Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Nature ; 628(8009): 878-886, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38509365

RESUMO

Targeted protein degradation and stabilization are promising therapeutic modalities because of their potency, versatility and their potential to expand the druggable target space1,2. However, only a few of the hundreds of E3 ligases and deubiquitinases in the human proteome have been harnessed for this purpose, which substantially limits the potential of the approach. Moreover, there may be other protein classes that could be exploited for protein stabilization or degradation3-5, but there are currently no methods that can identify such effector proteins in a scalable and unbiased manner. Here we established a synthetic proteome-scale platform to functionally identify human proteins that can promote the degradation or stabilization of a target protein in a proximity-dependent manner. Our results reveal that the human proteome contains a large cache of effectors of protein stability. The approach further enabled us to comprehensively compare the activities of human E3 ligases and deubiquitinases, identify and characterize non-canonical protein degraders and stabilizers and establish that effectors have vastly different activities against diverse targets. Notably, the top degraders were more potent against multiple therapeutically relevant targets than the currently used E3 ligases cereblon and VHL. Our study provides a functional catalogue of stability effectors for targeted protein degradation and stabilization and highlights the potential of induced proximity screens for the discovery of new proximity-dependent protein modulators.


Assuntos
Enzimas Desubiquitinantes , Estabilidade Proteica , Proteólise , Proteoma , Proteômica , Ubiquitina-Proteína Ligases , Humanos , Enzimas Desubiquitinantes/análise , Enzimas Desubiquitinantes/metabolismo , Proteoma/metabolismo , Ubiquitina-Proteína Ligases/análise , Ubiquitina-Proteína Ligases/metabolismo , Especificidade por Substrato , Quimera de Direcionamento de Proteólise/metabolismo , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo
2.
J Mol Biol ; 435(23): 168316, 2023 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-37858708

RESUMO

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.


Assuntos
Enzimas Desubiquitinantes , Citometria de Fluxo , Inibidores de Proteases , Humanos , Compostos de Anilina/farmacologia , Benzamidas/farmacologia , Enzimas Desubiquitinantes/análise , Enzimas Desubiquitinantes/antagonistas & inibidores , Doenças Neurodegenerativas/enzimologia , Ubiquitina/metabolismo , Ubiquitina Tiolesterase/antagonistas & inibidores , Peptidase 7 Específica de Ubiquitina/antagonistas & inibidores , Ubiquitinação/efeitos dos fármacos , Citometria de Fluxo/métodos , Inibidores de Proteases/química , Inibidores de Proteases/farmacologia , Proteases Semelhantes à Papaína de Coronavírus/análise , Proteases Semelhantes à Papaína de Coronavírus/antagonistas & inibidores , Anticorpos de Domínio Único
3.
Aging (Albany NY) ; 13(6): 8510-8523, 2021 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-33714206

RESUMO

Protein ubiquitination has been reported to be involved in many biological processes that affect cancer cell growth or death. In this study, we identified differentially expressed E3s/DUB-related genes associated with the prognosis of lung adenocarcinoma and then constructed an E3s/DUB enzyme signature prediction model for the training group and validated its accuracy for prognosis prediction in the validation group. According to our constructed model, all patients were divided into the high- or low-risk group, and a comparison of the two groups revealed that the high-risk group had poorer survival and higher mortality than the low-risk group. The calculated risk score was also an independent prognostic factor when analyzed together with other clinical factors. To explore the functions of the signature genes, we predicted the substrate proteins with which they interact and then performed enrichment analysis. Interestingly, we found that the signature genes were enriched in multiple treatment resistance and immune-related pathways. Therefore, we continued to analyze immune infiltration in the samples and found a variety of differences in immune cell infiltration. According to our constructed model, these differences in immune cell infiltration may predict different immune statuses after grouping and are associated with worse prognosis in high-risk patients.


Assuntos
Adenocarcinoma de Pulmão/enzimologia , Adenocarcinoma de Pulmão/genética , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/genética , Ubiquitinação/fisiologia , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Enzimas Desubiquitinantes/análise , Enzimas Desubiquitinantes/metabolismo , Humanos , Prognóstico , Transcriptoma , Ubiquitina-Proteína Ligases/análise , Ubiquitina-Proteína Ligases/metabolismo
4.
J Am Chem Soc ; 142(46): 19493-19501, 2020 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-33141564

RESUMO

Photocaged cell-permeable ubiquitin probe holds promise in profiling the activity of cellular deubiquitinating enzymes (DUBs) with the much needed temporal control. Here we report a new photocaged cell-permeable ubiquitin probe that undergoes photoactivation upon 365 nm UV treatment and enables intracellular deubiquitinating enzyme profiling. We used a semisynthetic approach to generate modular ubiquitin-based probe containing a tetrazole-derived warhead at the C-terminus of ubiquitin and employed a cyclic polyarginine cell-penetrating peptide (cR10) conjugated to the N-terminus of ubiquitin via a disulfide linkage to deliver the probe into live cells. Upon 365 nm UV irradiation, the tetrazole group is converted to a nitrilimine intermediate in situ, which reacts with nearby nucleophilic cysteine residue from the DUB active site. The new photocaged cell-permeable probe showed good reactivity toward purified DUBs, including USP2, UCHL1, and UCHL3, upon photoirradiation. The Ub-tetrazole probe was also assessed in HeLa cell lysate and showed robust labeling only upon photoactivation. We further carried out protein profiling in intact HeLa cells using the new photocaged cell-permeable ubiquitin probe and identified DUBs captured by the probe using label-free quantitative (LFQ) mass spectrometry. Importantly, the photocaged cell-permeable ubiquitin probe captured DUBs specifically in respective G1/S and G2/M phases in synchronized HeLa cells. Moreover, using this probe DUBs were profiled at different time points following the release of HeLa cells from G1/S phase. Our results showed that photocaged cell-permeable probe represents a valuable new tool for achieving a better understanding of the cellular functions of DUBs.


Assuntos
Enzimas Desubiquitinantes/análise , Sondas Moleculares/química , Ubiquitina/química , Domínio Catalítico , Permeabilidade da Membrana Celular , Reagentes de Ligações Cruzadas/química , Cisteína/química , Ativação Enzimática , Células HeLa , Humanos , Cinética , Espectrometria de Massas , Peptídeos/química , Processos Fotoquímicos , Exposição à Radiação , Fatores de Tempo , Raios Ultravioleta
5.
J Am Chem Soc ; 140(39): 12424-12433, 2018 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-30240200

RESUMO

Advancement in our knowledge of deubiquitinases (DUBs) and their biological functions requires biochemical tools permitting interrogation of DUB activities under physiologically relevant conditions. Activity-based DUB probes (DUB ABPs) have been widely used in investigating the function and activity of DUBs. However, most ubiquitin (Ub)-based DUB ABPs are not cell-permeable, limiting their utility to purified proteins and cell lysates. Lysis of cells usually leads to dilution of the cytoplasm and disruption of the normal cellular organization, which may alter the activity of many DUBs and DUB complexes. Here, we report a new class of cell-permeable DUB ABPs that enable intracellular DUB profiling. We used a semisynthetic approach to generate modular ubiquitin-based DUB probes containing a reactive warhead for covalent trapping of DUBs with a catalytic cysteine. We employed cell-penetrating peptides (CPPs), particualrly cyclic polyarginine (cR10), to deliver the DUB ABPs into cells, as confirmed using live-cell fluorescence microscopy and DUB ABPs containing a fluorophore at the C-terminus of Ub. In comparison to TAT, enhanced intacellular delivery was observed through conjugation of a cyclic polyarginine (cR10) to the N-terminus of ubiquitin via a disulfide linkage. Using the new cell-permeable DUB ABPs, we carried out DUB profiling in intact HeLa cells, and identified active DUBs using immunocapture and label-free quantitative mass spectrometry. Additionally, we demonstrated that the cell-permeable DUB ABPs can be used in assessing the inhibition of DUBs by small-molecule inhibitors in intact cells. Our results indicate that cell-permeable DUB ABPs hold great promise in providing a better understanding of the cellular functions of DUBs and advancing drug discovery efforts targeting human DUBs.


Assuntos
Enzimas Desubiquitinantes/metabolismo , Corantes Fluorescentes/química , Sondas Moleculares/química , Ubiquitinas/química , Permeabilidade da Membrana Celular , Enzimas Desubiquitinantes/análise , Enzimas Desubiquitinantes/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Corantes Fluorescentes/farmacocinética , Células HeLa , Humanos , Microscopia de Fluorescência , Sondas Moleculares/farmacocinética , Peptídeos/química , Peptídeos/farmacocinética , Ubiquitinas/farmacocinética
6.
Chem Commun (Camb) ; 53(73): 10208-10211, 2017 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-28857095

RESUMO

K27-linkage poly-ubiquitination plays important roles in DNA damage repair and autoimmunity. Identification of K27-linkage targeting deubiquitinases (DUBs) is essential for understanding their regulatory mechanisms. Here we report an aryl-azide-based photoaffinity diubiquitin (diUb) probe for profiling K27-linkage targeting DUBs. This probe shows high selectivity and sensitivity towards K27-linkage DUBs in vitro. It can also be used in cell lysate for identifying and probing K27-linkage targeting DUBs in real proteomes. Our work suggests that the photoaffinity-based strategy may provide a new useful approach for the construction of other isopeptide linkage targeting DUB probes.


Assuntos
Enzimas Desubiquitinantes/análise , Técnicas de Sonda Molecular , Marcadores de Fotoafinidade/química , Ubiquitina/química , Enzimas Desubiquitinantes/metabolismo , Células HEK293 , Humanos
7.
Methods Mol Biol ; 1513: 223-232, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27807841

RESUMO

Activity-based diubiquitin probes are highly useful in probing the deubiquitinase (DUB) activity and ubiquitin chain linkage specificity. Here we describe a detailed protocol to synthesize a new class of diubiquitin DUB probes. In this method, two ubiquitin moieties are connected through a linker resembling the native linkage in size and containing a Michael acceptor for trapping the DUB active-site cysteine. Detailed procedures for generating the linker molecule are also described.


Assuntos
Derivados de Benzeno/química , Técnicas de Química Sintética , Enzimas Desubiquitinantes/análise , Sondas Moleculares/síntese química , Ubiquitina/metabolismo , Domínio Catalítico , Cisteína/química , Enzimas Desubiquitinantes/metabolismo , Humanos , Mesna/química , Sondas Moleculares/química , Plasmídeos/química , Plasmídeos/metabolismo , Ubiquitinação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA