminus and the secondary structure content, with the latter being strongly coupled with the knotted topological state.


Subject(s)
Ubiquitin Thiolesterase , Humans , Amino Acid Sequence , Biocatalysis , Molecular Dynamics Simulation , Protein Conformation , Protein Structure, Secondary , Ubiquitin Thiolesterase/chemistry , Ubiquitin Thiolesterase/metabolism
3.
Cells ; 13(11)2024 May 31.
Article in English | MEDLINE | ID: mdl-38891087

ABSTRACT

Ubiquitin-specific protease 14 (USP14), one of the three major proteasome-associated deubiquitinating enzymes (DUBs), is known to be activated by the AKT-mediated phosphorylation at Ser432. Thereby, AKT can regulate global protein degradation by controlling the ubiquitin-proteasome system (UPS). However, the exact molecular mechanism of USP14 activation by AKT phosphorylation at the atomic level remains unknown. By performing the molecular dynamics (MD) simulation of the USP14 catalytic domain at three different states (inactive, active, and USP14-ubiquitin complex), we characterized the change in structural dynamics by phosphorylation. We observed that the Ser432 phosphorylation induced substantial conformational changes of USP14 in the blocking loop (BL) region to fold it from an open loop into a ß-sheet, which is critical for USP14 activation. Furthermore, phosphorylation also increased the frequency of critical hydrogen bonding and salt bridge interactions between USP14 and ubiquitin, which is essential for DUB activity. Structural dynamics insights from this study pinpoint the important local conformational landscape of USP14 by the phosphorylation event, which would be critical for understanding USP14-mediated proteasome regulation and designing future therapeutics.


Subject(s)
Molecular Dynamics Simulation , Proto-Oncogene Proteins c-akt , Ubiquitin Thiolesterase , Phosphorylation , Ubiquitin Thiolesterase/metabolism , Ubiquitin Thiolesterase/chemistry , Proto-Oncogene Proteins c-akt/metabolism , Humans , Ubiquitin/metabolism , Enzyme Activation , Catalytic Domain , Protein Binding , Protein Conformation
4.
EMBO Rep ; 25(7): 2950-2973, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38816515

ABSTRACT

The development of cancer therapeutics is often hindered by the fact that specific oncogenes cannot be directly pharmaceutically addressed. Targeting deubiquitylases that stabilize these oncogenes provides a promising alternative. USP28 and USP25 have been identified as such target deubiquitylases, and several small-molecule inhibitors indiscriminately inhibiting both enzymes have been developed. To obtain insights into their mode of inhibition, we structurally and functionally characterized USP28 in the presence of the three different inhibitors AZ1, Vismodegib and FT206. The compounds bind into a common pocket acting as a molecular sink. Our analysis provides an explanation why the two enzymes are inhibited with similar potency while other deubiquitylases are not affected. Furthermore, a key glutamate residue at position 366/373 in USP28/USP25 plays a central structural role for pocket stability and thereby for inhibition and activity. Obstructing the inhibitor-binding pocket by mutation of this glutamate may provide a tool to accelerate future drug development efforts for selective inhibitors of either USP28 or USP25 targeting distinct binding pockets.


Subject(s)
Ubiquitin Thiolesterase , Ubiquitin Thiolesterase/chemistry , Ubiquitin Thiolesterase/antagonists & inhibitors , Ubiquitin Thiolesterase/metabolism , Ubiquitin Thiolesterase/genetics , Humans , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Binding Sites , Pyridines/chemistry , Pyridines/pharmacology , Protein Binding , Models, Molecular
5.
Nat Commun ; 15(1): 4519, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38806474

