Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 48
Filtrer
1.
J Am Chem Soc ; 2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38848252

RÉSUMÉ

ß-catenin (CTNNB1) is an oncogenic transcription factor that is important in cell-cell adhesion and transcription of cell proliferation and survival genes that drive the pathogenesis of many different types of cancers. However, direct pharmacological targeting of CTNNB1 has remained challenging. Here, we have performed a screen with a library of cysteine-reactive covalent ligands to identify the monovalent degrader EN83 that depletes CTNNB1 in a ubiquitin-proteasome-dependent manner. We show that EN83 directly and covalently targets CTNNB1 three cysteines C466, C520, and C619, leading to destabilization and degradation of CTNNB1. Through structural optimization, we generate a highly potent and relatively selective destabilizing degrader that acts through the targeting of only C619 on CTNNB1. Our results show that chemoproteomic approaches can be used to covalently target and degrade challenging transcription factors like CTNNB1 through destabilization-mediated degradation.

2.
ACS Chem Biol ; 19(2): 442-450, 2024 02 16.
Article de Anglais | MEDLINE | ID: mdl-38305738

RÉSUMÉ

Targeted protein degradation with proteolysis targeting chimeras (PROTACs) is a powerful therapeutic modality for eliminating disease-causing proteins through targeted ubiquitination and proteasome-mediated degradation. Most PROTACs have exploited substrate receptors of Cullin-RING E3 ubiquitin ligases such as cereblon and VHL. Whether core, shared, and essential components of the Cullin-RING E3 ubiquitin ligase complex can be used for PROTAC applications remains less explored. Here, we discovered a cysteine-reactive covalent recruiter EN884 against the SKP1 adapter protein of the SKP1-CUL1-F-box containing the SCF complex. We further showed that this recruiter can be used in PROTAC applications to degrade neo-substrate proteins such as BRD4 and the androgen receptor in a SKP1- and proteasome-dependent manner. Our studies demonstrate that core and essential adapter proteins within the Cullin-RING E3 ubiquitin ligase complex can be exploited for targeted protein degradation applications and that covalent chemoproteomic strategies can enable recruiter discovery against these targets.


Sujet(s)
Cullines , Ubiquitin-protein ligases , Ubiquitin-protein ligases/métabolisme , Cullines/métabolisme , Protéolyse , Proteasome endopeptidase complex/métabolisme , Protéines nucléaires/métabolisme , Facteurs de transcription/métabolisme , Protéines associées aux kinases de la phase S/métabolisme , Protéines adaptatrices de la transduction du signal/métabolisme
3.
ACS Chem Biol ; 19(1): 22-36, 2024 01 19.
Article de Anglais | MEDLINE | ID: mdl-38150587

RÉSUMÉ

The papain-like protease of SARS-COV-2 is essential for viral replication and pathogenesis. Its location within a much larger multifunctional protein, NSP3, makes it an ideal candidate for a targeted degradation approach capable of eliminating multiple functions with a single-molecule treatment. In this work, we have developed a HiBiT-based cellular model to study NSP3 degradation and used this platform for the discovery of monovalent NSP3 degraders. We present previously unreported degradation activity of published papain-like protease inhibitors. Follow-up exploration of structure-activity relationships and mechanism-of-action studies points to the recruitment of the ubiquitin-proteasome machinery that is solely driven by site occupancy, regardless of molecular features of the ligand. Supported by HDX data, we hypothesize that binding-induced structural changes in NSP3 trigger the recruitment of an E3 ligase and lead to proteasomal degradation.


Sujet(s)
COVID-19 , Protéases de type papaïne des coronavirus , Papaïne , Humains , Papaïne/métabolisme , Protéines virales non structurales/métabolisme , SARS-CoV-2/composition chimique , Inhibiteurs de protéases/métabolisme
4.
bioRxiv ; 2023 Nov 02.
Article de Anglais | MEDLINE | ID: mdl-37961622

RÉSUMÉ

ß-catenin (CTNNB1) is an oncogenic transcription factor that is important in cell-cell adhesion and transcription of cell proliferation and survival genes that drives the pathogenesis of many different types of cancers. However, direct pharmacological targeting of CTNNB1 has remained challenging deeming this transcription factor as "undruggable." Here, we have performed a screen with a library of cysteine-reactive covalent ligands to identify a monovalent degrader EN83 that depletes CTNNB1 in a ubiquitin-proteasome-dependent manner. We show that EN83 directly and covalently targets CTNNB1 through targeting four distinct cysteines within the armadillo repeat domain-C439, C466, C520, and C619-leading to a destabilization of CTNNB1. Using covalent chemoproteomic approaches, we show that EN83 directly engages CTNNB1 in cells with a moderate degree of selectivity. We further demonstrate that direct covalent targeting of three of these four cysteines--C466, C520, and C619--in cells contributes to CTNNB1 degradation in cells. We also demonstrate that EN83 can be further optimized to yield more potent CTNNB1 binders and degraders. Our results show that chemoproteomic approaches can be used to covalently target and degrade challenging transcription factors like CTNNB1 through a destabilization-mediated degradation.

5.
bioRxiv ; 2023 Nov 02.
Article de Anglais | MEDLINE | ID: mdl-37904950

RÉSUMÉ

Targeted protein degradation with Proteolysis Targeting Chimeras (PROTACs) is a powerful therapeutic modality for eliminating disease-causing proteins through targeted ubiquitination and proteasome-mediated degradation. Most PROTACs have exploited substrate receptors of Cullin-RING E3 ubiquitin ligases such as cereblon and VHL. Whether core, shared, and essential components of the Cullin-RING E3 ubiquitin ligase complex can be used for PROTAC applications remains less explored. Here, we discovered a cysteine-reactive covalent recruiter EN884 against the SKP1 adapter protein of the SKP1-CUL1-F-box containing SCF complex. We further showed that this recruiter can be used in PROTAC applications to degrade neo-substrate proteins such as BRD4 and the androgen receptor in a SKP1- and proteasome-dependent manner. Our studies demonstrate that core and essential adapter proteins within the Cullin-RING E3 ubiquitin ligase complex can be exploited for targeted protein degradation applications and that covalent chemoproteomic strategies can enable recruiter discovery against these targets.

6.
Am J Hematol ; 98(3): 449-463, 2023 03.
Article de Anglais | MEDLINE | ID: mdl-36594167

RÉSUMÉ

The treatment of patients with relapsed or refractory lymphoid neoplasms represents a significant clinical challenge. Here, we identify the pro-survival BCL-2 protein family member MCL-1 as a resistance factor for the BCL-2 inhibitor venetoclax in non-Hodgkin lymphoma (NHL) cell lines and primary NHL samples. Mechanistically, we show that the antibody-drug conjugate polatuzumab vedotin promotes MCL-1 degradation via the ubiquitin/proteasome system. This targeted MCL-1 antagonism, when combined with venetoclax and the anti-CD20 antibodies obinutuzumab or rituximab, results in tumor regressions in preclinical NHL models, which are sustained even off-treatment. In a Phase Ib clinical trial (NCT02611323) of heavily pre-treated patients with relapsed or refractory NHL, 25/33 (76%) patients with follicular lymphoma and 5/17 (29%) patients with diffuse large B-cell lymphoma achieved complete or partial responses with an acceptable safety profile when treated with the recommended Phase II dose of polatuzumab vedotin in combination with venetoclax and an anti-CD20 antibody.


Sujet(s)
Immunoconjugués , Lymphome malin non hodgkinien , Humains , Protéine Mcl-1/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Lymphome malin non hodgkinien/traitement médicamenteux , Lymphome malin non hodgkinien/anatomopathologie , Rituximab/usage thérapeutique , Immunoconjugués/usage thérapeutique
7.
Nat Chem Biol ; 19(1): 55-63, 2023 01.
Article de Anglais | MEDLINE | ID: mdl-36577875

RÉSUMÉ

Engineered destruction of target proteins by recruitment to the cell's degradation machinery has emerged as a promising strategy in drug discovery. The majority of molecules that facilitate targeted degradation do so via a select number of ubiquitin ligases, restricting this therapeutic approach to tissue types that express the requisite ligase. Here, we describe a new strategy of targeted protein degradation through direct substrate recruitment to the 26S proteasome. The proteolytic complex is essential and abundantly expressed in all cells; however, proteasomal ligands remain scarce. We identify potent peptidic macrocycles that bind directly to the 26S proteasome subunit PSMD2, with a 2.5-Å-resolution cryo-electron microscopy complex structure revealing a binding site near the 26S pore. Conjugation of this macrocycle to a potent BRD4 ligand enabled generation of chimeric molecules that effectively degrade BRD4 in cells, thus demonstrating that degradation via direct proteasomal recruitment is a viable strategy for targeted protein degradation.


Sujet(s)
Protéines nucléaires , Facteurs de transcription , Protéines nucléaires/métabolisme , Cryomicroscopie électronique , Facteurs de transcription/métabolisme , Proteasome endopeptidase complex/métabolisme , Protéolyse , Ligases/métabolisme , Ubiquitin-protein ligases/métabolisme
8.
Science ; 378(6619): 549-553, 2022 11 04.
Article de Anglais | MEDLINE | ID: mdl-36378961

RÉSUMÉ

Cereblon (CRBN) is a ubiquitin ligase (E3) substrate receptor protein co-opted by CRBN E3 ligase modulatory drug (CELMoD) agents that target therapeutically relevant proteins for degradation. Prior crystallographic studies defined the drug-binding site within CRBN's thalidomide-binding domain (TBD), but the allostery of drug-induced neosubstrate binding remains unclear. We performed cryo-electron microscopy analyses of the DNA damage-binding protein 1 (DDB1)-CRBN apo complex and compared these structures with DDB1-CRBN in the presence of CELMoD compounds alone and complexed with neosubstrates. Association of CELMoD compounds to the TBD is necessary and sufficient for triggering CRBN allosteric rearrangement from an open conformation to the canonical closed conformation. The neosubstrate Ikaros only stably associates with the closed CRBN conformation, illustrating the importance of allostery for CELMoD compound efficacy and informing structure-guided design strategies to improve therapeutic efficacy.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Ubiquitin-protein ligases , Protéines adaptatrices de la transduction du signal/composition chimique , Cryomicroscopie électronique , Thalidomide/composition chimique , Ubiquitin-protein ligases/composition chimique , Domaines protéiques , Régulation allostérique
9.
Ann N Y Acad Sci ; 1510(1): 79-99, 2022 04.
Article de Anglais | MEDLINE | ID: mdl-35000205

RÉSUMÉ

Targeted protein degradation is critical for proper cellular function and development. Protein degradation pathways, such as the ubiquitin proteasomes system, autophagy, and endosome-lysosome pathway, must be tightly regulated to ensure proper elimination of misfolded and aggregated proteins and regulate changing protein levels during cellular differentiation, while ensuring that normal proteins remain unscathed. Protein degradation pathways have also garnered interest as a means to selectively eliminate target proteins that may be difficult to inhibit via other mechanisms. On June 7 and 8, 2021, several experts in protein degradation pathways met virtually for the Keystone eSymposium "Targeting protein degradation: from small molecules to complex organelles." The event brought together researchers working in different protein degradation pathways in an effort to begin to develop a holistic, integrated vision of protein degradation that incorporates all the major pathways to understand how changes in them can lead to disease pathology and, alternatively, how they can be leveraged for novel therapeutics.


Sujet(s)
Proteasome endopeptidase complex , Ubiquitine , Autophagie/physiologie , Humains , Organites , Proteasome endopeptidase complex/métabolisme , Protéines/métabolisme , Protéolyse , Ubiquitine/métabolisme
10.
Nat Biotechnol ; 40(1): 86-93, 2022 01.
Article de Anglais | MEDLINE | ID: mdl-34531539

RÉSUMÉ

Genetic and non-genetic heterogeneity within cancer cell populations represent major challenges to anticancer therapies. We currently lack robust methods to determine how preexisting and adaptive features affect cellular responses to therapies. Here, by conducting clonal fitness mapping and transcriptional characterization using expressed barcodes and single-cell RNA sequencing (scRNA-seq), we have developed tracking differential clonal response by scRNA-seq (TraCe-seq). TraCe-seq is a method that captures at clonal resolution the origin, fate and differential early adaptive transcriptional programs of cells in a complex population in response to distinct treatments. We used TraCe-seq to benchmark how next-generation dual epidermal growth factor receptor (EGFR) inhibitor-degraders compare to standard EGFR kinase inhibitors in EGFR-mutant lung cancer cells. We identified a loss of antigrowth activity associated with targeted degradation of EGFR protein and an essential role of the endoplasmic reticulum (ER) protein processing pathway in anti-EGFR therapeutic efficacy. Our results suggest that targeted degradation is not always superior to enzymatic inhibition and establish TraCe-seq as an approach to study how preexisting transcriptional programs affect treatment responses.


Sujet(s)
Antinéoplasiques , Tumeurs du poumon , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Récepteurs ErbB/génétique , Récepteurs ErbB/métabolisme , Humains , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Analyse sur cellule unique/méthodes
11.
J Med Chem ; 64(16): 11841-11856, 2021 08 26.
Article de Anglais | MEDLINE | ID: mdl-34251202

RÉSUMÉ

Breast cancer remains a leading cause of cancer death in women, representing a significant unmet medical need. Here, we disclose our discovery efforts culminating in a clinical candidate, 35 (GDC-9545 or giredestrant). 35 is an efficient and potent selective estrogen receptor degrader (SERD) and a full antagonist, which translates into better antiproliferation activity than known SERDs (1, 6, 7, and 9) across multiple cell lines. Fine-tuning the physiochemical properties enabled once daily oral dosing of 35 in preclinical species and humans. 35 exhibits low drug-drug interaction liability and demonstrates excellent in vitro and in vivo safety profiles. At low doses, 35 induces tumor regressions either as a single agent or in combination with a CDK4/6 inhibitor in an ESR1Y537S mutant PDX or a wild-type ERα tumor model. Currently, 35 is being evaluated in Phase III clinical trials.


Sujet(s)
Antinéoplasiques/usage thérapeutique , Tumeurs du sein/traitement médicamenteux , Carbolines/usage thérapeutique , Antagonistes des récepteurs des oestrogènes/usage thérapeutique , Récepteur alpha des oestrogènes/métabolisme , Animaux , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacocinétique , Carbolines/composition chimique , Carbolines/pharmacocinétique , Chiens , Antagonistes des récepteurs des oestrogènes/composition chimique , Antagonistes des récepteurs des oestrogènes/pharmacocinétique , Femelle , Humains , Cellules MCF-7 , Macaca fascicularis , Souris , Structure moléculaire , Rats , Relation structure-activité , Tests d'activité antitumorale sur modèle de xénogreffe
12.
Nat Commun ; 12(1): 4608, 2021 07 29.
Article de Anglais | MEDLINE | ID: mdl-34326324

RÉSUMÉ

The ubiquitin conjugating enzyme UBE2W catalyzes non-canonical ubiquitination on the N-termini of proteins, although its substrate repertoire remains unclear. To identify endogenous N-terminally-ubiquitinated substrates, we discover four monoclonal antibodies that selectively recognize tryptic peptides with an N-terminal diglycine remnant, corresponding to sites of N-terminal ubiquitination. Importantly, these antibodies do not recognize isopeptide-linked diglycine (ubiquitin) modifications on lysine. We solve the structure of one such antibody bound to a Gly-Gly-Met peptide to reveal the molecular basis for its selective recognition. We use these antibodies in conjunction with mass spectrometry proteomics to map N-terminal ubiquitination sites on endogenous substrates of UBE2W. These substrates include UCHL1 and UCHL5, where N-terminal ubiquitination distinctly alters deubiquitinase (DUB) activity. This work describes an antibody toolkit for enrichment and global profiling of endogenous N-terminal ubiquitination sites, while revealing functionally relevant substrates of UBE2W.


Sujet(s)
Anticorps/composition chimique , Peptides/composition chimique , Ubiquitin-conjugating enzymes/métabolisme , Protéines ubiquitinées/métabolisme , Séquence d'acides aminés , Animaux , Anticorps/immunologie , Cellules cultivées , Cristallographie aux rayons X/méthodes , Humains , Spectrométrie de masse/méthodes , Liaison aux protéines , Protéomique/méthodes , Lapins , Ubiquitin-conjugating enzymes/composition chimique , Ubiquitin-conjugating enzymes/immunologie , Ubiquitination
14.
Cell Death Differ ; 28(2): 591-605, 2021 02.
Article de Anglais | MEDLINE | ID: mdl-33432113

RÉSUMÉ

The ubiquitin system is complex, multifaceted, and is crucial for the modulation of a vast number of cellular processes. Ubiquitination is tightly regulated at different levels by a range of enzymes including E1s, E2s, and E3s, and an array of DUBs. The UPS directs protein degradation through the proteasome, and regulates a wide array of cellular processes including transcription and epigenetic factors as well as key oncoproteins. Ubiquitination is key to the dynamic regulation of programmed cell death. Notably, the TNF signaling pathway is controlled by competing ubiquitin conjugation and deubiquitination, which governs both proteasomal degradation and signaling complex formation. In the inflammatory response, ubiquitination is capable of both activating and dampening inflammasome activation through the control of either protein stability, complex formation, or, in some cases, directly affecting receptor activity. In this review, we discuss the enzymes and targets in the ubiquitin system that regulate fundamental cellular processes regulating cell death, and inflammation, as well as disease consequences resulting from their dysregulation. Finally, we highlight several pre-clinical and clinical compounds that regulate ubiquitin system enzymes, with the aim of restoring homeostasis and ameliorating diseases.


Sujet(s)
Inflammation/métabolisme , Tumeurs/métabolisme , Ubiquitin-protein ligases/métabolisme , Ubiquitination , Animaux , Apoptose , Humains , Proteasome endopeptidase complex/métabolisme , Transduction du signal , Ubiquitine/métabolisme
15.
Mol Cell ; 77(5): 1092-1106.e9, 2020 03 05.
Article de Anglais | MEDLINE | ID: mdl-31973889

RÉSUMÉ

Co-opting Cullin4 RING ubiquitin ligases (CRL4s) to inducibly degrade pathogenic proteins is emerging as a promising therapeutic strategy. Despite intense efforts to rationally design degrader molecules that co-opt CRL4s, much about the organization and regulation of these ligases remains elusive. Here, we establish protein interaction kinetics and estimation of stoichiometries (PIKES) analysis, a systematic proteomic profiling platform that integrates cellular engineering, affinity purification, chemical stabilization, and quantitative mass spectrometry to investigate the dynamics of interchangeable multiprotein complexes. Using PIKES, we show that ligase assemblies of Cullin4 with individual substrate receptors differ in abundance by up to 200-fold and that Cand1/2 act as substrate receptor exchange factors. Furthermore, degrader molecules can induce the assembly of their cognate CRL4, and higher expression of the associated substrate receptor enhances degrader potency. Beyond the CRL4 network, we show how PIKES can reveal systems level biochemistry for cellular protein networks important to drug development.


Sujet(s)
Chromatographie en phase liquide à haute performance , Protéomique/méthodes , Spectrométrie de masse ESI , Spectrométrie de masse en tandem , Ubiquitin-protein ligases/métabolisme , Cullines/génétique , Cullines/métabolisme , Cellules HEK293 , Humains , Cinétique , Protéines du muscle/génétique , Protéines du muscle/métabolisme , Protéine NEDD8/génétique , Protéine NEDD8/métabolisme , Cartes d'interactions protéiques , Protéolyse , Transduction du signal , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Ubiquitin-protein ligases/génétique
16.
Bioorg Med Chem Lett ; 30(4): 126907, 2020 02 15.
Article de Anglais | MEDLINE | ID: mdl-31902710

RÉSUMÉ

Chimeric molecules which effect intracellular degradation of target proteins via E3 ligase-mediated ubiquitination (e.g., PROTACs) are currently of high interest in medicinal chemistry. However, these entities are relatively large compounds that often possess molecular characteristics which may compromise oral bioavailability, solubility, and/or in vivo pharmacokinetic properties. Accordingly, we explored whether conjugation of chimeric degraders to monoclonal antibodies using technologies originally developed for cytotoxic payloads might provide alternate delivery options for these novel agents. In this report we describe the construction of several degrader-antibody conjugates comprised of two distinct ERα-targeting degrader entities and three independent ADC linker modalities. We subsequently demonstrate the antigen-dependent delivery to MCF7-neo/HER2 cells of the degrader payloads that are incorporated into these conjugates. We also provide evidence for efficient intracellular degrader release from one of the employed linkers. In addition, preliminary data are described which suggest that reasonably favorable in vivo stability properties are associated with the linkers utilized to construct the degrader conjugates.


Sujet(s)
Anticorps monoclonaux/immunologie , Vecteurs de médicaments/composition chimique , Récepteur alpha des oestrogènes/immunologie , Anticorps monoclonaux/composition chimique , Antinéoplasiques/composition chimique , Antinéoplasiques/immunologie , Antinéoplasiques/pharmacologie , Conception de médicament , Récepteur alpha des oestrogènes/métabolisme , Humains , Immunoconjugués/composition chimique , Immunoconjugués/immunologie , Immunoconjugués/pharmacologie , Cellules MCF-7 , Protéolyse/effets des médicaments et des substances chimiques , Récepteur ErbB-2/métabolisme
17.
Cell ; 178(4): 949-963.e18, 2019 08 08.
Article de Anglais | MEDLINE | ID: mdl-31353221

RÉSUMÉ

Estrogen receptor-positive (ER+) breast cancers frequently remain dependent on ER signaling even after acquiring resistance to endocrine agents, prompting the development of optimized ER antagonists. Fulvestrant is unique among approved ER therapeutics due to its capacity for full ER antagonism, thought to be achieved through ER degradation. The clinical potential of fulvestrant is limited by poor physicochemical features, spurring attempts to generate ER degraders with improved drug-like properties. We show that optimization of ER degradation does not guarantee full ER antagonism in breast cancer cells; ER "degraders" exhibit a spectrum of transcriptional activities and anti-proliferative potential. Mechanistically, we find that fulvestrant-like antagonists suppress ER transcriptional activity not by ER elimination, but by markedly slowing the intra-nuclear mobility of ER. Increased ER turnover occurs as a consequence of ER immobilization. These findings provide proof-of-concept that small molecule perturbation of transcription factor mobility may enable therapeutic targeting of this challenging target class.


Sujet(s)
Tumeurs du sein/métabolisme , Antagonistes des récepteurs des oestrogènes/pharmacologie , Fulvestrant/pharmacologie , Récepteurs des oestrogènes/antagonistes et inhibiteurs , Récepteurs des oestrogènes/métabolisme , Animaux , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cinnamates/pharmacologie , Résistance aux médicaments antinéoplasiques , Antagonistes des récepteurs des oestrogènes/usage thérapeutique , Femelle , Fulvestrant/usage thérapeutique , Cellules HEK293 , Hétérogreffes , Humains , Indazoles/pharmacologie , Ligands , Cellules MCF-7 , Souris , Souris de lignée NOD , Souris nude , Souris SCID , Polymorphisme de nucléotide simple , Protéolyse/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Transcription génétique/effets des médicaments et des substances chimiques
18.
Drug Discov Today Technol ; 31: 109-123, 2019 Apr.
Article de Anglais | MEDLINE | ID: mdl-31200854

RÉSUMÉ

The Ubiquitin/Proteasome System comprises an essential cellular mechanism for regulated protein degradation. Ubiquitination may also promote the assembly of protein complexes that initiate intracellular signaling cascades. Thus, proper regulation of substrate protein ubiquitination is essential for maintaining normal cellular physiology. Deubiquitinases are the class of enzymes responsible for removing ubiquitin modifications from target proteins and have been implicated in regulating human disease. As such, deubiquitinases are now recognized as emerging drug targets. Small molecule deubiquitinase inhibitors have been developed; among those, inhibitors for the deubiquitinases USP7 and USP14 are the best-characterized given that they are structurally validated. In this review we discuss the normal physiological roles of the USP7 and USP14 deubiquitinases as well as the pathological conditions associated with their dysfunction, with a focus on oncology and neurodegenerative diseases. We also review structural biology of USP7 and USP14 enzymes and the characterization of their respective inhibitors, highlighting the various molecular mechanisms by which these deubiquitinases may be functionally inhibited. Finally, we summarize the cellular and in vivo studies performed using the structurally-validated USP7 and USP14 inhibitors.


Sujet(s)
Pyrroles/pharmacologie , Ubiquitin thiolesterase/antagonistes et inhibiteurs , Animaux , Humains , Structure moléculaire , Pyrroles/composition chimique , Ubiquitin thiolesterase/composition chimique , Ubiquitin thiolesterase/métabolisme
20.
Cell Chem Biol ; 26(2): 156-177, 2019 02 21.
Article de Anglais | MEDLINE | ID: mdl-30554913

RÉSUMÉ

The ubiquitin/proteasome system is a primary conduit for selective intracellular protein degradation. Since its discovery over 30 years ago, this highly regulated system continues to be an active research area for drug discovery that is exemplified by several approved drugs. Here we review compounds in preclinical testing, clinical trials, and approved drugs, with the aim of highlighting innovative discoveries and breakthrough therapies that target the ubiquitin system.


Sujet(s)
Antienzymes/composition chimique , Proteasome endopeptidase complex/métabolisme , Ubiquitine/métabolisme , Évaluation préclinique de médicament , Antienzymes/métabolisme , Humains , Protéines proto-oncogènes c-mdm2/antagonistes et inhibiteurs , Protéines proto-oncogènes c-mdm2/métabolisme , Ubiquitine/composition chimique , Ubiquitin-activating enzymes/composition chimique , Ubiquitin-activating enzymes/métabolisme , Ubiquitin-conjugating enzymes/composition chimique , Ubiquitin-conjugating enzymes/métabolisme , Ubiquitin-protein ligases/composition chimique , Ubiquitin-protein ligases/métabolisme , Ubiquitination
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...