Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 121(31): e2403331121, 2024 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-39052835

RESUMEN

Androgen receptor (AR) is a main driver for castration-resistant prostate cancer (CRPC). c-Myc is an oncogene underlying prostate tumorigenesis. Here, we find that the deubiquitinase USP11 targets both AR and c-Myc in prostate cancer (PCa). USP11 expression was up-regulated in metastatic PCa and CRPC. USP11 knockdown (KD) significantly inhibited PCa cell growth. Our RNA-seq studies revealed AR and c-Myc as the top transcription factors altered after USP11 KD. ChIP-seq analysis showed that either USP11 KD or replacement of endogenous USP11 with a catalytic-inactive USP11 mutant significantly decreased chromatin binding by AR and c-Myc. We find that USP11 employs two mechanisms to up-regulate AR and c-Myc levels: namely, deubiquitination of AR and c-Myc proteins to increase their stability and deubiquitination of H2A-K119Ub, a repressive histone mark, on promoters of AR and c-Myc genes to increase their transcription. AR and c-Myc reexpression in USP11-KD PCa cells partly rescued cell growth defects. Thus, our studies reveal a tumor-promoting role for USP11 in aggressive PCa through upregulation of AR and c-Myc activities and support USP11 as a potential target against PCa.


Asunto(s)
Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Neoplasias de la Próstata , Proteínas Proto-Oncogénicas c-myc , Receptores Androgénicos , Tioléster Hidrolasas , Humanos , Masculino , Línea Celular Tumoral , Proliferación Celular/genética , Histonas/metabolismo , Regiones Promotoras Genéticas/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/patología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Receptores Androgénicos/metabolismo , Receptores Androgénicos/genética , Tioléster Hidrolasas/metabolismo , Tioléster Hidrolasas/genética , Ubiquitinación , Regulación hacia Arriba
2.
J Biol Chem ; : 107727, 2024 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-39214302

RESUMEN

Ubiquitin-specific proteases (USPs) are a family of multi-domain deubiquitinases (DUBs) with variable architectures, some containing regulatory auxiliary domains. Among the USP family, all occurrences of intramolecular regulation presently known are autoactivating. USP8 remains the sole exception as its putative WW-like domain, conserved only in vertebrate orthologs, is autoinhibitory. Here, we present a comprehensive structure-function analysis describing the autoinhibition of USP8 and provide evidence of the physical interaction between the WW-like and catalytic domains. The solution structure of full-length USP8 reveals an extended, monomeric conformation. Coupled with DUB assays, the WW-like domain is confirmed to be the minimal autoinhibitory unit. Strikingly, autoinhibition is only observed with the WW-like domain in cis and depends on the length of the linker tethering it to the catalytic domain. Modelling of the WW:CD complex structure and mutagenesis of interface residues suggests a novel binding site in the S1 pocket. To investigate the interplay between phosphorylation and USP8 autoinhibition, we identify AMP-activated protein kinase as a highly selective modifier of S718 in the 14-3-3 binding motif. We show that 14-3-3γ binding to phosphorylated USP8 potentiates autoinhibition in a WW-like domain-dependent manner by stabilizing an autoinhibited conformation. These findings provide mechanistic details on the autoregulation of USP8 and shed light on its evolutionary significance.

3.
Nucleic Acids Res ; 49(16): 9374-9388, 2021 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-34390346

RESUMEN

The Y-family DNA polymerase η (Polη) is critical for the synthesis past damaged DNA nucleotides in yeast through translesion DNA synthesis (TLS). TLS is initiated by monoubiquitination of proliferating cell nuclear antigen (PCNA) and the subsequent recruitment of TLS polymerases. Although individual structures of the Polη catalytic core and PCNA have been solved, a high-resolution structure of the complex of Polη/PCNA or Polη/monoubiquitinated PCNA (Ub-PCNA) still remains elusive, partly due to the disordered Polη C-terminal region and the flexibility of ubiquitin on PCNA. To circumvent these obstacles and obtain structural insights into this important TLS polymerase complex, we developed photo-activatable PCNA and Ub-PCNA probes containing a p-benzoyl-L-phenylalanine (pBpa) crosslinker at selected positions on PCNA. By photo-crosslinking the probes with full-length Polη, specific crosslinking sites were identified following tryptic digestion and tandem mass spectrometry analysis. We discovered direct interactions of the Polη catalytic core and its C-terminal region with both sides of the PCNA ring. Model building using the crosslinking site information as a restraint revealed multiple conformations of Polη in the polymerase complex. Availability of the photo-activatable PCNA and Ub-PCNA probes will also facilitate investigations into other PCNA-containing complexes important for DNA replication, repair and damage tolerance.


Asunto(s)
ADN Polimerasa Dirigida por ADN/genética , ADN/biosíntesis , Antígeno Nuclear de Célula en Proliferación/genética , Proteínas de Saccharomyces cerevisiae/genética , Ubiquitina/genética , Benzofenonas/farmacología , ADN/genética , Replicación del ADN/genética , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/ultraestructura , ADN Polimerasa Dirigida por ADN/química , ADN Polimerasa Dirigida por ADN/ultraestructura , Sustancias Macromoleculares/química , Sustancias Macromoleculares/ultraestructura , Mutación/genética , Fenilalanina/análogos & derivados , Fenilalanina/farmacología , Antígeno Nuclear de Célula en Proliferación/química , Antígeno Nuclear de Célula en Proliferación/ultraestructura , Unión Proteica/efectos de los fármacos , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/ultraestructura , Ubiquitina/química , Ubiquitina/ultraestructura
4.
Sensors (Basel) ; 23(18)2023 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-37766014

RESUMEN

Cloud observation serves as the fundamental bedrock for acquiring comprehensive cloud-related information. The categorization of distinct ground-based clouds holds profound implications within the meteorological domain, boasting significant applications. Deep learning has substantially improved ground-based cloud classification, with automated feature extraction being simpler and far more accurate than using traditional methods. A reengineering of the DenseNet architecture has given rise to an innovative cloud classification method denoted as CloudDenseNet. A novel CloudDense Block has been meticulously crafted to amplify channel attention and elevate the salient features pertinent to cloud classification endeavors. The lightweight CloudDenseNet structure is designed meticulously according to the distinctive characteristics of ground-based clouds and the intricacies of large-scale diverse datasets, which amplifies the generalization ability and elevates the recognition accuracy of the network. The optimal parameter is obtained by combining transfer learning with designed numerous experiments, which significantly enhances the network training efficiency and expedites the process. The methodology achieves an impressive 93.43% accuracy on the large-scale diverse dataset, surpassing numerous published methods. This attests to the substantial potential of the CloudDenseNet architecture for integration into ground-based cloud classification tasks.

5.
Entropy (Basel) ; 25(1)2023 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-36673265

RESUMEN

To solve the problem of feature distribution discrepancy in cross-corpus speech emotion recognition tasks, this paper proposed an emotion recognition model based on multi-task learning and subdomain adaptation, which alleviates the impact on emotion recognition. Existing methods have shortcomings in speech feature representation and cross-corpus feature distribution alignment. The proposed model uses a deep denoising auto-encoder as a shared feature extraction network for multi-task learning, and the fully connected layer and softmax layer are added before each recognition task as task-specific layers. Subsequently, the subdomain adaptation algorithm of emotion and gender features is added to the shared network to obtain the shared emotion features and gender features of the source domain and target domain, respectively. Multi-task learning effectively enhances the representation ability of features, a subdomain adaptive algorithm promotes the migrating ability of features and effectively alleviates the impact of feature distribution differences in emotional features. The average results of six cross-corpus speech emotion recognition experiments show that, compared with other models, the weighted average recall rate is increased by 1.89~10.07%, the experimental results verify the validity of the proposed model.

6.
Proc Natl Acad Sci U S A ; 116(15): 7288-7297, 2019 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-30914461

RESUMEN

USP9X is a conserved deubiquitinase (DUB) that regulates multiple cellular processes. Dysregulation of USP9X has been linked to cancers and X-linked intellectual disability. Here, we report the crystal structure of the USP9X catalytic domain at 2.5-Å resolution. The structure reveals a canonical USP-fold comprised of fingers, palm, and thumb subdomains, as well as an unusual ß-hairpin insertion. The catalytic triad of USP9X is aligned in an active configuration. USP9X is exclusively active against ubiquitin (Ub) but not Ub-like modifiers. Cleavage assays with di-, tri-, and tetraUb chains show that the USP9X catalytic domain has a clear preference for K11-, followed by K63-, K48-, and K6-linked polyUb chains. Using a set of activity-based diUb and triUb probes (ABPs), we demonstrate that the USP9X catalytic domain has an exo-cleavage preference for K48- and endo-cleavage preference for K11-linked polyUb chains. The structure model and biochemical data suggest that the USP9X catalytic domain harbors three Ub binding sites, and a zinc finger in the fingers subdomain and the ß-hairpin insertion both play important roles in polyUb chain processing and linkage specificity. Furthermore, unexpected labeling of a secondary, noncatalytic cysteine located on a blocking loop adjacent to the catalytic site by K11-diUb ABP implicates a previously unreported mechanism of polyUb chain recognition. The structural features of USP9X revealed in our study are critical for understanding its DUB activity. The new Ub-based ABPs form a set of valuable tools to understand polyUb chain processing by the cysteine protease class of DUBs.


Asunto(s)
Modelos Moleculares , Poliubiquitina/química , Ubiquitina Tiolesterasa/química , Cristalografía por Rayos X , Humanos , Poliubiquitina/metabolismo , Relación Estructura-Actividad , Especificidad por Sustrato , Ubiquitina Tiolesterasa/metabolismo
7.
J Am Chem Soc ; 142(46): 19493-19501, 2020 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-33141564

RESUMEN

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.


