Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 86
Filtrar
1.
Antiviral Res ; 228: 105944, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38914283

RESUMEN

SARS-CoV-2 papain-like protease (PLpro) could facilitate viral replication and host immune evasion by respectively hydrolyzing viral polyprotein and host ubiquitin conjugates, thereby rendering itself as an important antiviral target. Yet few noncovalent PLpro inhibitors of SARS-CoV-2 have been reported with improved directed towards pathogenic deubiquitinating activities inhibition. Herein, we report that coronavirus PLpro proteases have distinctive substrate bias and are conserved to deubiquitylate K63-linked polyubiquitination, thereby attenuating host type I interferon response. We identify a noncovalent compound specifically optimized towards halting the K63-deubiquitinase activity of SARS-CoV-2 PLpro, but not other coronavirus (CoV) counterparts or host deubiquitinase. Contrasting with GRL-0617, a SARS-CoV-1 PLpro inhibitor, SIMM-036 is 50-fold and 7-fold (half maximal inhibitory concentration (IC50)) more potent to inhibit viral replication during SARS-CoV-2 infection and restore the host interferon-ß (IFN-ß) response in human angiotensin-converting enzyme 2 (hACE2)-HeLa cells, respectively. Structure-activity relationship (SAR) analysis further reveals the importance of BL2 groove of PLpro, which could determine the selectivity of K63-deubiquitinase activity of the enzyme.


Asunto(s)
Antivirales , SARS-CoV-2 , Replicación Viral , Humanos , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/enzimología , Replicación Viral/efectos de los fármacos , Antivirales/farmacología , Antivirales/química , Proteasas Similares a la Papaína de Coronavirus/antagonistas & inhibidores , Proteasas Similares a la Papaína de Coronavirus/metabolismo , Proteasas Similares a la Papaína de Coronavirus/química , Proteasas 3C de Coronavirus/antagonistas & inhibidores , Proteasas 3C de Coronavirus/metabolismo , Proteasas 3C de Coronavirus/química , COVID-19/virología , Enzimas Desubicuitinizantes/antagonistas & inhibidores , Enzimas Desubicuitinizantes/metabolismo , Ubiquitinación/efectos de los fármacos , Tratamiento Farmacológico de COVID-19 , Células Vero , Chlorocebus aethiops , Inhibidores de Proteasas/farmacología , Inhibidores de Proteasas/química , Animales , Células HEK293
2.
J Med Chem ; 67(13): 10710-10742, 2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-38897928

RESUMEN

Upon infection by an intracellular pathogen, host cells activate apoptotic pathways to limit pathogen replication. Consequently, efficient proliferation of the obligate intracellular pathogen Chlamydia trachomatis, a major cause of trachoma and sexually transmitted diseases, depends on the suppression of host cell apoptosis. C. trachomatis secretes deubiquitinase ChlaDUB1 into the host cell, leading among other interactions to the stabilization of antiapoptotic proteins and, thus, suppression of host cell apoptosis. Targeting the bacterial effector protein may, therefore, lead to new therapeutic possibilities. To explore the active site of ChlaDUB1, an iterative cycle of computational docking, synthesis, and enzymatic screening was applied with the aim of lead structure development. Hereby, covalent inhibitors were developed, which show enhanced inhibition with a 22-fold increase in IC50 values compared to previous work. Comprehensive insights into the binding prerequisites to ChlaDUB1 are provided, establishing the foundation for an additional specific antichlamydial therapy by small molecules.


Asunto(s)
Chlamydia trachomatis , Diseño de Fármacos , Chlamydia trachomatis/efectos de los fármacos , Chlamydia trachomatis/enzimología , Relación Estructura-Actividad , Simulación del Acoplamiento Molecular , Acetiltransferasas/antagonistas & inhibidores , Acetiltransferasas/metabolismo , Humanos , Antibacterianos/farmacología , Antibacterianos/síntesis química , Antibacterianos/química , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Enzimas Desubicuitinizantes/antagonistas & inhibidores , Enzimas Desubicuitinizantes/metabolismo , Estructura Molecular , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/metabolismo
3.
Virus Res ; 344: 199368, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38588924