ABSTRACT

Protein ubiquitination regulates a wide range of cellular processes. The degree of protein ubiquitination is determined by the delicate balance between ubiquitin ligase (E3)-mediated ubiquitination and deubiquitinase (DUB)-mediated deubiquitination. In comparison to the E3-substrate interactions, the DUB-substrate interactions (DSIs) remain insufficiently investigated. To address this challenge, we introduce a protein sequence-based ab initio method, TransDSI, which transfers proteome-scale evolutionary information to predict unknown DSIs despite inadequate training datasets. An explainable module is integrated to suggest the critical protein regions for DSIs while predicting DSIs. TransDSI outperforms multiple machine learning strategies against both cross-validation and independent test. Two predicted DUBs (USP11 and USP20) for FOXP3 are validated by "wet lab" experiments, along with two predicted substrates (AR and p53) for USP22. TransDSI provides new functional perspective on proteins by identifying regulatory DSIs, and offers clues for potential tumor drug target discovery and precision drug application.


Subject(s)
Deubiquitinating Enzymes , Proteome , Ubiquitination , Humans , Proteome/metabolism , Deubiquitinating Enzymes/metabolism , Deubiquitinating Enzymes/genetics , Deep Learning , Ubiquitin Thiolesterase/metabolism , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/chemistry , Substrate Specificity , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Machine Learning , Protein Binding , Amino Acid Sequence , Thiolester Hydrolases
6.
J Med Chem ; 67(6): 4496-4524, 2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38488146

ABSTRACT

Dysregulation of the ubiquitin-proteasome systems is a hallmark of various disease states including neurodegenerative diseases and cancer. Ubiquitin C-terminal hydrolase L1 (UCHL1), a deubiquitinating enzyme, is expressed primarily in the central nervous system under normal physiological conditions, however, is considered an oncogene in various cancers, including melanoma, lung, breast, and lymphoma. Thus, UCHL1 inhibitors could serve as a viable treatment strategy against these aggressive cancers. Herein, we describe a covalent fragment screen that identified the chloroacetohydrazide scaffold as a covalent UCHL1 inhibitor. Subsequent optimization provided an improved fragment with single-digit micromolar potency against UCHL1 and selectivity over the closely related UCHL3. The molecule demonstrated efficacy in cellular assays of metastasis. Additionally, we report a ligand-bound crystal structure of the most potent molecule in complex with UCHL1, providing insight into the binding mode and information for future optimization.


Subject(s)
Neoplasms , Ubiquitin Thiolesterase , Humans , Ubiquitin Thiolesterase/chemistry , Ubiquitin Thiolesterase/metabolism , Ubiquitin/metabolism , Breast , Proteasome Endopeptidase Complex
7.
Int J Biol Macromol ; 263(Pt 1): 130309, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38382779

ABSTRACT

Maintaining protein balance within a cell is essential for proper cellular function, and disruptions in the ubiquitin-proteasome pathway, which is responsible for degrading and recycling unnecessary or damaged proteins, can lead to various diseases. Deubiquitinating enzymes play a vital role in regulating protein homeostasis by removing ubiquitin chains from substrate proteins, thereby controlling important cellular processes, such as apoptosis and DNA repair. Among these enzymes, ubiquitin-specific protease 7 (USP7) is of particular interest. USP7 is a cysteine protease consisting of a TRAF region, catalytic region, and C-terminal ubiquitin-like (UBL) region, and it interacts with tumor suppressors, transcription factors, and other key proteins involved in cell cycle regulation and epigenetic control. Moreover, USP7 has been implicated in the pathogenesis and progression of various diseases, including cancer, inflammation, neurodegenerative conditions, and viral infections. Overall, characterizing the functions of USP7 is crucial for understanding the pathophysiology of diverse diseases and devising innovative therapeutic strategies. This article reviews the structure and function of USP7 and its complexes, its association with diseases, and its known inhibitors and thus represents a valuable resource for advancing USP7 inhibitor development and promoting potential future treatment options for a wide range of diseases.


Subject(s)
Proteostasis , Ubiquitin , Ubiquitin-Specific Peptidase 7/genetics , Ubiquitin-Specific Peptidase 7/chemistry , Ubiquitin-Specific Peptidase 7/metabolism , Ubiquitin/chemistry , Catalytic Domain , Ubiquitin Thiolesterase/chemistry
8.
J Mol Biol ; 436(4): 168438, 2024 02 15.
Article in English | MEDLINE | ID: mdl-38185323

ABSTRACT

A mutant of ubiquitin C-terminal hydrolase L1 (UCHL1) detected in early-onset neurodegenerative patients, UCHL1R178Q, showed higher catalytic activity than wild-type UCHL1 (UCHL1WT). Lying within the active-site pocket, the arginine is part of an interaction network that holds the catalytic histidine in an inactive arrangement. However, the structural basis and mechanism of enzymatic activation upon glutamine substitution was not understood. We combined X-ray crystallography, protein nuclear magnetic resonance (NMR) analysis, enzyme kinetics, covalent inhibition analysis, and biophysical measurements to delineate activating factors in the mutant. While the crystal structure of UCHL1R178Q showed nearly the same arrangement of the catalytic residues and active-site pocket, the mutation caused extensive alteration in the chemical environment and dynamics of more than 30 residues, some as far as 15 Å away from the site of mutation. Significant broadening of backbone amide resonances in the HSQC spectra indicates considerable backbone dynamics changes in several residues, in agreement with solution small-angle X-ray scattering (SAXS) analyses which indicate an overall increase in protein flexibility. Enzyme kinetics show the activation is due to a kcat effect despite a slightly weakened substrate affinity. In line with this, the mutant shows a higher second-order rate constant (kinact/Ki) in a reaction with a substrate-derived irreversible inhibitor, Ub-VME, compared to the wild-type enzyme, an observation indicative of a more reactive catalytic cysteine in the mutant. Together, the observations underscore structural plasticity as a factor contributing to enzyme kinetic behavior which can be modulated through mutational effects.


Subject(s)
Catalytic Domain , Cysteine , Neurodegenerative Diseases , Ubiquitin Thiolesterase , Humans , Binding Sites/genetics , Cysteine/chemistry , Cysteine/genetics , Kinetics , Mutagenesis, Site-Directed , Nuclear Magnetic Resonance, Biomolecular , Scattering, Small Angle , Ubiquitin Thiolesterase/chemistry , Ubiquitin Thiolesterase/genetics , X-Ray Diffraction , Neurodegenerative Diseases/genetics
9.
J Biol Chem ; 299(7): 104911, 2023 07.
Article in English | MEDLINE | ID: mdl-37311534

ABSTRACT

Reversible lysine-63 (K63) polyubiquitination regulates proinflammatory signaling in vascular smooth muscle cells (SMCs) and plays an integral role in atherosclerosis. Ubiquitin-specific peptidase 20 (USP20) reduces NFκB activation triggered by proinflammatory stimuli, and USP20 activity attenuates atherosclerosis in mice. The association of USP20 with its substrates triggers deubiquitinase activity; this association is regulated by phosphorylation of USP20 on Ser334 (mouse) or Ser333 (human). USP20 Ser333 phosphorylation was greater in SMCs of atherosclerotic segments of human arteries as compared with nonatherosclerotic segments. To determine whether USP20 Ser334 phosphorylation regulates proinflammatory signaling, we created USP20-S334A mice using CRISPR/Cas9-mediated gene editing. USP20-S334A mice developed ∼50% less neointimal hyperplasia than congenic WT mice after carotid endothelial denudation. WT carotid SMCs showed substantial phosphorylation of USP20 Ser334, and WT carotids demonstrated greater NFκB activation, VCAM-1 expression, and SMC proliferation than USP20-S334A carotids. Concordantly, USP20-S334A primary SMCs in vitro proliferated and migrated less than WT SMCs in response to IL-1ß. An active site ubiquitin probe bound to USP20-S334A and USP20-WT equivalently, but USP20-S334A associated more avidly with TRAF6 than USP20-WT. IL-1ß induced less K63-linked polyubiquitination of TRAF6 and less downstream NFκB activity in USP20-S334A than in WT SMCs. Using in vitro phosphorylation with purified IRAK1 and siRNA-mediated gene silencing of IRAK1 in SMCs, we identified IRAK1 as a novel kinase for IL-1ß-induced USP20 Ser334 phosphorylation. Our findings reveal novel mechanisms regulating IL-1ß-induced proinflammatory signaling: by phosphorylating USP20 Ser334, IRAK1 diminishes the association of USP20 with TRAF6 and thus augments NFκB activation, SMC inflammation, and neointimal hyperplasia.


Subject(s)
Atherosclerosis , Inflammation , Interleukin-1 Receptor-Associated Kinases , Interleukin-1beta , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Phosphoserine , Ubiquitin Thiolesterase , Animals , Humans , Mice , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cells, Cultured , Hyperplasia/metabolism , Hyperplasia/pathology , Inflammation/metabolism , Inflammation/pathology , Interleukin-1 Receptor-Associated Kinases/chemistry , Interleukin-1 Receptor-Associated Kinases/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Phosphorylation , Phosphoserine/metabolism , TNF Receptor-Associated Factor 6/metabolism , Ubiquitin Thiolesterase/chemistry , Ubiquitin Thiolesterase/metabolism , NF-kappa B/metabolism , Carotid Arteries/metabolism , Carotid Arteries/pathology , Interleukin-1beta/metabolism , Ubiquitination
10.
Protein Pept Lett ; 30(7): 552-561, 2023.
Article in English | MEDLINE | ID: mdl-37246324

ABSTRACT

Cancer is an important chronic non-communicable disease that endangers human health and has become the main cause of death of residents around the world in the 21st century. At present, most of the mature treatment methods stay at the level of cell and tissue, which is difficult to fundamentally solve the problem of cancer. Therefore, explaining the pathogenesis of cancer at the molecular level becomes the answer to the key problem of cancer regulation. BRCA-associated protein 1 (brca1- associated protein 1) is a kind of ubiquitination enzyme encoded by the BAP1 gene and composed of 729 amino acids. As a carcinogenic protein, BAP1 can affect the cancer cell cycle and proliferation capacity, mutation, and deletion. For example, depending on catalytic activity, it participates in the regulation of intracellular function through transcription, epigenetic, and DNA damage repair. This article mainly reviews the basic structure and function of BAP1 in cells, its role in cancer development, and cancer-related mutants.


Subject(s)
DNA Repair , Neoplasms , Humans , Cell Line, Tumor , Mutation , Ubiquitination , Cell Cycle , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/chemistry , Ubiquitin Thiolesterase/metabolism , Neoplasms/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
11.
Nature ; 616(7955): 176-182, 2023 04.
Article in English | MEDLINE | ID: mdl-36991118

ABSTRACT

Repression of gene expression by protein complexes of the Polycomb group is a fundamental mechanism that governs embryonic development and cell-type specification1-3. The Polycomb repressive deubiquitinase (PR-DUB) complex removes the ubiquitin moiety from monoubiquitinated histone H2A K119 (H2AK119ub1) on the nucleosome4, counteracting the ubiquitin E3 ligase activity of Polycomb repressive complex 1 (PRC1)5 to facilitate the correct silencing of genes by Polycomb proteins and safeguard active genes from inadvertent silencing by PRC1 (refs. 6-9). The intricate biological function of PR-DUB requires accurate targeting of H2AK119ub1, but PR-DUB can deubiquitinate monoubiquitinated free histones and peptide substrates indiscriminately; the basis for its exquisite nucleosome-dependent substrate specificity therefore remains unclear. Here we report the cryo-electron microscopy structure of human PR-DUB, composed of BAP1 and ASXL1, in complex with the chromatosome. We find that ASXL1 directs the binding of the positively charged C-terminal extension of BAP1 to nucleosomal DNA and histones H3-H4 near the dyad, an addition to its role in forming the ubiquitin-binding cleft. Furthermore, a conserved loop segment of the catalytic domain of BAP1 is situated near the H2A-H2B acidic patch. This distinct nucleosome-binding mode displaces the C-terminal tail of H2A from the nucleosome surface, and endows PR-DUB with the specificity for H2AK119ub1.


Subject(s)
Deubiquitinating Enzymes , Histones , Polycomb Repressive Complex 1 , Polycomb-Group Proteins , Humans , Cryoelectron Microscopy , Histones/chemistry , Histones/metabolism , Nucleosomes/chemistry , Nucleosomes/genetics , Nucleosomes/metabolism , Polycomb Repressive Complex 1/chemistry , Polycomb Repressive Complex 1/metabolism , Polycomb Repressive Complex 1/ultrastructure , Polycomb-Group Proteins/chemistry , Polycomb-Group Proteins/metabolism , Polycomb-Group Proteins/ultrastructure , Ubiquitin/metabolism , Ubiquitin Thiolesterase/chemistry , Ubiquitin Thiolesterase/metabolism , Ubiquitin Thiolesterase/ultrastructure , Ubiquitination , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Repressor Proteins/ultrastructure , Catalytic Domain , Deubiquitinating Enzymes/classification , Deubiquitinating Enzymes/metabolism , Deubiquitinating Enzymes/ultrastructure , Substrate Specificity , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/ultrastructure
12.
Angew Chem Int Ed Engl ; 61(33): e202204395, 2022 08 15.
Article in English | MEDLINE | ID: mdl-35691827

ABSTRACT

The tumor suppressor p53 is the most frequently mutated gene in human cancer and more than half of cancers contain p53 mutations. The development of novel and effective therapeutic strategies for p53 mutant cancer therapy is a big challenge and highly desirable. Ubiquitin-specific protease 7 (USP7), also known as HAUSP, is a deubiquitinating enzyme and proposed to stabilize the oncogenic E3 ubiquitin ligase MDM2 that promotes the proteosomal degradation of p53. Herein, we report the design and characterization of U7D-1 as the first selective USP7-degrading Proteolysis Targeting Chimera (PROTAC). U7D-1 showed selective and effective USP7 degradation, and maintained potent cell growth inhibition in p53 mutant cancer cells, with USP7 inhibitor showing no activity. These data clearly demonstrated the practicality and importance of PROTAC as a preliminary chemical tool for investigating USP7 protein functions and a promising method for potential p53 mutant cancer therapy.


Subject(s)
Neoplasms , Tumor Suppressor Protein p53 , Cell Line, Tumor , Humans , Neoplasms/metabolism , Proteolysis , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ubiquitin Thiolesterase/chemistry , Ubiquitin Thiolesterase/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Specific Peptidase 7/metabolism
13.
Biomol NMR Assign ; 16(2): 197-203, 2022 10.
Article in English | MEDLINE | ID: mdl-35536398

ABSTRACT

Ubiquitin specific protease 7 (USP7) is a deubiquitinating enzyme, which removes ubiquitin tag from numerous protein substrates involved in diverse cellular processes such as cell cycle regulation, apoptosis and DNA damage response. USP7 affects stability, interaction network and cellular localization of its cellular and viral substrates by controlling their ubiquitination status. The large 41 kDa catalytic domain of USP7 harbors the active site of the enzyme. Here we present a nearly complete (93%) NMR resonance assignment of isoleucine, leucine and valine (ILV) side-chains of the USP7 catalytic domain along with a refined nearly complete (93%) assignment of its backbone resonances. The reported ILV methyl group assignment will facilitate further NMR investigations of structure, interactions and conformational dynamics of the USP7 enzyme.


Subject(s)
Isoleucine , Valine , Catalytic Domain , Humans , Leucine , Nuclear Magnetic Resonance, Biomolecular , Ubiquitin Thiolesterase/chemistry , Ubiquitin-Specific Peptidase 7/metabolism , Ubiquitins
14.
J Mol Biol ; 434(9): 167553, 2022 05 15.
Article in English | MEDLINE | ID: mdl-35317997

ABSTRACT

BRAC1 associated protein-1 (BAP1) is a major tumor suppressor involved in many cancers. The deubiquitinase (DUB) activity of BAP1 is essential for its nuclear localization, histone remodeling and proteostasis associated with mitochondrial calcium flux. Loss of the DUB activity due to catalytic mutations within the ubiquitin C-terminal hydrolase (UCH) domain of BAP1 (BAP1-UCH) directly contributes to oncogenesis. Nevertheless, it is non-trivial to rationalize how the other high-frequency but non-catalytic mutations within the BAP1-UCH lead to malignancies. Here we used multiplex spectroscopic, thermodynamic and biophysical analyses to investigate the impacts of eleven high-occurrence mutations within BAP1-UCH on the structure, folding and function. Several mutations significantly destabilize BAP1-UCH and increase its aggregation propensity. Hydrogen-deuterium exchange mass spectrometry data revealed allosteric destabilizations caused by mutations distant from the catalytic site. Our findings gave a comprehensive and multiscale account of the molecular basis of how these non-catalytic mutations within BAP1-UCH may be implicated in oncogenesis.


Subject(s)
Carcinogenesis , Tumor Suppressor Proteins , Ubiquitin Thiolesterase , Carcinogenesis/genetics , Humans , Mutation , Protein Domains , Structure-Activity Relationship , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/chemistry , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism
15.
Biochem Biophys Res Commun ; 599: 57-62, 2022 04 09.
Article in English | MEDLINE | ID: mdl-35176625

ABSTRACT

Deubiquitinating enzymes (DUBs) form a large protease family involved in a myriad of biological and pathological processes, including ROS sensors. ROS-mediated inhibition of their DUB activities is critical for fine-tuning the stress-activated signaling pathways. Here, we demonstrate that the ubiquitin C-terminal hydrolase (UCH) domain of BAP1 (BAP1-UCH) is highly sensitive to moderate oxidative stress. Oxidation of the catalytic C91 significantly destabilizes BAP1-UCH and increases the population of partially unfolded form, which is prone to aggregation. Unlike other DUBs, the oxidation-induced structural and functional loss of BAP1-UCH cannot be fully reversed by reducing agents. The oligomerization of oxidized BAP1-UCH is attributed to inter-molecular disulfide bond formation. Hydrogen-deuterium mass exchange spectrometry (HDX-MS) reveals increased fluctuations of the central ß-sheet upon oxidation. Our findings suggest that oxidation-mediated functional loss and increased aggregation propensity may contribute to oncogenesis associated with BAP1.


Subject(s)
Cysteine/metabolism , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/chemistry , Ubiquitin Thiolesterase/metabolism , Carcinogenesis , Circular Dichroism , Disulfides/chemistry , Humans , Hydrogen Deuterium Exchange-Mass Spectrometry , Hydrogen Peroxide/chemistry , Oxidation-Reduction , Oxidative Stress , Protein Aggregation, Pathological/metabolism , Protein Domains , Protein Folding , Spectrometry, Fluorescence , Tumor Suppressor Proteins/genetics , Ubiquitin Thiolesterase/genetics
16.
Biomolecules ; 12(1)2022 01 01.
Article in English | MEDLINE | ID: mdl-35053210

ABSTRACT

There is currently a lack of reliable methods and strategies to probe the deubiquitinating enzyme UCHL3. Current small molecules reported for this purpose display reduced potency and selectivity in cellular assays. To bridge this gap and provide an alternative approach to probe UCHL3, our group has carried out the rational design of ubiquitin-variant activity-based probes with selectivity for UCHL3 over the closely related UCHL1 and other DUBs. The approach successfully produced a triple-mutant ubiquitin variant activity-based probe, UbVQ40V/T66K/V70F-PRG, that was ultimately 20,000-fold more selective for UCHL3 over UCHL1 when assessed by rate of inactivation assays. This same variant was shown to selectively form covalent adducts with UCHL3 in MDA-MB-231 breast cancer cells and no reactivity toward other DUBs expressed. Overall, this study demonstrates the feasibility of the approach and also provides insight into how this approach may be applied to other DUB targets.


Subject(s)
Amino Acid Substitution , Mutation, Missense , Ubiquitin Thiolesterase , Ubiquitin , Cell Line, Tumor , Humans , Ubiquitin/chemistry , Ubiquitin/genetics , Ubiquitin/metabolism , Ubiquitin Thiolesterase/chemistry , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism
17.
Eur J Med Chem ; 227: 113970, 2022 Jan 05.
Article in English | MEDLINE | ID: mdl-34752952

ABSTRACT

In the past few years, researchers have shed light on the immense importance of ubiquitin in numerous regulatory pathways. The post-translational addition of mono or poly-ubiquitin molecules namely "ubiquitinoylation" is therefore pivotal to maintain the cell's vitality, maturation, differentiation, and division. Part of conserving homeostasis stems from maintaining the ubiquitin pool in the vicinity of the cell's intracellular environment; this crucial role is played by deubiquitylating enzymes (DUBs) that cleave ubiquitin molecules from target molecules. To date, they are categorized into 7 families with ubiquitin carboxyl c-terminal de-hydrolase family (UCH) as the most common and well-studied. Ubiquitin C-terminal hydrolase L (UCH-L3) is a significant protein in this family as it has been implicated in many molecular and cellular processes with its mRNA identified in a range of body tissues including the brain. It goes without saying that it manifests in maintaining health and when abnormally regulated in disease. As it is an attractive small molecule drug target, scientists have used high throughput screening (HTS) and other drug discovery methods to discover inhibitors for this enzyme for the treatment of cancer and neurodegenerative diseases. In this review we present an overview of UCH-L3 catalytic mechanism, structure, its role in DNA repair and cancer along with the inhibitors discovered so far to halt its activity.


Subject(s)
Enzyme Inhibitors/pharmacology , Ubiquitin Thiolesterase/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Humans , Structure-Activity Relationship , Ubiquitin Thiolesterase/chemistry , Ubiquitin Thiolesterase/metabolism
18.
Proc Natl Acad Sci U S A ; 118(48)2021 11 30.
Article in English | MEDLINE | ID: mdl-34815344

ABSTRACT

Carriers of heterozygous germline BAP1 mutations (BAP1+/-) are affected by the "BAP1 cancer syndrome." Although they can develop almost any cancer type, they are unusually susceptible to asbestos carcinogenesis and mesothelioma. Here we investigate why among all carcinogens, BAP1 mutations cooperate with asbestos. Asbestos carcinogenesis and mesothelioma have been linked to a chronic inflammatory process promoted by the extracellular release of the high-mobility group box 1 protein (HMGB1). We report that BAP1+/- cells secrete increased amounts of HMGB1, and that BAP1+/- carriers have detectable serum levels of acetylated HMGB1 that further increase when they develop mesothelioma. We linked these findings to our discovery that BAP1 forms a trimeric protein complex with HMGB1 and with histone deacetylase 1 (HDAC1) that modulates HMGB1 acetylation and its release. Reduced BAP1 levels caused increased ubiquitylation and degradation of HDAC1, leading to increased acetylation of HMGB1 and its active secretion that in turn promoted mesothelial cell transformation.


Subject(s)
Asbestos , HMGB1 Protein/chemistry , Histone Deacetylase 1/chemistry , Tumor Suppressor Proteins/chemistry , Ubiquitin Thiolesterase/chemistry , Animals , Biomarkers, Tumor/metabolism , Carcinogenesis , Cell Nucleus/metabolism , Female , Gene-Environment Interaction , Germ-Line Mutation , HMGB1 Protein/genetics , Heterozygote , Histone Deacetylase 1/genetics , Incidence , Inflammation , Male , Mesothelioma/metabolism , Mice , Mutation , Prognosis , Protein Binding , Tumor Suppressor Proteins/metabolism , Ubiquitin/chemistry , Ubiquitin Thiolesterase/metabolism
19.
Cells ; 10(10)2021 10 04.
Article in English | MEDLINE | ID: mdl-34685632

ABSTRACT

Squamous cell carcinomas are therapeutically challenging tumor entities. Low response rates to radiotherapy and chemotherapy are commonly observed in squamous patients and, accordingly, the mortality rate is relatively high compared to other tumor entities. Recently, targeting USP28 has been emerged as a potential alternative to improve the therapeutic response and clinical outcomes of squamous patients. USP28 is a catalytically active deubiquitinase that governs a plethora of biological processes, including cellular proliferation, DNA damage repair, apoptosis and oncogenesis. In squamous cell carcinoma, USP28 is strongly expressed and stabilizes the essential squamous transcription factor ΔNp63, together with important oncogenic factors, such as NOTCH1, c-MYC and c-JUN. It is presumed that USP28 is an oncoprotein; however, recent data suggest that the deubiquitinase also has an antineoplastic effect regulating important tumor suppressor proteins, such as p53 and CHK2. In this review, we discuss: (1) The emerging role of USP28 in cancer. (2) The complexity and mutational landscape of squamous tumors. (3) The genetic alterations and cellular pathways that determine the function of USP28 in squamous cancer. (4) The development and current state of novel USP28 inhibitors.


Subject(s)
Carcinoma, Squamous Cell/genetics , Genes, Tumor Suppressor , Oncogenes , Ubiquitin Thiolesterase/genetics , Animals , Carcinoma, Squamous Cell/therapy , Humans , Models, Molecular , Ubiquitin Thiolesterase/antagonists & inhibitors , Ubiquitin Thiolesterase/chemistry , Ubiquitin Thiolesterase/metabolism
20.
J Biol Chem ; 297(4): 101107, 2021 10.
Article in English | MEDLINE | ID: mdl-34425109

ABSTRACT

Ubiquitination is a crucial posttranslational protein modification involved in a myriad of biological pathways. This modification is reversed by deubiquitinases (DUBs) that deconjugate the single ubiquitin (Ub) moiety or poly-Ub chains from substrates. In the past decade, tremendous efforts have been focused on targeting DUBs for drug discovery. However, most chemical compounds with inhibitory activity for DUBs suffer from mild potency and low selectivity. To overcome these obstacles, we developed a phage display-based protein engineering strategy for generating Ub variant (UbV) inhibitors, which was previously successfully applied to the Ub-specific protease (USP) family of cysteine proteases. In this work, we leveraged the UbV platform to selectively target STAMBP, a member of the JAB1/MPN/MOV34 (JAMM) metalloprotease family of DUB enzymes. We identified two UbVs (UbVSP.1 and UbVSP.3) that bind to STAMBP with high affinity but differ in their selectivity for the closely related paralog STAMBPL1. We determined the STAMBPL1-UbVSP.1 complex structure by X-ray crystallography, revealing hotspots of the JAMM-UbV interaction. Finally, we show that UbVSP.1 and UbVSP.3 are potent inhibitors of STAMBP isopeptidase activity, far exceeding the reported small-molecule inhibitor BC-1471. This work demonstrates that UbV technology is suitable to develop molecules as tools to target metalloproteases, which can be used to further understand the cellular function of JAMM family DUBs.


Subject(s)
Endosomal Sorting Complexes Required for Transport , Peptide Hydrolases , Peptide Library , Protease Inhibitors/chemistry , Ubiquitin Thiolesterase , Ubiquitin , Crystallography, X-Ray , Endosomal Sorting Complexes Required for Transport/antagonists & inhibitors , Endosomal Sorting Complexes Required for Transport/chemistry , Humans , Peptide Hydrolases/chemistry , Protein Structure, Quaternary , Ubiquitin/chemistry , Ubiquitin/genetics , Ubiquitin Thiolesterase/antagonists & inhibitors , Ubiquitin Thiolesterase/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL