Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Neuromolecular Med ; 25(3): 313-329, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-36739586

RESUMEN

Parkinson's disease (PD) is the most common neurodegenerative movement disorder. There are no available therapeutics that slow or halt the progressive loss of dopamine-producing neurons, which underlies the primary clinical symptoms. Currently approved PD drugs can provide symptomatic relief by increasing brain dopamine content or activity; however, the alleviation is temporary, and the effectiveness diminishes with the inevitable progression of neurodegeneration. Discovery and development of disease-modifying neuroprotective therapies has been hampered by insufficient understanding of the root cause of PD-related neurodegeneration. The etiology of PD involves a combination of genetic and environmental factors. Although a single cause has yet to emerge, genetic, cell biological and neuropathological evidence implicates mitochondrial dysfunction and protein aggregation. Postmortem PD brains show pathognomonic Lewy body intraneuronal inclusions composed of aggregated α-synuclein, indicative of failure to degrade misfolded protein. Mutations in the genes that code for α-synuclein, as well as the E3 ubiquitin ligase Parkin, cause rare inherited forms of PD. While many ubiquitin ligases label proteins with ubiquitin chains to mark proteins for degradation by the proteasome, Parkin has been shown to mark dysfunctional mitochondria for degradation by mitophagy. The ubiquitin proteasome system participates in several aspects of the cell's response to mitochondrial damage, affording numerous therapeutic opportunities to augment mitophagy and potentially stop PD progression. This review examines the role and therapeutic potential of such UPS modulators, exemplified by both ubiquitinating and deubiquitinating enzymes.


Asunto(s)
Enfermedades Neurodegenerativas , Enfermedad de Parkinson , Humanos , Enfermedad de Parkinson/genética , alfa-Sinucleína/genética , Complejo de la Endopetidasa Proteasomal , Ubiquitina/metabolismo , Dopamina , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
2.
Eur J Immunol ; 50(1): 48-55, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31531847

RESUMEN

TAM receptors (Tyro3, Axl, and Mer) are receptor tyrosine kinases (RTKs) that are expressed by multiple immune cells including NK cells. Although RTKs typically enhance cellular functions, TAM receptor ligation blocks NK-cell activation. The mechanisms by which RTKs block NK-cell signaling downstream of activating receptors are unknown. In this report, we demonstrate that TAM receptors attenuate NK cell responses via the activity of E3 ubiquitin ligase Casitas B lineage lymphoma b (Cbl-b). Specifically, we show that Tyro3, Axl, and Mer phosphorylate Cbl-b, and Tyro3 ligation activates Cbl-b by phosphorylating tyrosine residues 133 and 363. Ligation of TAM receptors by their ligand Gas6 suppresses activating receptor-stimulated NK-cell functions such as IFN-γ production and degranulation, in a TAM receptor kinase- and Cbl-b-dependent manner. Moreover, Gas6 ligation induces the degradation of LAT1, a transmembrane adaptor protein required for NK cell activating receptor signaling, in WT but not in Cbl-b knock-out NK cells. Together, these results suggest that TAM receptors may attenuate NK-cell function by phosphorylating Cbl-b, which in turn dampens NK-cell activation signaling by promoting the degradation of LAT1. Our data therefore support a mechanism by which RTKs attenuate, rather than stimulate, signaling pathways via the activation of ubiquitin ligases.


Asunto(s)
Células Asesinas Naturales/metabolismo , Activación de Linfocitos/fisiología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Proteínas Adaptadoras Transductoras de Señales/inmunología , Sistema de Transporte de Aminoácidos y+L/metabolismo , Animales , Células Asesinas Naturales/inmunología , Ratones , Fosforilación , Proteínas Proto-Oncogénicas c-cbl/inmunología , Proteínas Tirosina Quinasas Receptoras/inmunología , Transducción de Señal/inmunología
3.
Trends Biochem Sci ; 44(7): 599-615, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30819414

RESUMEN

Post-translational modifications (PTMs) by ubiquitin (Ub) are versatile, highly dynamic, and involved in nearly all aspects of eukaryote biological function. The reversibility and heterogeneity of Ub chains attached to protein substrates have complicated their isolation, quantification, and characterization. Strategies have emerged to isolate endogenous ubiquitylated targets, including technologies based on the use of Ub-binding peptides, such as tandem-repeated Ub-binding entities (TUBEs). TUBEs allow the identification and characterization of Ub chains, and novel substrates for deubiquitylases (DUBs) and Ub ligases (E3s). Here we review their impact on purification, analysis of pan or chain-selective polyubiquitylated proteins and underline the biological relevance of this information. Together with peptide aptamers and other Ub affinity-based approaches, TUBEs will contribute to unraveling the secrets of the Ub code.


Asunto(s)
Ubiquitina/metabolismo , Ubiquitinación , Animales , Humanos , Unión Proteica , Procesamiento Proteico-Postraduccional
4.
J Med Chem ; 61(2): 422-443, 2018 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-28768102

RESUMEN

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.


Asunto(s)
Antineoplásicos/farmacología , Evaluación Preclínica de Medicamentos/métodos , Inhibidores Enzimáticos/farmacología , Peptidasa Específica de Ubiquitina 7/antagonistas & inhibidores , Peptidasa Específica de Ubiquitina 7/metabolismo , Antineoplásicos/química , Ensayos Clínicos como Asunto , Cristalografía por Rayos X , Inhibidores Enzimáticos/química , Humanos , Terapia Molecular Dirigida/métodos , Inhibidores de Proteasoma/farmacología , Conformación Proteica , Ubiquitina/metabolismo , Peptidasa Específica de Ubiquitina 7/química , Peptidasa Específica de Ubiquitina 7/inmunología
5.
PLoS One ; 12(12): e0189744, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29236775

RESUMEN

Accumulation of Foxp3+ T-regulatory (Treg) cells in the tumor microenvironment is associated with tumor immune evasion and poor patient outcome in the case of many solid tumors. Current therapeutic strategies for blocking Treg functions are not Treg-specific, and display only modest and transient efficacy. Recent studies revealed that ubiquitin-specific protease 7 (USP7) is essential for Treg functions by stabilizing expression of Tip60 and Foxp3, which together are central to the development and maintenance of the Treg cell lineage. Pharmacological inhibition of USP7 is therefore a promising strategy for suppressing Treg functions and promoting anti-tumor immunity. Previously, we reported the P5091 series of small molecule USP7 inhibitors and demonstrated their direct anti-tumor activity in vivo using xenograft models. However, the precise mechanism of action of these compounds was not well defined. In this study, we report the development and characterization of P217564, a second-generation USP7 inhibitor with improved potency and selectivity. P217564 selectively targets the catalytic cleft of USP7 and modifies its active site cysteine (C223) by forming a covalent adduct. Irreversible inhibition of USP7 results in durable downstream biological responses in cells, including down-regulation of Tip60 and consequent impairment of Treg suppressive function. In addition, we demonstrate that both USP7 and various USP7 substrates are subjected to Lys48-mediated ubiquitin modification, consistent with increased proteasomal degradation of these proteins because of USP7 inhibition.


Asunto(s)
Proteínas de Drosophila/metabolismo , Factores de Transcripción Forkhead/metabolismo , Histona Acetiltransferasas/metabolismo , Linfocitos T Reguladores/inmunología , Peptidasa Específica de Ubiquitina 7/antagonistas & inhibidores , Dominio Catalítico , Línea Celular Tumoral , Humanos , Inhibidores de Proteasas/farmacología , Peptidasa Específica de Ubiquitina 7/metabolismo , Ubiquitinación
6.
Opt Express ; 24(24): 27371-27381, 2016 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-27906309

RESUMEN

Particle tracking velocimetry (PTV) is a valuable tool for microfluidic analysis. Especially mixing processes and the environmental interaction of fluids on a microscopic scale are of particular importance for pharmaceutical and biomedical applications. However, currently applied techniques suffer from the lag of instantaneous depth information. Here we present a scan-free, shadow-imaging PTV-technique for 3D trajectory and velocity measurement of flow fields in micro-channels with 2 µm spatial resolution. By using an incoherent light source, one camera and a spatial light modulator (LCoS-SLM) that generates double-images of the seeding particle shadows, it is a simply applicable and highly scalable technique.

7.
Cell Biochem Biophys ; 67(1): 161-7, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23695783

RESUMEN

The ubiquitin pathway regulates diverse functions including protein localization and stability. The complexity of the pathway involving nearly 40 identified E2 conjugating enzymes and over 600 E3 ligases raises the issue of specificity. With the E2s and E3s fitting into a limited number of classes based on bioinformatics, structures, and proven activities, there is not a clear picture as to what would determine which E2/E3 enzyme pair would be functional. There have been many reports of limited E2/E3 activity profiling with a small number of E2s and E3s. We have expanded on this to investigate the activity of ubiquitin E2s covering the majority of the reported classes/families in concert with a number of E3s implicated in a variety of diseases. Using an ELISA-based assay we screened 10 E3 ligases against a panel of 11 E2s to determine which E2/E3 pairs exhibited E3 autoubiquitylation activity. In addition, the ubiquitin chain linkage preference by certain E2/E3 pairs was investigated. Finally, substrate ubiquitylation was assayed for the E3 ligase MuRF1 using various E2/MuRF1 pairs. These studies demonstrate the utility of identifying the correct E2/E3 pair to monitor specific substrate ubiquitylation.


Asunto(s)
Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Humanos , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Especificidad por Sustrato , Enzimas Ubiquitina-Conjugadoras/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
8.
Biochim Biophys Acta ; 1823(11): 2094-7, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22721718

RESUMEN

The ubiquitin proteasome pathway controls the cellular degradation of ~80-90% of the proteome in a highly regulated manner. In this pathway, E3 ligases are responsible for the conjugation of ubiquitin to protein substrates which can lead to their destruction by the 26S proteasome. Aberrant E3 ligases have been implicated in several diseases and are widely recognized as attractive targets for drug discovery. As researchers continue to characterize E3 ligases, additional associations with various disease states are being exposed. The availability of assays that allow rapid analysis of E3 ligase activity is paramount to both biochemical studies and drug discovery efforts aimed at E3 ligases. To address this need, we have developed a homogenous assay for monitoring ubiquitin chain formation using Tandem Ubiquitin Binding Entities (TUBEs). TUBEs bind selectively to polyubiquitin chains versus mono-ubiquitin thus enabling the detection of polyubiquitin chains in the presence of mono-ubiquitin. This assay reports on the proximity between the protein substrate and TUBEs as a result of polyubiquitin chain formation by an E3 ligase. This homogenous assay is a step forward in streamlining an approach for characterizing and quantitating E3 ligase activity in a rapid and cost effective manner. This article is part of a Special Issue entitled: Ubiquitin Drug Discovery and Diagnostics.


Asunto(s)
Proteínas Musculares/metabolismo , Poliubiquitina/metabolismo , Factores de Transcripción/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina/metabolismo , Ensayos Analíticos de Alto Rendimiento , Humanos , Unión Proteica , Secuencias Repetidas en Tándem , Proteínas de Motivos Tripartitos , Ubiquitinación
9.
Biochim Biophys Acta ; 1823(11): 2014-21, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22610084

RESUMEN

The proteasome inhibitor bortezomib remains the only ubiquitin pathway effector to become a drug (VELCADE®) and has become a successful treatment for hematological malignancies. While producing a global cellular effect, proteasome inhibitors have not triggered the catastrophe articulated initially in terms such as "buildup of cellular garbage". Proteasome inhibitors, in fact, do have a therapeutic window, although in the case of the prototype bortezomib it is small owing to peripheral neuropathy, myelosuppression and, as recently reported, cardiotoxicity [1]. Currently, several second-generation molecules are undergoing clinical evaluation to increase this window. An alternative strategy is to target ubiquitin pathway enzymes acting at non-proteasomal sites-E1, E2, and E3, associated with ubiquitin conjugation, and deubiquitylating enzymes ("DUBs")-that act locally on selected targets rather than on the whole cell. Inhibitors (or activators, in some cases) of these enzymes should be developable as selective antitumor agents with toxicity profiles superior to that of bortezomib. Various therapeutic hypotheses follow from known cellular mechanisms of these target enzymes; most hypotheses relate to cancer, reminiscent of the FDA-approved protein kinase inhibitors now marketed. Since ubiquitin tagging controls the cellular content, activity, or compartmentation of proteins associated with disease, inhibitors or activators of ubiquitin conjugation or deconjugation are predicted to have an impact on disease. For practical and empirical reasons, inhibitors of ubiquitin pathway enzymes have been the favored therapeutic avenue. In approximately the time that has elapsed since the approval of bortezomib in 2003, there has been some progress in developing potential anticancer drugs that target various ubiquitin pathway enzymes. An E1 inhibitor and inhibitors of E3 are now in clinical trial, with some objective responses reported. Appropriate assays and/or rational design may uncover improved inhibitors of these enzymes, as well as E2 and DUBs, for further development. Presently, it should become clear whether one or both of the two general strategies for ubiquitin-based drug discovery will lead to truly superior new medicines for cancer and other diseases. This article is part of a Special Issue entitled: Ubiquitin Drug Discovery and Diagnostics.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Inhibidores de Proteasas/uso terapéutico , Complejo de la Endopetidasa Proteasomal/química , Ubiquitina/metabolismo , Animales , Ensayos Clínicos como Asunto , Humanos
10.
ACS Med Chem Lett ; 3(10): 789-92, 2012 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-24900381

RESUMEN

Inhibitors of the cancer-related cysteine isopeptidase human ubiquitin-specific proteases 7 (USP7) and 47 (USP47) are considered to have potential as cancer therapeutics, owing to their ability to stabilize the tumor suppressor p53 and to decrease DNA polymerase ß (Polß), both of which are potential anticancer effects. A new class of dual small molecule inhibitors of these enzymes has been discovered. Compound 1, a selective inhibitor of USP7 and USP47 with moderate potency, demonstrates inhibition of USP7 in cells and induces elevated p53 and apoptosis in cancer cell lines. Compound 1 has been shown to demonstrate modest activity in human xenograft multiple myeloma and B-cell leukemia in vivo models. This activity may be the result of dual inhibition of USP7 and USP47. To address issues regarding potency and developability, analogues of compound 1 have been synthesized and tested, leading to improvements in potency, solubility, and metabolic reactivity profile. Further optimization is expected to yield preclinical candidates and, ultimately, clinical candidates for the treatment of multiple myeloma, prostate cancer, and other cancers.

11.
Cell Biochem Biophys ; 60(1-2): 113-8, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21448668

RESUMEN

Progressive muscle wasting, also known as myopathy or muscle atrophy is a debilitating and life-threatening disorder. Myopathy is a pathological condition of many diseases including cancer, diabetes, COPD, and AIDS and is a natural consequence of inactivity and aging (sarcopenia). Muscle atrophy occurs when there is a net loss of muscle mass resulting in a change in the balance between protein synthesis and protein degradation. The ubiquitin pathway and specific ubiquitin pathway enzymes have been directly implicated in the progression of atrophy. The ubiquitin E3 ligase Muscle-specific RING Finger E3 ligase (MuRF1) is upregulated and increases protein degradation and muscle wasting in numerous muscle atrophy models. The inhibition of MuRF1 could be a novel mechanism to prevent or reverse muscle wasting associated with various pathologies. We screened a small molecule library for inhibitors to MuRF1 activity and identified P013222, an inhibitor of MuRF1 autoubiquitylation. Further, P013222 was shown to inhibit MuRF1-dependent substrate ubiquitylation, and was active in inhibiting MuRF1 in a cellular atrophy model. Thus MuRF1 can be targeted in a specific manner and produce positive results in cellular atrophy models.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Proteínas Musculares/antagonistas & inhibidores , Atrofia Muscular/prevención & control , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Animales , Biocatálisis/efectos de los fármacos , Western Blotting , Línea Celular , Dexametasona/farmacología , Relación Dosis-Respuesta a Droga , Glucocorticoides/farmacología , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Atrofia Muscular/metabolismo , Mioblastos Esqueléticos/citología , Mioblastos Esqueléticos/efectos de los fármacos , Mioblastos Esqueléticos/metabolismo , Cadenas Pesadas de Miosina/metabolismo , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/metabolismo , Bibliotecas de Moléculas Pequeñas , Especificidad por Sustrato , Proteínas de Motivos Tripartitos , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación/efectos de los fármacos
12.
Assay Drug Dev Technol ; 9(2): 165-73, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21133675

RESUMEN

The reversible conjugation of ubiquitin and ubiquitin-like (UbL) proteins to protein substrates plays a critical role in the regulation of many cellular pathways. The removal of ubiquitin from target proteins is performed by ubiquitin proteases also known as deubiquitylases (DUBs). Owing to their substrate specificity and the central role ubiquitylation plays in cell signaling pathways, DUB are attractive targets for therapeutic development. The development of DUB inhibitors requires assays that are amenable to high-throughput screening and provide rapid assessment of inhibitor selectivity. Determination of inhibitor selectivity at an early stage of drug discovery will reduce drug failure in the clinic as well as reduce overall drug development costs. We have developed two novel assays, UbL-Enterokinase light chain and UbL-Granzyme B, for quantifying ubiquitin and UbL protease activity. In our quest to discover and characterize novel chemical entities, we have combined these assays with a previously developed assay in a multiplex format. This multiplex format allows for the detection of three distinct protease activities simultaneously, in a single well. We have demonstrated that the multiplex format is able to distinguish between selective and nonselective protease inhibitors. Specifically, we have used this assay format to characterize P022077, a selective ubiquitin-specific protease 7 inhibitor discovered at Progenra.


Asunto(s)
Colorantes Fluorescentes/análisis , Inhibidores de Proteasas/química , Ubiquitina/antagonistas & inhibidores , Ubiquitinas/antagonistas & inhibidores , Técnicas de Laboratorio Clínico , Fluorescencia , Colorantes Fluorescentes/metabolismo , Pichia , Inhibidores de Proteasas/análisis , Inhibidores de Proteasas/metabolismo , Especificidad por Sustrato/fisiología , Ubiquitina/metabolismo , Ubiquitinas/metabolismo
13.
J Biomol Screen ; 15(10): 1220-8, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20864734

RESUMEN

The ubiquitin-proteasome system is central to the regulation of numerous cellular events, and dysregulation may lead to disease pathogenesis. E3 ubiquitin ligases typically function in concert with E1 and E2 enzymes to recruit specific substrates, thereby coordinating their ubiquitylation and subsequent proteasomal degradation or cellular activity. E3 ligases have been implicated in a wide range of pathologies, and monitoring their activity in a rapid and cost-effective manner would be advantageous in drug discovery. The relative lack of high-throughput screening (HTS)-compliant E3 ligase assays has significantly hindered the discovery of E3 inhibitors. Herein, the authors describe a novel HTS-compliant E3 ligase assay platform that takes advantage of a ubiquitin binding domain's inherent affinity for polyubiquitin chains, permitting the analysis of ubiquitin chain formation in an E3 ligase-dependent manner. This assay has been used successfully with members of both the RING and HECT families, demonstrating the platform's broad utility for analyzing a wide range of E3 ligases. The utility of the assay platform is demonstrated by the identification of inhibitors of the E3 ligase CARP2. As the number of E3 ligases associated with various disease states increases, the ability to quantitate the activity of these enzymes in an expeditious manner becomes imperative in drug discovery.


Asunto(s)
Enzimas Reparadoras del ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/antagonistas & inhibidores , Ensayos Analíticos de Alto Rendimiento/métodos , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas de Saccharomyces cerevisiae/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Enzimas Reparadoras del ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Descubrimiento de Drogas , Humanos , Luminiscencia , Proteínas del Tejido Nervioso/metabolismo , Poliubiquitina/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Bibliotecas de Moléculas Pequeñas , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Enzimas Activadoras de Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
14.
Biochem Soc Trans ; 38(Pt 1): 132-6, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20074047

RESUMEN

Dysregulation of the UPS (ubiquitin-proteasome system) has been implicated in a wide range of pathologies including cancer, neurodegeneration and viral infection. Inhibiting the proteasome has been shown to be an effective therapeutic strategy in humans; however, toxicity with this target remains high. E3s (Ub-protein ligases) represent an alternative attractive therapeutic target in the UPS. In this paper, we will discuss current platforms that report on E3 ligase activity and can detect E3 inhibitors, and underline the advantages and disadvantages of each approach.


Asunto(s)
Inhibidores de Proteasas/metabolismo , Inhibidores de Proteasoma , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Ubiquitina/metabolismo , Bioensayo/métodos , Humanos , Enfermedades del Sistema Inmune/tratamiento farmacológico , Enfermedades del Sistema Inmune/enzimología , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/enzimología , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
15.
Methods Mol Biol ; 497: 269-81, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19107424

RESUMEN

In this chapter we describe a novel, sensitive, homogenous high throughput reporter-based in vitro assay for SUMO protease activity developed by Progenra, Inc. A reporter construct was created by fusing His(6)-tagged small ubiquitin-like modifier (SUMO) to the amino terminus of the reporter enzyme phospholipase A(2) (PLA(2)). Following cleavage by a member of the sentrin specific proteases (SENPs), free PLA(2) is able to turn over its substrate, resulting in the release of a fluorescent product which is readily quantifiable using a fluorimeter or a fluorescence plate reader. The utility of this SUMO-CHOP-Reporter assay platform is demonstrated by its ability to determine K(m) values and to characterize inhibitors of SUMO proteases.


Asunto(s)
Técnicas Biosensibles/métodos , Péptido Hidrolasas/aislamiento & purificación , Péptido Hidrolasas/metabolismo , Proteína SUMO-1/metabolismo , Catálisis , Técnicas de Laboratorio Clínico , Cisteína Endopeptidasas/metabolismo , Endopeptidasas/metabolismo , Humanos , Cinética , Péptido Hidrolasas/química , Fosfolipasas A2/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Sensibilidad y Especificidad , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Factor de Transcripción CHOP/química , Factor de Transcripción CHOP/metabolismo , Factor de Transcripción CHOP/fisiología
16.
Biochem Soc Trans ; 36(Pt 5): 828-32, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18793145

RESUMEN

Dysregulation of the UPS (ubiquitin-proteasome system) has been implicated in a wide range of pathologies including cancer, neurodegeneration and viral infection. Inhibiting the proteasome has been shown to be an effective therapeutic strategy in humans; yet toxicity with this target remains high. DUBs (deubiquitinating enzymes) represent an alternative target in the UPS with low predicted toxicity. Currently, there are no DUB inhibitors that have been used clinically. To address this situation, Progenra has developed a novel assay to measure the proteolytic cleavage of Ub (ubiquitin) or UBL (Ub-like protein) conjugates such as SUMO (small Ub-related modifier), NEDD8 (neural-precursor-cell-expressed, developmentally down-regulated 8) or ISG15 (interferon-stimulated gene 15) by isopeptidases. In this review, current platforms for detecting DUB inhibitors are discussed and the advantages and disadvantages of the approaches are underlined.


Asunto(s)
Bioensayo/métodos , Endopeptidasas/metabolismo , Inhibidores Enzimáticos/metabolismo , Ubiquitina/metabolismo , Enfermedad , Endopeptidasas/genética , Inhibidores Enzimáticos/uso terapéutico , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Fosfolipasas A2/genética , Fosfolipasas A2/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/genética
17.
Protein Sci ; 17(6): 1035-43, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18424514

RESUMEN

Conjugation or deconjugation of ubiquitin (Ub) or ubiquitin-like proteins (UBLs) to or from cellular proteins is a multifaceted and universal means of regulating cellular physiology, controlling the lifetime, localization, and activity of many critical proteins. Deconjugation of Ub or UBL from proteins is performed by a class of proteases called isopeptidases. Herein is described a readily quantifiable novel isopeptidase assay platform consisting of Ub or UBL fused to the reporter enzyme phospholipase A(2) (PLA(2)). Isopeptidase activity releases PLA(2), which cleaves its substrate, generating a signal that is linear with deubiquitylase (DUB) concentration and is able to discriminate DUB, deSUMOylase, deNEDDylase, and deISGylase activities. The power and sensitivity of the UBL-PLA(2) assay are demonstrated by its ability to differentiate the contrasting deISGylase and DUB activities of two coronavirus proteases: severe acute respiratory syndrome papain-like protease (SARS-CoV PLpro) and NL63 CoV papain-like protease 2 (PLP2). Furthermore, direct comparisons with the current Ub-7-amino-4-methylcoumarin (Ub-AMC) assay demonstrated that the Ub-PLA(2) assay is an effective tool for characterizing modulators of isopeptidase activity. This observation was expanded by profiling the inhibitory activity of the nonselective isopeptidase inhibitor NSC 632839 against DUBs and deSUMOylases. Taken together, these studies illustrate the utility of the reporter-based approach to measuring isopeptidase activity.


Asunto(s)
Endopeptidasas/metabolismo , Ubiquitina/metabolismo , Animales , Endopeptidasas/aislamiento & purificación , Ratones , Inhibidores de Proteasas/farmacología
18.
J Med Chem ; 50(16): 3777-85, 2007 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-17636946

RESUMEN

High-throughput screening for inhibitors of the human metalloprotease, methionine aminopeptidase-2 (MetAP2), identified a potent class of 3-anilino-5-benzylthio-1,2,4-triazole compounds. Efficient array and interative synthesis of triazoles led to rapid SAR development around the aniline, benzylthio, and triazole moeities. Evaluation of these analogs in a human MetAP2 enzyme assay led to the identification of several inhibitors with potencies in the 50-100 picomolar range. The deleterious effects on inhibitor potency by methylation of the anilino-triazole nitrogens, as well as the X-ray crystal structure of triazole 102 bound in the active site of MetAP2, confirm the key interactions between the triazole nitrogens, the active site cobalt atoms, and the His-231 side-chain. The structure has also provided a rationale for interpreting SAR within the triazole series. Key aniline (2-isopropylphenyl) and sulfur substituents (furanylmethyl) identified in the SAR studies led to the identification of potent inhibitors (103 and 104) of endothelial cell proliferation. Triazoles 103 and 104 also exhibited dose-dependent activity in an aortic ring tissue model of angiogenesis highlighting the potential utility of MetAP2 inhibitors as anticancer agents.


Asunto(s)
Aminopeptidasas/antagonistas & inhibidores , Inhibidores de la Angiogénesis/síntesis química , Furanos/síntesis química , Metaloendopeptidasas/antagonistas & inhibidores , Tiazoles/síntesis química , Tiofenos/síntesis química , Triazoles/síntesis química , Inhibidores de la Angiogénesis/química , Inhibidores de la Angiogénesis/farmacología , Animales , Aorta Torácica/efectos de los fármacos , Capilares/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Cristalografía por Rayos X , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/citología , Furanos/química , Furanos/farmacología , Técnicas In Vitro , Masculino , Modelos Moleculares , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad , Tiazoles/química , Tiazoles/farmacología , Tiofenos/química , Tiofenos/farmacología , Triazoles/química , Triazoles/farmacología
19.
Future Oncol ; 3(2): 191-9, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17381419

RESUMEN

Tagging proteins with mono- or poly-ubiquitin is now recognized as a multifaceted and universal means of regulating cell growth and physiology. It does so by controlling the cellular lifetime of nearly all eukaryotic proteins and the cellular localization of many critical proteins. Enzymes of the ubiquitin pathway add (ligases) or remove (deubiquitinases [DUBs]) ubiquitin tags to or from their target proteins in a selective fashion. Similarly to the kinases and their corresponding phosphatases, ubiquitin ligases and DUBs have become actively studied molecular oncology targets for drug discovery. Approximately 79 functional DUBs exist in the human proteome, suggesting that selective intervention is a reasonable therapeutic objective, with the goal of downregulating or ablating oncogene products or, alternatively, upregulating or sparing tumor suppressors. In the following review, this fascinating class of regulatory enzymes will be described, and specific examples of DUBs that are viable targets for anticancer therapy will be considered.


Asunto(s)
Antineoplásicos/farmacología , Sistemas de Liberación de Medicamentos , Ubiquitina/efectos de los fármacos , Ubiquitina/metabolismo , Humanos , Proteína NEDD8 , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/efectos de los fármacos , Ubiquitina Tiolesterasa/efectos de los fármacos , Ubiquitinas/efectos de los fármacos
20.
Anal Biochem ; 350(2): 214-21, 2006 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-16356462

RESUMEN

The ubiquitin-proteasome pathway is the major nonlysosomal proteolytic system in eukaryotic cells responsible for regulating the level of many key regulatory molecules within the cells. Modification of cellular proteins by ubiquitin and ubiquitin-like proteins, such as small ubiquitin-like modifying protein (SUMO), plays an essential role in a number of biological schemes, and ubiquitin pathway enzymes have become important therapeutic targets. Ubiquitination is a dynamic reversible process; a multitude of ubiquitin ligases and deubiquitinases (DUBs) are responsible for the wide-ranging influence of this pathway as well as its selectivity. The DUB enzymes serve to maintain adequate pools of free ubiquitin and regulate the ubiquitination status of cellular proteins. Using SUMO fusions, a novel assay system, based on poliovirus RNA-dependent RNA polymerase activity, is described here. The method simplifies the isopeptidase assay and facilitates high-throughput analysis of these enzymes. The principle of the assay is the dependence of the viral polymerase on a free N terminus for activity; accordingly, the polymerase is inactive when fused at its N terminus to SUMO or any other ubiquitin-like protein. The assay is sensitive, reproducible, and adaptable to a high-throughput format for use in screens for inhibitors/activators of clinically relevant SUMO proteases and deubiquitinases.


Asunto(s)
Liasas de Carbono-Nitrógeno/análisis , ARN Polimerasas Dirigidas por ADN/metabolismo , Poliovirus/enzimología , Proteína SUMO-1/análisis , Ubiquitinas/análisis , Proteínas Recombinantes de Fusión/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA