Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
J Chem Inf Model ; 64(10): 4134-4148, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38703206

ABSTRACT

Ubiquitin-specific protease 7 (USP7) is a deubiquitinase enzyme that plays a critical role in regulating various cellular processes by cleaving ubiquitin molecules from target proteins. The C-terminal loop (CTL) motif is a specific region at the C-terminal end of the USP7 enzyme. Recent experiments suggest that the CTL motif plays a role in USP7's catalytic activity by contributing to the enzyme's structural stability, substrate recognition, and catalytic efficiency. The objective of this work is to elucidate these roles through the utilization of computational methods for molecular simulations. For this, we conducted extensive molecular dynamics (MD) simulations to investigate the conformational dynamics and protein-protein interactions within the USP7 enzyme-substrate complex with the substrate consisting of the ubiquitin tagged with the fluorescent label rhodamine 110-gly (Ub-Rho). Our results demonstrate that the CTL motif plays a crucial role in stabilizing the Ubl domains' conformation and augmenting the stability of active conformations within the enzyme-substrate complex. Conversely, the absence of the CTL motif results in increased flexibility and variability in Ubl domains' motion, leading to a reduced percentage of active conformations. Furthermore, our analysis of protein-protein interactions highlights the significance of the CTL motif in anchoring the Ubl45 domains to the catalytic domain (CD), thereby facilitating stable interactions with the substrate. Overall, our findings provide valuable insights into the conformational dynamics and protein-protein interactions inherent in the USP7 enzyme-substrate complex. These insights shed light on some mechanistic details of USP7 concerning the substrate's recognition before its catalytic action.


Subject(s)
Molecular Dynamics Simulation , Protein Binding , Ubiquitin-Specific Peptidase 7 , Ubiquitin , Ubiquitin-Specific Peptidase 7/metabolism , Ubiquitin-Specific Peptidase 7/chemistry , Substrate Specificity , Ubiquitin/metabolism , Ubiquitin/chemistry , Protein Domains , Humans , Rhodamines/chemistry , Rhodamines/metabolism , Protein Conformation
2.
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
3.
J Pharm Biomed Anal ; 227: 115305, 2023 Apr 01.
Article in English | MEDLINE | ID: mdl-36812797

ABSTRACT

Deubiquitinating enzyme ubiquitin-specific protease 7 (USP7) is a promising therapeutic target. Several USP7 inhibitors accommodated in the catalytic triad of USP7 have been reported with the aid of high-throughput screening (HTS) methods using USP7 catalytic domain truncation. However, the drawbacks of previously reported biochemical cleavage assays, including poor stability, fluorescence interference, time-consuming, expensive, more importantly the selectivity issue, have challenged the USP7-targeted drug discovery. In this work, we demonstrated the functional heterogeneity and essential role of different structural elements in the USP7 full activation, highlighting the necessity of USP7 full length in drug discovery. Apart from reported two pockets in the catalytic triad, five additional ligandable pockets were predicted based on the proposed USP7 full length models by AlphaFold and homology modelling. A reliable homogeneous time-resolved fluorescence (HTRF) HTS method was established based on the cleavage mechanism of USP7 towards the ubiquitin precursor UBA10. The USP7 full length protein was successfully expressed in the relatively cost-effective E. coli prokaryotic system and used to simulate the auto-activated USP7 in nature. Via screening our in-house library (∼ 1500 compounds), 19 hit compounds with >20% of inhibition rate were identified for further optimization. This assay will enrich the toolbox for the identification of highly potent and selective USP7 inhibitors for clinical use.


Subject(s)
Escherichia coli , High-Throughput Screening Assays , Escherichia coli/genetics , Escherichia coli/metabolism , Ubiquitin-Specific Peptidase 7/genetics , Ubiquitin-Specific Peptidase 7/chemistry , Catalytic Domain , Drug Discovery
4.
Mol Inform ; 41(7): e2100273, 2022 07.
Article in English | MEDLINE | ID: mdl-35037416

ABSTRACT

Ubiquitin-specific protease 7 (USP7) is one of the most extensively studied deubiquitinases. USP7 exhibits a high expression signature in various malignant tumors, suggesting that it is a marker of tumor prognosis and a potential drug target for anti-tumor therapy. In this study, virtual screening based on pharmacophore model and biological evaluation have been applied for the discovery of novel USP7 inhibitors targeting the catalytic active site. The TS-4 was screened from 215,480 small molecules and was found to have USP7 inhibitory activity. Preliminary in vitro studies disclosed its antiproliferative activity on human colon cancer cell lines (HCT-116 and RKO), compared with normal colon cell line (CCD841CoN). Molecular dynamics (MD) simulation revealed the combine mechanism between USP7 with the TS-4. The TS-4 formed stable interactions with Asp295, Phe409 and Tyr514, which were critical to enhance its biological activity. This compound will serve as a promising hit compound for facilitating the further design of novel USP7 inhibitors.


Subject(s)
Molecular Dynamics Simulation , Neoplasms , Ubiquitin-Specific Peptidase 7 , Humans , Molecular Docking Simulation , Neoplasms/drug therapy , Ubiquitin-Specific Peptidase 7/antagonists & inhibitors , Ubiquitin-Specific Peptidase 7/chemistry
5.
Int J Oncol ; 59(5)2021 11.
Article in English | MEDLINE | ID: mdl-34558642

ABSTRACT

Tat interactive protein, 60 kDa (TIP60) is an important partner of ubiquitin­like, containing PHD and RING finger domains 1 (UHRF1), ensuring various cellular processes through its acetyltransferase activity. TIP60 is believed to play a tumor suppressive role, partly explained by its downregulated expression in a number of cancers. The aim of the present study was to investigate the role and mechanisms of action of TIP60 in the regulation of UHRF1 expression. The results revealed that TIP60 overexpression downregulated the UHRF1 and DNA methyltransferase 1 (DNMT1) expression levels. TIP60 interfered with USP7­UHRF1 association and induced the degradation of UHRF1 in an auto­ubiquitination­dependent manner. Moreover, TIP60 activated the p73­mediated apoptotic pathway. Taken together, the data of the present study suggest that the tumor suppressor role of TIP60 is mediated by its regulation to UHRF1.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , Lysine Acetyltransferase 5/physiology , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Specific Peptidase 7/chemistry , Apoptosis , CCAAT-Enhancer-Binding Proteins/chemistry , Computational Biology , HeLa Cells , Humans , Tumor Protein p73/physiology , Ubiquitin-Protein Ligases/chemistry , Ubiquitination
6.
J Mol Biol ; 433(2): 166733, 2021 01 22.
Article in English | MEDLINE | ID: mdl-33279577

ABSTRACT

Reversible protein ubiquitination is an essential signaling mechanism within eukaryotes. Deubiquitinating enzymes are critical to this process, as they mediate removal of ubiquitin from substrate proteins. Ubiquitin-specific protease 7 (USP7) is a prominent deubiquitinating enzyme, with an extensive network of interacting partners and established roles in cell cycle activation, immune responses and DNA replication. Characterized USP7 substrates primarily interact with one of two major binding sites outside the catalytic domain. These are located on the USP7 N-terminal TRAF-like (TRAF) domain and the first and second UBL domains (UBL1-2) within the C-terminal tail. Here, we report that DNA polymerase iota (Pol ι) is a novel USP7 substrate that interacts with both TRAF and UBL1-2. Through the use of biophysical approaches and mutational analysis, we characterize both interfaces and demonstrate that bipartite binding to both USP7 domains is required for efficient Pol ι deubiquitination. Together, these data establish a new bipartite mode of USP7 substrate binding.


Subject(s)
DNA-Directed DNA Polymerase/chemistry , DNA-Directed DNA Polymerase/metabolism , Protein Interaction Domains and Motifs , Ubiquitin-Specific Peptidase 7/chemistry , Ubiquitin-Specific Peptidase 7/metabolism , Binding Sites , Catalytic Domain , Cell Line , DNA-Directed DNA Polymerase/genetics , Humans , Kinetics , Models, Molecular , Protein Binding , Protein Conformation , Recombinant Proteins , Structure-Activity Relationship , Substrate Specificity , Ubiquitin-Specific Peptidase 7/genetics , Ubiquitination , DNA Polymerase iota
7.
Curr Pharm Biotechnol ; 22(7): 995-1004, 2021.
Article in English | MEDLINE | ID: mdl-32744966

ABSTRACT

BACKGROUND: ZUFSP (Zinc-finger and UFSP domain protein) is a novel representative member of the recently characterized seventh class of deubiquitinating enzymes (DUBs). Due to the roles DUBs play in genetic instability, they have become a major drug target in cancer and neurodegenerative diseases. ZUFSP, being a DUB enzyme has also been implicated in genetic stability. However, no lead compound has been developed to target ZUFSP. OBJECTIVE/METHODS: Therefore, in this study, we used a combined drug repurposing, virtual screening and per-Residue Energy Decomposition (PRED) to identify ZUFSP inhibitors with therapeutic potential. 3-bromo-6-{[4-hydroxy-1-3(3-phenylbutanoyl)piperidin-4-yl]methyl}-4H,5H,6H,7H-thieno[2,3- C]pyridine-7-one (BHPTP) which is an inhibitor of USP7 was repurposed to target ZUFSP. The rationale behind this is based on the similarity of the active between USP7 and ZUFSP. RESULTS: PRED of the binding between BHPTP and ZUFSP revealed Cys223, Arg408, Met410, Asn460, and Tyr465 as the crucial residues responsible for this interaction. The pharmacophoric moieties of BHPTP responsible for this binding along with other physiochemical properties were used as a filter to retrieve potential ligands. 799 compounds were retrieved, ZINC083241427, ZINC063648749, and ZINC063648753 were selected due to the binding energy they exhibited. Cheminformatics analysis revealed that the compounds possess high membrane permeability, however, BHPTP had a low membrane permeability. Furthermore, the compounds are drug like, having obeyed Lipinski's rule of five. CONCLUSION: Taken together, findings from this study put ZINC083241427, ZINC063648749, and ZINC063648753 as potential ZUFSP inhibitor, however, more experimental validation is required to unravel the mechanism of actions of these compounds.


Subject(s)
Catalytic Domain/drug effects , Ubiquitin-Specific Peptidase 7/antagonists & inhibitors , Ubiquitin-Specific Peptidase 7/chemistry , Zinc Fingers/drug effects , Catalytic Domain/physiology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Molecular Docking Simulation/methods , Protein Binding/physiology , Protein Structure, Secondary , Protein Structure, Tertiary , Zinc Fingers/physiology
8.
Biochemistry ; 59(40): 3796-3801, 2020 10 13.
Article in English | MEDLINE | ID: mdl-33006472

ABSTRACT

Ubiquitination and SUMOylation of protein are crucial for various biological responses. The recent unraveling of cross-talk between SUMO and ubiquitin (Ub) has shown the pressing needs to develop the platform for the synthesis of Ub tagged SUMO2 dimers to decipher its biological functions. Still, the platforms for facile synthesis of dimers under native condition are less explored and remain major challenges. Here, we have developed the platform that can expeditiously synthesize all eight Ub tagged SUMO2 and SUMOylated proteins under native condition. Expanding genetic code (EGC) method was employed to incorporate Se-alkylselenocysteine at lysine positions. Oxidative selenoxide elimination generates the electrophilic center, dehydroalanine, which upon Michael addition with C-terminal modified ubiquitin, a nucleophile, yield Ub tagged SUMO2. The dimers were further interrogated with USP7, a SUMO2 deubiquitinase, which is involved in DNA repair, to understand specificity toward the Ub tagged SUMO2 dimer. Our results have shown that the C-terminal domain of USP7 is crucial for USP7 efficiency and selectivity for the Ub tagged SUMO2 dimer.


Subject(s)
Small Ubiquitin-Related Modifier Proteins/metabolism , Ubiquitin-Specific Peptidase 7/metabolism , Ubiquitin/metabolism , Humans , Models, Molecular , Protein Domains , Protein Multimerization , Small Ubiquitin-Related Modifier Proteins/chemistry , Substrate Specificity , Sumoylation , Ubiquitin/chemistry , Ubiquitin-Specific Peptidase 7/chemistry , Ubiquitination
9.
J Biol Chem ; 295(33): 11754-11763, 2020 08 14.
Article in English | MEDLINE | ID: mdl-32587091

ABSTRACT

The transcription factor NF-ĸB is a master regulator of the innate immune response and plays a central role in inflammatory diseases by mediating the expression of pro-inflammatory cytokines. Ubiquitination-triggered proteasomal degradation of DNA-bound NF-ĸB strongly limits the expression of its target genes. Conversely, USP7 (deubiquitinase ubiquitin-specific peptidase 7) opposes the activities of E3 ligases, stabilizes DNA-bound NF-ĸB, and thereby promotes NF-ĸB-mediated transcription. Using gene expression and synthetic peptide arrays on membrane support and overlay analyses, we found here that inhibiting USP7 increases NF-ĸB ubiquitination and degradation, prevents Toll-like receptor-induced pro-inflammatory cytokine expression, and represents an effective strategy for controlling inflammation. However, the broad regulatory roles of USP7 in cell death pathways, chromatin, and DNA damage responses limit the use of catalytic inhibitors of USP7 as anti-inflammatory agents. To this end, we identified an NF-ĸB-binding site in USP7, ubiquitin-like domain 2, that selectively mediates interactions of USP7 with NF-ĸB subunits but is dispensable for interactions with other proteins. Moreover, we found that the amino acids 757LDEL760 in USP7 critically contribute to the interaction with the p65 subunit of NF-ĸB. Our findings support the notion that USP7 activity could be potentially targeted in a substrate-selective manner through the development of noncatalytic inhibitors of this deubiquitinase to abrogate NF-ĸB activity.


Subject(s)
Transcription Factor RelA/metabolism , Ubiquitin-Specific Peptidase 7/metabolism , Ubiquitination , Animals , Cells, Cultured , Female , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Models, Molecular , Protein Domains , Protein Interaction Domains and Motifs , Proteolysis , Ubiquitin-Specific Peptidase 7/chemistry
10.
ACS Chem Biol ; 15(6): 1392-1400, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32302100

ABSTRACT

Ubiquitin specific protease 7 (USP7) regulates the protein stability of key cellular regulators in pathways ranging from apoptosis to neuronal development, making it a promising therapeutic target. Here we used an engineered, activated variant of the USP7 catalytic domain to perform structure-activity studies of electrophilic peptidomimetic inhibitors. Employing this USP7 variant, we found that inhibitors with a cyanopyrrolidine warhead unexpectedly promoted a ß-elimination reaction of the initial covalent adducts, thereby converting the active-site cysteine residue to dehydroalanine. We determined that this phenomenon is specific for the USP7 catalytic cysteine and that structural features of the inhibitor and protein microenvironment impact elimination rates. Using comprehensive docking studies, we propose that the characteristic conformational dynamics of USP7 allow access to conformations that promote the ligand-induced elimination. Unlike in conventional reversible-covalent inhibition, the compounds described here irreversibly destroy a catalytic residue while simultaneously converting the inhibitor to a nonelectrophilic byproduct. Accordingly, this unexpected finding expands the scope of covalent inhibitor modalities and offers intriguing insights into enzyme-inhibitor dynamics.


Subject(s)
Catalytic Domain/drug effects , Pyrrolidines/chemistry , Pyrrolidines/pharmacology , Ubiquitin-Specific Peptidase 7/antagonists & inhibitors , Cysteine/chemistry , Cysteine/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Molecular Docking Simulation , Peptidomimetics/chemistry , Peptidomimetics/pharmacology , Ubiquitin-Specific Peptidase 7/chemistry , Ubiquitin-Specific Peptidase 7/metabolism
11.
J Med Chem ; 63(10): 5398-5420, 2020 05 28.
Article in English | MEDLINE | ID: mdl-32302140

ABSTRACT

USP7 is a promising target for cancer therapy as its inhibition is expected to decrease function of oncogenes, increase tumor suppressor function, and enhance immune function. Using a structure-based drug design strategy, a new class of reversible USP7 inhibitors has been identified that is highly potent in biochemical and cellular assays and extremely selective for USP7 over other deubiquitinases. The succinimide was identified as a key potency-driving motif, forming two strong hydrogen bonds to the allosteric pocket of USP7. Redesign of an initial benzofuran-amide scaffold yielded a simplified ether series of inhibitors, utilizing acyclic conformational control to achieve proper amine placement. Further improvements were realized upon replacing the ether-linked amines with carbon-linked morpholines, a modification motivated by free energy perturbation (FEP+) calculations. This led to the discovery of compound 41, a highly potent, selective, and orally bioavailable USP7 inhibitor. In xenograft studies, compound 41 demonstrated tumor growth inhibition in both p53 wildtype and p53 mutant cancer cell lines, demonstrating that USP7 inhibitors can suppress tumor growth through multiple different pathways.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Drug Discovery/methods , Ubiquitin-Specific Peptidase 7/antagonists & inhibitors , Ubiquitin-Specific Peptidase 7/chemistry , Administration, Oral , Animals , Cell Line, Tumor , Crystallography, X-Ray/methods , Humans , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Protein Structure, Tertiary , Ubiquitin-Specific Peptidase 7/metabolism , Xenograft Model Antitumor Assays/methods
12.
Sci Rep ; 10(1): 5324, 2020 03 24.
Article in English | MEDLINE | ID: mdl-32210275

ABSTRACT

Ubiquitin specific peptidase 7 (USP7) is a deubiquitinating enzyme (DUB) that removes ubiquitin tags from specific protein substrates in order to alter their degradation rate and sub-cellular localization. USP7 has been proposed as a therapeutic target in several cancers because it has many reported substrates with a role in cancer progression, including FOXO4, MDM2, N-Myc, and PTEN. The multi-substrate nature of USP7, combined with the modest potency and selectivity of early generation USP7 inhibitors, has presented a challenge in defining predictors of response to USP7 and potential patient populations that would benefit most from USP7-targeted drugs. Here, we describe the structure-guided development of XL177A, which irreversibly inhibits USP7 with sub-nM potency and selectivity across the human proteome. Evaluation of the cellular effects of XL177A reveals that selective USP7 inhibition suppresses cancer cell growth predominantly through a p53-dependent mechanism: XL177A specifically upregulates p53 transcriptional targets transcriptome-wide, hotspot mutations in TP53 but not any other genes predict response to XL177A across a panel of ~500 cancer cell lines, and TP53 knockout rescues XL177A-mediated growth suppression of TP53 wild-type (WT) cells. Together, these findings suggest TP53 mutational status as a biomarker for response to USP7 inhibition. We find that Ewing sarcoma and malignant rhabdoid tumor (MRT), two pediatric cancers that are sensitive to other p53-dependent cytotoxic drugs, also display increased sensitivity to XL177A.


Subject(s)
Protease Inhibitors/pharmacology , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Specific Peptidase 7/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , MCF-7 Cells , Protease Inhibitors/chemistry , Ubiquitin-Specific Peptidase 7/chemistry , Ubiquitin-Specific Peptidase 7/metabolism , Ubiquitination/drug effects
13.
Proc Natl Acad Sci U S A ; 117(9): 4664-4674, 2020 03 03.
Article in English | MEDLINE | ID: mdl-32071216

ABSTRACT

During protein degradation by the ubiquitin-proteasome pathway, latent 26S proteasomes in the cytosol must assume an active form. Proteasomes are activated when ubiquitylated substrates bind to them and interact with the proteasome-bound deubiquitylase Usp14/Ubp6. The resulting increase in the proteasome's degradative activity was recently shown to be mediated by Usp14's ubiquitin-like (Ubl) domain, which, by itself, can trigger proteasome activation. Many other proteins with diverse cellular functions also contain Ubl domains and can associate with 26S proteasomes. We therefore tested if various Ubl-containing proteins that have important roles in protein homeostasis or disease also activate 26S proteasomes. All seven Ubl-containing proteins tested-the shuttling factors Rad23A, Rad23B, and Ddi2; the deubiquitylase Usp7, the ubiquitin ligase Parkin, the cochaperone Bag6, and the protein phosphatase UBLCP1-stimulated peptide hydrolysis two- to fivefold. Rather than enhancing already active proteasomes, Rad23B and its Ubl domain activated previously latent 26S particles. Also, Ubl-containing proteins (if present with an unfolded protein) increased proteasomal adenosine 5'-triphosphate (ATP) hydrolysis, the step which commits substrates to degradation. Surprisingly, some of these proteins also could stimulate peptide hydrolysis even when their Ubl domains were deleted. However, their Ubl domains were required for the increased ATPase activity. Thus, upon binding to proteasomes, Ubl-containing proteins not only deliver substrates (e.g., the shuttling factors) or provide additional enzymatic activities (e.g., Parkin) to proteasomes, but also increase their capacity for proteolysis.


Subject(s)
Proteasome Endopeptidase Complex/metabolism , Proteolysis , Ubiquitin/metabolism , Binding Sites , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Endopeptidases/chemistry , Endopeptidases/metabolism , Proteasome Endopeptidase Complex/chemistry , Protein Binding , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Ubiquitin/chemistry , Ubiquitin Thiolesterase/chemistry , Ubiquitin Thiolesterase/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Specific Peptidase 7/chemistry , Ubiquitin-Specific Peptidase 7/metabolism
14.
Yakugaku Zasshi ; 139(5): 827-835, 2019 May 01.
Article in Japanese | MEDLINE | ID: mdl-30842349

ABSTRACT

Proteolysis mediated by the ubiquitin-proteome system plays an important role in cancer. Recently, a deubiquitinating enzyme, ubiquitin-specific protease 7 (USP7) has attracted attention as a key regulator of the p53-human double minute 2 (HDM2) pathway in cancer cells. Although some USP7 enzyme inhibitors have been identified, issues related to activity and selectivity prevent their therapeutic application. In this study, we aimed to search for novel USP7-HDM2 protein-protein interaction (PPI) inhibitors that do not affect the USP7 enzyme activity. Using the fragment-mapping program Fsubsite and the canonical subsite-fragment database (CSFDB) developed in our laboratory, we mapped a variety of fragments onto USP7 protein and constructed 3D-pharmacophore models based on the arrangement patterns of the mapped fragments. Finally, we performed 3D pharmacophore-based virtual screening of a commercial compound database and successfully selected promising USP7-HDM2 PPI inhibitor candidates.


Subject(s)
Antineoplastic Agents , Computer Simulation , Drug Discovery , Protease Inhibitors , Protein Interaction Maps , Proto-Oncogene Proteins c-mdm2 , Restriction Mapping/methods , Ubiquitin-Specific Peptidase 7 , Models, Molecular , Protease Inhibitors/chemistry , Protein Structure, Quaternary , Proteolysis , Proto-Oncogene Proteins c-mdm2/chemistry , Ubiquitin-Specific Peptidase 7/chemistry
15.
DNA Repair (Amst) ; 76: 30-39, 2019 04.
Article in English | MEDLINE | ID: mdl-30807924

ABSTRACT

Turnover of cellular proteins is regulated by Ubiquitin Proteasome System (UPS). Components of this pathway, including the proteasome, ubiquitinating enzymes and deubiquitinating enzymes, are highly specialized and tightly regulated. In this mini-review we focus on the de-ubiquitinating enzyme USP7, and summarize latest advances in understanding its structure, substrate specificity and relevance to human cancers. There is increasing interest in UPS components as targets for cancer therapy and here we also overview the recent progress in the development of small molecule inhibitors that target USP7.


Subject(s)
Enzyme Inhibitors/pharmacology , Ubiquitin-Specific Peptidase 7/chemistry , Ubiquitin-Specific Peptidase 7/metabolism , Animals , Humans , Substrate Specificity , Ubiquitin-Specific Peptidase 7/antagonists & inhibitors
16.
Nat Commun ; 10(1): 231, 2019 01 16.
Article in English | MEDLINE | ID: mdl-30651545

ABSTRACT

USP7 is a highly abundant deubiquitinating enzyme (DUB), involved in cellular processes including DNA damage response and apoptosis. USP7 has an unusual catalytic mechanism, where the low intrinsic activity of the catalytic domain (CD) increases when the C-terminal Ubl domains (Ubl45) fold onto the CD, allowing binding of the activating C-terminal tail near the catalytic site. Here we delineate how the target protein promotes the activation of USP7. Using NMR analysis and biochemistry we describe the order of activation steps, showing that ubiquitin binding is an instrumental step in USP7 activation. Using chemically synthesised p53-peptides we also demonstrate how the correct ubiquitinated substrate increases catalytic activity. We then used transient reaction kinetic modelling to define how the USP7 multistep mechanism is driven by target recognition. Our data show how this pleiotropic DUB can gain specificity for its cellular targets.


Subject(s)
Protein Processing, Post-Translational , Ubiquitin-Specific Peptidase 7/metabolism , Ubiquitin/metabolism , Carbon Isotopes/chemistry , Catalytic Domain/genetics , Enzyme Assays/methods , Kinetics , Models, Chemical , Mutagenesis, Site-Directed , Nitrogen Isotopes/chemistry , Nuclear Magnetic Resonance, Biomolecular/methods , Peptides/chemistry , Peptides/metabolism , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Substrate Specificity , Surface Plasmon Resonance , Tumor Suppressor Protein p53/chemistry , Ubiquitin/chemistry , Ubiquitin-Specific Peptidase 7/chemistry , Ubiquitin-Specific Peptidase 7/genetics , Ubiquitin-Specific Peptidase 7/isolation & purification
17.
Cell Res ; 28(12): 1186-1194, 2018 12.
Article in English | MEDLINE | ID: mdl-30254335

ABSTRACT

The ubiquitin system is important for drug discovery, and the discovery of selective small-molecule inhibitors of deubiquitinating enzymes (DUBs) remains an active yet extremely challenging task. With a few exceptions, previously developed inhibitors have been found to bind the evolutionarily conserved catalytic centers of DUBs, resulting in poor selectivity. The small molecule IU1 was the first-ever specific inhibitor identified and exhibited surprisingly excellent selectivity for USP14 over other DUBs. However, the molecular mechanism for this selectivity was elusive. Herein, we report the high-resolution co-crystal structures of the catalytic domain of USP14 bound to IU1 and three IU1 derivatives. All the structures of these complexes indicate that IU1 and its analogs bind to a previously unknown steric binding site in USP14, thus blocking the access of the C-terminus of ubiquitin to the active site of USP14 and abrogating USP14 activity. Importantly, this steric site in USP14 is very unique, as suggested by structural alignments of USP14 with several known DUB X-ray structures. These results, in conjunction with biochemical characterization, indicate a coherent steric blockade mechanism for USP14 inhibition by compounds of the IU series. In light of the recent report of steric blockade of USP7 by FT671, this work suggests a potential generally applicable allosteric mechanism for the regulation of DUBs via steric blockade, as showcased by our discovery of IU1-248 which is 10-fold more potent than IU1.


Subject(s)
Enzyme Inhibitors/chemistry , Pyrroles/chemistry , Pyrrolidines/chemistry , Ubiquitin Thiolesterase/antagonists & inhibitors , Ubiquitin Thiolesterase/chemistry , Allosteric Regulation , Binding Sites , Catalytic Domain , Crystallization/methods , Crystallography, X-Ray/methods , Enzyme Inhibitors/pharmacology , Humans , Models, Molecular , Protein Structure, Tertiary , Pyrroles/pharmacology , Pyrrolidines/pharmacology , Substrate Specificity , Ubiquitin/metabolism , Ubiquitin-Specific Peptidase 7/chemistry
18.
Epigenetics Chromatin ; 11(1): 9, 2018 02 27.
Article in English | MEDLINE | ID: mdl-29482658

ABSTRACT

BACKGROUND: It has been reported that USP7 (ubiquitin-specific protease 7) prevents ubiquitylation and degradation of DNA methyltransferase 1 (DNMT1) by direct binding of USP7 to the glycine-lysine (GK) repeats that join the N-terminal regulatory domain of DNMT1 to the C-terminal methyltransferase domain. The USP7-DNMT1 interaction was reported to be mediated by acetylation of lysine residues within the (GK) repeats. RESULTS: We found that DNMT1 is present at normal levels in mouse and human cells that contain undetectable levels of USP7. Substitution of the (GK) repeats by (GQ) repeats prevents lysine acetylation but does not affect the stability of DNMT1 or the ability of the mutant protein to restore genomic methylation levels when expressed in Dnmt1-null ES cells. Furthermore, both USP7 and PCNA are recruited to sites of DNA replication independently of the presence of DNMT1, and there is no evidence that DNMT1 is degraded in cycling cells after S phase. CONCLUSIONS: Multiple lines of evidence indicate that homeostasis of DNMT1 in somatic cells is controlled primarily at the level of transcription and that interaction of USP7 with the (GK) repeats of DNMT1 is unlikely to play a major role in the stabilization of DNMT1 protein.


Subject(s)
DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA Replication , DNA/genetics , Lysine/chemistry , Ubiquitin-Specific Peptidase 7/metabolism , Acetylation , Animals , Binding Sites , Cell Line , DNA (Cytosine-5-)-Methyltransferase 1/chemistry , Humans , Mice , Mouse Embryonic Stem Cells , Mutation , Protein Binding , Protein Stability , Ubiquitin-Specific Peptidase 7/chemistry
19.
J Med Chem ; 61(2): 422-443, 2018 01 25.
Article in English | MEDLINE | ID: mdl-28768102

ABSTRACT

Ubiquitin specific protease 7 (USP7), the most widely studied among the nearly 100 deubiquitinating enzymes, supports cancer by positively affecting tumor growth and negatively affecting the patient's immune response to tumors. Great interest exists, therefore, in developing USP7 inhibitors for clinical evaluation. While the proteasome inhibitor field has enjoyed clinical success, very few clinically appropriate effectors of deubiquitinating (protease) or ubiquitinating (ligase) enzymes have appeared. The ubiquitin protease/ligase field is moving from the initial discovery of potent, selective modulators with cell proof of concept and in vivo activity to the optimization of these molecules to impart drug-like properties or the discovery of new inhibitor scaffolds by improved screening or rational design. This Perspective focuses on the current status of USP7 inhibitors from various organizations active in developing these compounds for the clinic and suggests undertakings that are both achievable and necessary to lead to successful clinical outcomes for USP7 inhibitors in cancer treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/pharmacology , Ubiquitin-Specific Peptidase 7/antagonists & inhibitors , Ubiquitin-Specific Peptidase 7/metabolism , Antineoplastic Agents/chemistry , Clinical Trials as Topic , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Humans , Molecular Targeted Therapy/methods , Proteasome Inhibitors/pharmacology , Protein Conformation , Ubiquitin/metabolism , Ubiquitin-Specific Peptidase 7/chemistry , Ubiquitin-Specific Peptidase 7/immunology
20.
Structure ; 26(1): 72-84.e7, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29249604

ABSTRACT

Ubiquitin-specific protease 7 (USP7) deubiquitinase activity is controlled by a number of regulatory factors, including stimulation by intramolecular accessory domains. Alone, the USP7 catalytic domain (USP7cd) shows limited activity and apo USP7cd crystal structures reveal a disrupted catalytic triad. By contrast, ubiquitin-conjugated USP7cd structures demonstrate the canonical cysteine protease active-site geometry; however, the structural features of the USP7cd that stabilize the inactive conformation and the mechanism of transition between inactive and active states remain unclear. Here we use comparative structural analyses, molecular dynamics simulations, and in silico sequence re-engineering via directed sampling by RosettaDesign to identify key molecular determinants of USP7cd activation and successfully engineer USP7cd for improved activity. Full kinetic analysis and multiple X-ray crystal structures of our designs indicate that electrostatic interactions in the distal "switching loop" region and local packing in the hydrophobic core mediate subtle but significant conformational changes that modulate USP7cd activation.


Subject(s)
Enzyme Inhibitors/chemistry , Mutation , Peptidomimetics/chemistry , Ubiquitin-Specific Peptidase 7/chemistry , Amino Acid Sequence , Amino Acid Substitution , Catalytic Domain , Cloning, Molecular , Crystallography, X-Ray , Enzyme Activation , Enzyme Inhibitors/chemical synthesis , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Kinetics , Molecular Dynamics Simulation , Peptidomimetics/chemical synthesis , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Static Electricity , Substrate Specificity , Thermodynamics , Ubiquitin-Specific Peptidase 7/antagonists & inhibitors , Ubiquitin-Specific Peptidase 7/genetics , Ubiquitin-Specific Peptidase 7/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...