RESUMEN

Several viruses are now known to code for deubiquitinating proteases in their genomes. Ubiquitination is an essential post-translational modification of cellular substrates involved in many processes in the cell, including in innate immune signalling. This post-translational modification is regulated by the ubiquitin conjugation machinery, as well as various host deubiquitinating enzymes. The conjugation of ubiquitin chains to several innate immune related factors is often needed to induce downstream signalling, shaping the antiviral response. Viral deubiquitinating proteins, besides often having a primary function in the viral replication cycle by cleaving the viral polyprotein, are also able to cleave ubiquitin chains from such host substrates, in that way exerting a function in innate immune evasion. The presence of viral deubiquitinating enzymes has been firmly established for numerous animal-infecting viruses, such as some well-researched and clinically important nidoviruses, and their presence has now been confirmed in several plant viruses as well. Viral proteases in general have long been highlighted as promising drug targets, with a current focus on small molecule inhibitors. In this review, we will discuss the range of viral deubiquitinating proteases known to date, summarise the various avenues explored to inhibit such proteases and discuss novel strategies and models intended to inhibit and study these specific viral enzymes.


Asunto(s)
Enzimas Desubicuitinizantes , Enzimas Desubicuitinizantes/metabolismo , Enzimas Desubicuitinizantes/antagonistas & inhibidores , Enzimas Desubicuitinizantes/genética , Humanos , Proteasas Virales/metabolismo , Procesamiento Proteico-Postraduccional , Ubiquitinación , Animales , Replicación Viral , Antivirales/farmacología , Inhibidores de Proteasas/farmacología , Virus/efectos de los fármacos , Virus/enzimología , Proteínas Virales/metabolismo , Proteínas Virales/genética , Ubiquitina/metabolismo , Inmunidad Innata
4.
J Mol Biol ; 435(23): 168316, 2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-37858708

RESUMEN

Deubiquitinases (DUBs) are proteolytic enzymes that catalyze the removal of ubiquitin from protein substrates. The critical role of DUBs in regulating protein ubiquitination makes them attractive drug targets in oncology, neurodegenerative disease, and antiviral development. Biochemical assays for quantifying DUB activity have enabled characterization of substrate preferences and discovery of small molecule inhibitors. However, assessing the efficacy of these inhibitors in cellular contexts to support clinical drug development has been limited by a lack of tractable cell-based assays. To address this gap, we developed a two-color flow cytometry-based assay that allows for sensitive quantification of DUB activity and inhibition in living cells. The utility of this system was demonstrated by quantifying the potency of GRL0617 against the viral DUB SARS-CoV-2 PLpro, identifying potential GRL0617 resistance mutations, and performing structure-function analysis of the vOTU domain from the recently emerged Yezo virus. In addition, the system was optimized for cellular DUBs by modifying a GFP-targeting nanobody to recruit USP7 and USP28 to benchmark a panel of reported inhibitors and assess inhibition kinetics. Together, these results demonstrate the utility of these assays for studying DUB biology in a cellular context with potential to aid in inhibitor discovery and development.


Asunto(s)
Enzimas Desubicuitinizantes , Citometría de Flujo , Inhibidores de Proteasas , Humanos , Compuestos de Anilina/farmacología , Benzamidas/farmacología , Enzimas Desubicuitinizantes/análisis , Enzimas Desubicuitinizantes/antagonistas & inhibidores , Enfermedades Neurodegenerativas/enzimología , Ubiquitina/metabolismo , Ubiquitina Tiolesterasa/antagonistas & inhibidores , Peptidasa Específica de Ubiquitina 7/antagonistas & inhibidores , Ubiquitinación/efectos de los fármacos , Citometría de Flujo/métodos , Inhibidores de Proteasas/química , Inhibidores de Proteasas/farmacología , Proteasas Similares a la Papaína de Coronavirus/análisis , Proteasas Similares a la Papaína de Coronavirus/antagonistas & inhibidores , Anticuerpos de Dominio Único
5.
Bioorg Med Chem Lett ; 94: 129458, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37634761

RESUMEN

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


Asunto(s)
Antimaláricos , Enzimas Desubicuitinizantes , Antagonistas del Ácido Fólico , Antimaláricos/farmacología , Eucariontes , Plasmodium falciparum , Complejo de la Endopetidasa Proteasomal , Enzimas Desubicuitinizantes/antagonistas & inhibidores , Enzimas Desubicuitinizantes/química , Proteínas Protozoarias
6.
Mol Cell Proteomics ; 22(8): 100609, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37385347

RESUMEN

Dampening functional levels of the mitochondrial deubiquitylating enzyme Ubiquitin-specific protease 30 (USP30) has been suggested as an effective therapeutic strategy against neurodegenerative disorders such as Parkinson's Disease. USP30 inhibition may counteract the deleterious effects of impaired turnover of damaged mitochondria, which is inherent to both familial and sporadic forms of the disease. Small-molecule inhibitors targeting USP30 are currently in development, but little is known about their precise nature of binding to the protein. We have integrated biochemical and structural approaches to gain novel mechanistic insights into USP30 inhibition by a small-molecule benzosulfonamide-containing compound, USP30inh. Activity-based protein profiling mass spectrometry confirmed target engagement, high selectivity, and potency of USP30inh for USP30 against 49 other deubiquitylating enzymes in a neuroblastoma cell line. In vitro characterization of USP30inh enzyme kinetics inferred slow and tight binding behavior, which is comparable with features of covalent modification of USP30. Finally, we blended hydrogen-deuterium exchange mass spectrometry and computational docking to elucidate the molecular architecture and geometry of USP30 complex formation with USP30inh, identifying structural rearrangements at the cleft of the USP30 thumb and palm subdomains. These studies suggest that USP30inh binds to this thumb-palm cleft, which guides the ubiquitin C terminus into the active site, thereby preventing ubiquitin binding and isopeptide bond cleavage, and confirming its importance in the inhibitory process. Our data will pave the way for the design and development of next-generation inhibitors targeting USP30 and associated deubiquitinylases.


Asunto(s)
Enzimas Desubicuitinizantes , Mitofagia , Enzimas Desubicuitinizantes/antagonistas & inhibidores , Enzimas Desubicuitinizantes/metabolismo , Proteínas Mitocondriales/metabolismo , Mitofagia/fisiología , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Sulfonamidas/farmacología
7.
Viruses ; 15(2)2023 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-36851696

RESUMEN

Ubiquitination and deubiquitination processes are widely involved in modulating the function, activity, localization, and stability of multiple cellular proteins regulating almost every aspect of cellular function. Several virus families have been shown to exploit the cellular ubiquitin-conjugating system to achieve a productive infection: enter the cell, promote genome replication, or assemble and release viral progeny. In this study, we analyzed the role of deubiquitinating enzymes (DUBs) during chikungunya virus (CHIKV) infection. HEK293T, Vero-E6, and Huh-7 cells were treated with two DUB inhibitors (PR619 or WP1130). Then, infected cells were evaluated by flow cytometry, and viral progeny was quantified using the plaque assay method. The changes in viral proteins and viral RNA were analyzed using Western blotting and RT-qPCR, respectively. Results indicate that treatment with DUB inhibitors impairs CHIKV replication due to significant protein and viral RNA synthesis deregulation. Therefore, DUB activity may be a pharmacological target for blocking CHIKV infection.


Asunto(s)
Fiebre Chikungunya , Virus Chikungunya , Enzimas Desubicuitinizantes , Inhibidores Enzimáticos , Replicación Viral , Humanos , Fiebre Chikungunya/tratamiento farmacológico , Virus Chikungunya/efectos de los fármacos , Enzimas Desubicuitinizantes/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Células HEK293 , ARN Viral , Replicación Viral/efectos de los fármacos
8.
Cell Mol Life Sci ; 79(2): 117, 2022 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-35118522

RESUMEN

Although damaged cells can be repaired, cells that are considered unlikely to be repaired are eliminated through apoptosis, a type of predicted cell death found in multicellular organisms. Apoptosis is a structured cell death involving alterations to the cell morphology and internal biochemical changes. This process involves the expansion and cracking of cells, changes in cell membranes, nuclear fragmentation, chromatin condensation, and chromosome cleavage, culminating in the damaged cells being eaten and processed by other cells. The ubiquitin-proteasome system (UPS) is a major cellular pathway that regulates the protein levels through proteasomal degradation. This review proposes that apoptotic proteins are regulated through the UPS and describes a unique direction for cancer treatment by controlling proteasomal degradation of apoptotic proteins, and small molecules targeted to enzymes associated with UPS.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis/fisiología , Enzimas Desubicuitinizantes/metabolismo , Mitocondrias/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/metabolismo , Apoptosis/efectos de los fármacos , Caspasas/metabolismo , Citocromos c/metabolismo , Enzimas Desubicuitinizantes/antagonistas & inhibidores , Humanos , Modelos Biológicos , Piperidonas/farmacología , Inhibidores de Proteasas/farmacología
9.
Mol Med Rep ; 25(1)2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34850961

RESUMEN

Cardiovascular ischemia/reperfusion (I/R) injury is primarily caused by oxygen recovery after prolonged hypoxia. Previous studies found that the long non coding RNA (lncRNA) nuclear enriched abundant transcript 1 (NEAT1) was involved in cardiovascular pathology, and that NOD­like receptor protein 3 (NLRP3) inflammasome activation­dependent pyroptosis played a key role in cardiovascular I/R injury. The present study aimed to explore the molecular mechanism of I/R pathogenesis in order to provide novel insights for potential future therapies. Cell viability and lactate dehydrogenase enzyme activity assays were used to detect cell injury after human umbilical vein endothelial cells (HUVECs) were subjected to hypoxia/reoxygenation (H/R). The expression of the NEAT1/microRNA (miR)­204/BRCA1/BRCA2­containing complex subunit 3 (BRCC3) axis was examined by reverse transcription­quantitative PCR, and the associations among genes were confirmed by luciferase reporter assays. Western blotting and ELISA were used to measure the level of NLRP3 inflammasome activation­dependent pyroptosis. The results demonstrated that NEAT1, BRCC3 expression and NLRP3 inflammasome activation­dependent pyroptosis were significantly increased in H/R­injured HUVECs, whereas silencing BRCC3 or NEAT1 attenuated H/R­induced injury and pyroptosis. NEAT1 positively regulated BRCC3 expression via competitively binding with miR­204. Moreover, NEAT1 overexpression counteracted miR­204 mimic­induced injury, BRCC3 expression and NLRP3 inflammasome activation­dependent pyroptosis. Taken together, these findings demonstrated that inhibition of lncRNA NEAT1 protects HUVECs against H/R­induced NLRP3 inflammasome activation by targeting the miR­204/BRCC3 axis.


Asunto(s)
Enzimas Desubicuitinizantes/antagonistas & inhibidores , Endotelio Vascular/efectos de los fármacos , Hipoxia/fisiopatología , Inflamación/prevención & control , MicroARNs/antagonistas & inhibidores , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , ARN Largo no Codificante/antagonistas & inhibidores , Supervivencia Celular , Enzimas Desubicuitinizantes/genética , Enzimas Desubicuitinizantes/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inflamasomas/efectos de los fármacos , Inflamasomas/metabolismo , Inflamación/etiología , Inflamación/metabolismo , Inflamación/patología , MicroARNs/genética , Sustancias Protectoras/farmacología , Piroptosis , Daño por Reperfusión
10.
Leukemia ; 36(1): 210-220, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34326465

RESUMEN

Mutations in the Janus Kinase 2 (JAK2) gene resulting in constitutive kinase activation represent the most common genetic event in myeloproliferative neoplasms (MPN), a group of diseases involving overproduction of one or more kinds of blood cells, including red cells, white cells, and platelets. JAK2 kinase inhibitors, such as ruxolitinib, provide clinical benefit, but inhibition of wild-type (wt) JAK2 limits their clinical utility due to toxicity to normal cells, and small molecule inhibition of mutated JAK2 kinase activity can lead to drug resistance. Here, we present a strategy to target mutated JAK2 for degradation, using the cell's intracellular degradation machinery, while sparing non-mutated JAK2. We employed a chemical genetics screen, followed by extensive selectivity profiling and genetic studies, to identify the deubiquitinase (DUB), JOSD1, as a novel regulator of mutant JAK2. JOSD1 interacts with and stabilizes JAK2-V617F, and inactivation of the DUB leads to JAK2-V617F protein degradation by increasing its ubiquitination levels, thereby shortening its protein half-life. Moreover, targeting of JOSD1 leads to the death of JAK2-V617F-positive primary acute myeloid leukemia (AML) cells. These studies provide a novel therapeutic approach to achieving selective targeting of mutated JAK2 signaling in MPN.


Asunto(s)
Enzimas Desubicuitinizantes/antagonistas & inhibidores , Janus Quinasa 2/genética , Leucemia Mieloide Aguda/tratamiento farmacológico , Mutación , Trastornos Mieloproliferativos/tratamiento farmacológico , Bibliotecas de Moléculas Pequeñas/farmacología , Anciano , Anciano de 80 o más Años , Apoptosis , Proliferación Celular , Humanos , Leucemia Mieloide Aguda/enzimología , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Persona de Mediana Edad , Trastornos Mieloproliferativos/enzimología , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/patología , Fosforilación , Pronóstico , Células Tumorales Cultivadas
11.
Mol Cell ; 82(1): 15-29, 2022 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-34813758

RESUMEN

Deubiquitinases (DUBs) are specialized proteases that remove ubiquitin from substrates or cleave within ubiquitin chains to regulate ubiquitylation and therefore play important roles in eukaryotic biology. Dysregulation of DUBs is implicated in several human diseases, highlighting the importance of DUB function. In addition, many pathogenic bacteria and viruses encode and deploy DUBs to manipulate host immune responses and establish infectious diseases in humans and animals. Hence, therapeutic targeting of DUBs is an increasingly explored area that requires an in-depth mechanistic understanding of human and pathogenic DUBs. In this review, we summarize the multiple layers of regulation that control autoinhibition, activation, and substrate specificity of DUBs. We discuss different strategies to inhibit DUBs and the progress in developing selective small-molecule DUB inhibitors. Finally, we propose a classification system of DUB inhibitors based on their mode of action.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , COVID-19 , Enzimas Desubicuitinizantes , Inhibidores Enzimáticos/uso terapéutico , SARS-CoV-2 , Ubiquitinación/efectos de los fármacos , COVID-19/enzimología , Enzimas Desubicuitinizantes/antagonistas & inhibidores , Enzimas Desubicuitinizantes/metabolismo , Humanos
12.
Int J Mol Sci ; 22(21)2021 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-34769137

RESUMEN

Cisplatin-based chemotherapy is the standard treatment for bladder urothelial carcinoma (UC). Most patients experience chemoresistance, the primary cause of treatment failure, which leads to disease relapse. The underlying mechanism of chemoresistance involves reduced apoptosis. In this study, we investigated the antitumor effect of the deubiquitylating enzyme inhibitor PR-619 in cisplatin-resistant bladder UC. Deubiquitinase (ubiquitin-specific protease 14 (USP14) and USP21) immunohistochemical staining demonstrated that deubiquitination is related to chemoresistance in patients with metastatic UC and may be a target for overcoming chemoresistance. Cytotoxicity and apoptosis were assessed using fluorescence-activated flow cytometry and a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium assay, and PR-619 was found to enhance the cytotoxic and apoptotic effects of cisplatin in cisplatin-resistant T24/R cells. Mitigated cisplatin chemoresistance was associated with the concurrent suppression of c-Myc expression in T24/R cells. Moreover, the expression of c-Myc was upregulated in human bladder UC specimens from patients with chemoresistance. Experiments in a xenograft nude mouse model confirmed that PR-619 enhanced the antitumor effects of cisplatin. These results are promising for the development of therapeutic strategies to prevent UC chemoresistance through the combined use of chemotherapeutic agents/deubiquitination inhibitors (PR-619) by targeting the c-Myc pathway.


Asunto(s)
Aminopiridinas/uso terapéutico , Carcinoma/tratamiento farmacológico , Enzimas Desubicuitinizantes/antagonistas & inhibidores , Resistencia a Antineoplásicos/efectos de los fármacos , Tiocianatos/uso terapéutico , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Aminopiridinas/farmacología , Animales , Antineoplásicos/uso terapéutico , Carcinoma/metabolismo , Línea Celular Tumoral , Cisplatino/uso terapéutico , Humanos , Ratones Desnudos , Tiocianatos/farmacología , Ubiquitina Tiolesterasa/metabolismo , Neoplasias de la Vejiga Urinaria/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
13.
STAR Protoc ; 2(4): 100896, 2021 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-34723213

RESUMEN

Identification of selective deubiquitinase (DUB) inhibitors is critical for probe development to further understand and explore DUB biological function. Here, we detail the optimization and deployment of an in vitro fluorogenic ubiquitin-rhodamine assay to conduct high-throughput screening of a small molecule library against a panel of DUBs. In screening the compound library against multiple DUBs in parallel, we describe an approach for identifying selective DUB inhibitors and provide a roadmap for enabling selective DUB inhibitor discovery. For complete details on the use and execution of this protocol, please refer to Varca et al. (2021).


Asunto(s)
Enzimas Desubicuitinizantes , Inhibidores Enzimáticos , Ensayos Analíticos de Alto Rendimiento/métodos , Rodaminas/metabolismo , Ubiquitina/metabolismo , Enzimas Desubicuitinizantes/antagonistas & inhibidores , Enzimas Desubicuitinizantes/metabolismo , Pruebas de Enzimas , Escherichia coli/genética , Colorantes Fluorescentes/química , Proteínas Recombinantes/efectos de los fármacos , Proteínas Recombinantes/metabolismo , Bibliotecas de Moléculas Pequeñas
14.
J Pharm Pharm Sci ; 24: 390-399, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34319871

RESUMEN

PURPOSE: SARS-CoV-2 infection is associated with substantial mortality and high morbidity. This study tested the effect of angiotensin II type I receptor blocker, losartan, on SARS-CoV-2 replication and inhibition of the papain-like protease of the virus. METHODS: The dose-dependent inhibitory effect of losartan, in concentrations from 1µM to 100µM as determined by quantitative cell analysis combining fluorescence microscopy, image processing, and cellular measurements (Cellomics analysis) on SARS-CoV-2 replication was investigated in Vero E6 cells. The impact of losartan on deubiquitination and deISGylation of SARS-CoV-2 papain-like protease (PLpro) were also evaluated.  Results: Losartan reduced PLpro cleavage of tetraUbiquitin to diUbiquitin.  It was less effective in inhibiting PLpro's cleavage of ISG15-AMC than Ubiquitin-AMC.  To determine if losartan inhibited SARS-CoV-2 replication, losartan treatment of SARS-CoV-2 infected Vero E6 was examined. Losartan treatment one hour prior to SARS-CoV-2 infection reduced levels of SARS-CoV-2 nuclear protein, an indicator of virus replication, by 80% and treatment one-hour post-infection decreased viral replication by 70%. CONCLUSION: Losartan was not an effective inhibitor of deubiquitinase or deISGylase activity of the PLpro but affected the SARS-CoV-2 replication of Vero E6 cells in vitro.  As losartan has a favorable safety profile and is currently available it has features necessary for efficacious drug repurposing and treatment of COVID-19.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Antivirales/farmacología , Losartán/farmacología , SARS-CoV-2/efectos de los fármacos , Animales , Chlorocebus aethiops , Biología Computacional , Proteasas Similares a la Papaína de Coronavirus/antagonistas & inhibidores , Proteasas Similares a la Papaína de Coronavirus/metabolismo , Enzimas Desubicuitinizantes/antagonistas & inhibidores , Enzimas Desubicuitinizantes/metabolismo , SARS-CoV-2/metabolismo , SARS-CoV-2/fisiología , Ubiquitina/metabolismo , Células Vero , Replicación Viral/efectos de los fármacos , Tratamiento Farmacológico de COVID-19
15.
Biomolecules ; 11(6)2021 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-34071582

RESUMEN

The urgent need for novel and effective drugs against the SARS-CoV-2 coronavirus pandemic has stimulated research worldwide. The Papain-like protease (PLpro), which is essential for viral replication, shares a similar active site structural architecture to other cysteine proteases. Here, we have used representatives of the Ovarian Tumor Domain deubiquitinase family OTUB1 and OTUB2 along with the PLpro of SARS-CoV-2 to validate and rationalize the binding of inhibitors from previous SARS-CoV candidate compounds. By forming a new chemical bond with the cysteine residue of the catalytic triad, covalent inhibitors irreversibly suppress the protein's activity. Modeling covalent inhibitor binding requires detailed knowledge about the compounds' reactivities and binding. Molecular Dynamics refinement simulations of top poses reveal detailed ligand-protein interactions and show their stability over time. The recently discovered selective OTUB2 covalent inhibitors were used to establish and validate the computational protocol. Structural parameters and ligand dynamics are in excellent agreement with the ligand-bound OTUB2 crystal structures. For SARS-CoV-2 PLpro, recent covalent peptidomimetic inhibitors were simulated and reveal that the ligand-protein interaction is very dynamic. The covalent and non-covalent docking plus subsequent MD refinement of known SARS-CoV inhibitors into DUBs and the SARS-CoV-2 PLpro point out a possible approach to target the PLpro cysteine protease from SARS-CoV-2. The results show that such an approach gives insight into ligand-protein interactions, their dynamic character, and indicates a path for selective ligand design.


Asunto(s)
Enzimas Desubicuitinizantes/antagonistas & inhibidores , Inhibidores de Proteasas/química , SARS-CoV-2/metabolismo , Proteasas Virales/química , Sitios de Unión , COVID-19/patología , Dominio Catalítico , Enzimas Desubicuitinizantes/metabolismo , Diseño de Fármacos , Femenino , Humanos , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Ligandos , Simulación de Dinámica Molecular , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Inhibidores de Proteasas/metabolismo , SARS-CoV-2/aislamiento & purificación , Proteasas Virales/metabolismo
16.
Cell Chem Biol ; 28(12): 1758-1771.e13, 2021 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-34129829

RESUMEN

Deubiquitinating enzymes (DUBs) are a class of isopeptidases that regulate ubiquitin dynamics through catalytic cleavage of ubiquitin from protein substrates and ubiquitin precursors. Despite growing interest in DUB biological function and potential as therapeutic targets, few selective small-molecule inhibitors and no approved drugs currently exist. To identify chemical scaffolds targeting specific DUBs and establish a broader framework for future inhibitor development across the gene family, we performed high-throughput screening of a chemically diverse small-molecule library against eight different DUBs, spanning three well-characterized DUB families. Promising hit compounds were validated in a series of counter-screens and orthogonal assays, as well as further assessed for selectivity across expanded panels of DUBs. Through these efforts, we have identified multiple highly selective DUB inhibitors and developed a roadmap for rapidly identifying and validating selective inhibitors of related enzymes.


Asunto(s)
Enzimas Desubicuitinizantes/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Enzimas Desubicuitinizantes/metabolismo , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Femenino , Células HEK293 , Humanos , Estructura Molecular , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/química
17.
Cell Chem Biol ; 28(7): 1090-1100, 2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-34089649

RESUMEN

Deubiquitinating enzymes (DUBs) are a largely understudied and untapped resource in the toolkit of protein degradation functionalities. They comprise a large repertoire of enzymes that remove ubiquitin from substrates in a variety of cellular and pathophysiological contexts, and have enormous potential for research and clinical use. It is only within the last 5 years that potent, selective, and well-characterized small-molecule inhibitors of DUBs have been described. These compounds are now being used to study the biological roles of DUBs. Here, we describe downstream applications of small-molecule inhibitors for studying DUBs and provide a framework for future studies. We highlight recent examples of using these inhibitors to confirm and explore the role of these enzymes in both normal and pathological contexts. These studies represent the first steps in the burgeoning field of pharmacological and chemoproteomic studies of DUBs, which will be critical for the continued advancement of DUB field.


Asunto(s)
Enzimas Desubicuitinizantes/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Enzimas Desubicuitinizantes/metabolismo , Inhibidores Enzimáticos/química , Humanos , Bibliotecas de Moléculas Pequeñas/química , Ubiquitina/metabolismo , Ubiquitinación/efectos de los fármacos
18.
Sci Immunol ; 6(58)2021 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-33931568

RESUMEN

Pharmacologically inhibiting nucleotide-binding domain and leucine-rich repeat-containing (NLR) family, pyrin domain-containing protein 3 (NLRP3) inflammasome activation results in potent therapeutic effects in a wide variety of preclinical inflammatory disease models. NLRP3 deubiquitination is essential for efficient NLRP3 inflammasome activity, but it remains unclear whether this process can be harnessed for therapeutic benefit. Here, we show that thiolutin (THL), an inhibitor of the JAB1/MPN/Mov34 (JAMM) domain-containing metalloprotease, blocks NLRP3 inflammasome activation by canonical, noncanonical, alternative, and transcription-independent pathways at nanomolar concentrations. In addition, THL potently inhibited the activation of multiple NLRP3 mutants linked with cryopyrin-associated periodic syndromes (CAPS). Treatment with THL alleviated NLRP3-related diseases in mouse models of lipopolysaccharide-induced sepsis, monosodium urate-induced peritonitis, experimental autoimmune encephalomyelitis, CAPS, and methionine-choline-deficient diet-induced nonalcoholic fatty liver disease. Mechanistic studies revealed that THL inhibits the BRCC3-containing isopeptidase complex (BRISC)-mediated NLRP3 deubiquitination and activation. In addition, we show that holomycin, a natural methyl derivative of THL, displays an even higher inhibitory activity against NLRP3 inflammasome than THL. Our study validates that posttranslational modification of NLRP3 can be pharmacologically targeted to prevent or treat NLRP3-associated inflammatory diseases. Future clinical development of derivatives of THL may provide new therapies for NLRP3-related diseases.


Asunto(s)
Enzimas Desubicuitinizantes/antagonistas & inhibidores , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Inflamasomas/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Animales , Enzimas Desubicuitinizantes/genética , Enzimas Desubicuitinizantes/metabolismo , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Sangre Fetal , Humanos , Inflamasomas/inmunología , Inflamasomas/metabolismo , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Lactamas/farmacología , Lactamas/uso terapéutico , Lipopolisacáridos , Masculino , Ratones , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/inmunología , Embarazo , Cultivo Primario de Células , Pirrolidinonas/farmacología , Pirrolidinonas/uso terapéutico , Células THP-1 , Ubiquitinación/efectos de los fármacos
19.
Biochem Biophys Res Commun ; 554: 186-192, 2021 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-33798946

RESUMEN

Promyelocytic leukemia protein (PML) nuclear bodies (NBs) are dynamic and multiprotein complexes implicated in a variety of important biochemical events. Due to alternative mRNA splicing, PML has at least six nuclear isoforms that share a common N-terminus but differ in their C-terminal regions. However, the unique role of each PML isoform is not clear. Here, we report the characterization of the deubiquitinase ataxin-3 as a specific binding partner of PML isoform II (PML-II). Ataxin-3 was identified as a potential binding protein of PML-II in a yeast-hybrid screen employing the unique C-terminal region of PML-II as bait. Ataxin-3 only binds to the C-terminal region of PML-II and not that of other PML isoforms. The interaction between ataxin-3 and PML-II was confirmed by co-immunoprecipition assays, and immunofluorescent microscopy revealed that PML-II and ataxin-3 were co-localized in PML-NBs. In addition, PML-II not only interacts with ataxin-3 with a normal range of poly-Q repeats (13Q), but also with a pathological form of ataxin-3 with extended poly-Q repeats (79Q). Importantly, the deubiquitinase activity of ataxin-3 was inhibited by PML-II. Our results suggest that PML-II may be a negative regulator of ataxin-3.


Asunto(s)
Ataxina-3/metabolismo , Enzimas Desubicuitinizantes/antagonistas & inhibidores , Cuerpos de Inclusión Intranucleares/metabolismo , Proteína de la Leucemia Promielocítica/metabolismo , Proteínas Represoras/metabolismo , Empalme Alternativo , Ataxina-3/genética , Línea Celular Tumoral , Humanos , Proteína de la Leucemia Promielocítica/genética , Unión Proteica , Isoformas de Proteínas , Proteínas Represoras/genética , Proteínas Supresoras de Tumor/metabolismo , Ubiquitinación
20.
Cancer Sci ; 112(6): 2287-2298, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33738896

RESUMEN

Drug resistance is a significant obstacle to effective cancer treatment. Drug resistance develops from initially reversible drug-tolerant cancer cells, which offer therapeutic opportunities to impede cancer relapse. The mechanisms of resistance to proteasome inhibitor (PI) therapy have been investigated intensively, however the ways by which drug-tolerant cancer cells orchestrate their adaptive responses to drug challenges remain largely unknown. Here, we demonstrated that cyclin A1 suppression elicited the development of transient PI tolerance in mixed-lineage leukemia (MLL) cells. This adaptive process involved reversible downregulation of cyclin A1, which promoted PI resistance through cell-cycle arrest. PI-tolerant MLL cells acquired cyclin A1 dependency, regulated directly by MLL protein. Loss of cyclin A1 function resulted in the emergence of drug tolerance, which was associated with patient relapse and reduced survival. Combination treatment with PI and deubiquitinating enzyme (DUB) inhibitors overcame this drug resistance by restoring cyclin A1 expression through chromatin crosstalk between histone H2B monoubiquitination and MLL-mediated histone H3 lysine 4 methylation. These results reveal the importance of cyclin A1-engaged cell-cycle regulation in PI resistance in MLL cells, and suggest that cell-cycle re-entry by DUB inhibitors may represent a promising epigenetic therapeutic strategy to prevent acquired drug resistance.


Asunto(s)
Ciclina A1/metabolismo , Enzimas Desubicuitinizantes/antagonistas & inhibidores , Tolerancia a Medicamentos , Leucemia Bifenotípica Aguda/tratamiento farmacológico , Inhibidores de Proteasoma/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/genética , Línea Celular Tumoral , Cromatina/metabolismo , Ciclina A1/genética , Resistencia a Antineoplásicos , Tolerancia a Medicamentos/genética , Regulación Neoplásica de la Expresión Génica , Histonas/metabolismo , Humanos , Leucemia Bifenotípica Aguda/genética , Leucemia Bifenotípica Aguda/metabolismo , Leucemia Bifenotípica Aguda/patología , Metilación , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Pronóstico , Inhibidores de Proteasoma/uso terapéutico , Ubiquitinación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...