Asunto(s)
Enzimas Desubicuitinizantes/análisis , Sondas Moleculares/química , Ubiquitina/química , Dominio Catalítico , Permeabilidad de la Membrana Celular , Reactivos de Enlaces Cruzados/química , Cisteína/química , Activación Enzimática , Células HeLa , Humanos , Cinética , Espectrometría de Masas , Péptidos/química , Procesos Fotoquímicos , Exposición a la Radiación , Factores de Tiempo , Rayos Ultravioleta
8.
J Am Chem Soc ; 140(39): 12424-12433, 2018 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-30240200

RESUMEN

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.


Asunto(s)
Enzimas Desubicuitinizantes/metabolismo , Colorantes Fluorescentes/química , Sondas Moleculares/química , Ubiquitinas/química , Permeabilidad de la Membrana Celular , Enzimas Desubicuitinizantes/análisis , Enzimas Desubicuitinizantes/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Colorantes Fluorescentes/farmacocinética , Células HeLa , Humanos , Microscopía Fluorescente , Sondas Moleculares/farmacocinética , Péptidos/química , Péptidos/farmacocinética , Ubiquitinas/farmacocinética
9.
Biochemistry ; 55(33): 4708-19, 2016 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-27501351

RESUMEN

Deubiquitinating enzymes (DUBs) are responsible for reversing mono- and polyubiquitination of proteins and play essential roles in numerous cellular processes. Close to 100 human DUBs have been identified and are classified into five families, with the ubiquitin-specific protease (USP) family being the largest (>50 members). The binding of ubiquitin (Ub) to USP is strikingly different from that observed for the DUBs in the ubiquitin C-terminal hydrolase (UCH) and ovarian tumor domain protease (OTU) families. We generated a panel of mutant ubiquitins and used them to probe the ubiquitin's interaction with a number of USPs. Our results revealed a remarkable divergence of USP-Ub interactions among the USP catalytic domains. Our double-mutant cycle analysis targeting the ubiquitin residues located in the tip, the central body, and the tail of ubiquitin also demonstrated different crosstalk among the USP-Ub interactions. This work uncovered intriguing divergence in the ubiquitin-binding mode in the USP family DUBs and raised the possibility of targeting the ubiquitin-binding hot spots on USPs for selective inhibition of USPs by small molecule antagonists.


Asunto(s)
Péptido Hidrolasas/química , Ubiquitina/química , Catálisis , Mutagénesis Sitio-Dirigida , Péptido Hidrolasas/genética
10.
Chembiochem ; 17(11): 995-8, 2016 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-27113245

RESUMEN

We report a cysteine-based ligation strategy for generating a monoubiquitylated protein while preserving the native cysteine residues on the acceptor protein. In monoubiquitylation of proliferating cell nuclear antigen (PCNA) this method circumvents the need to mutate the native cysteine residues on PCNA. The chemically ubiquitylated PCNA contains a noncleavable linkage of the same length as the native isopeptide linkage. It also retains the normal function of the native Ub-PCNA in stimulating the ATPase activity of replication factor C (RFC) and lesion bypass synthesis by Polη. This method may be adapted for chemical ubiquitylation of other proteins and for site-specific modification of a target protein at a specific site through sulfhydryl chemistry.


Asunto(s)
Cisteína/química , Antígeno Nuclear de Célula en Proliferación/química , Cisteína/metabolismo , ADN Polimerasa Dirigida por ADN/metabolismo , Electroforesis en Gel de Poliacrilamida , Espectrometría de Masas , Antígeno Nuclear de Célula en Proliferación/metabolismo , Ubiquitinación , Rayos Ultravioleta
11.
Nat Chem Biol ; 10(4): 298-304, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24531842

RESUMEN

Protein ubiquitination and deubiquitination are central to the control of a large number of cellular pathways and signaling networks in eukaryotes. Although the essential roles of ubiquitination have been established in the eukaryotic DNA damage response, the deubiquitination process remains poorly defined. Chemical probes that perturb the activity of deubiquitinases (DUBs) are needed to characterize the cellular function of deubiquitination. Here we report ML323 (2), a highly potent inhibitor of the USP1-UAF1 deubiquitinase complex with excellent selectivity against human DUBs, deSUMOylase, deneddylase and unrelated proteases. Using ML323, we interrogated deubiquitination in the cellular response to UV- and cisplatin-induced DNA damage and revealed new insights into the requirement of deubiquitination in the DNA translesion synthesis and Fanconi anemia pathways. Moreover, ML323 potentiates cisplatin cytotoxicity in non-small cell lung cancer and osteosarcoma cells. Our findings point to USP1-UAF1 as a key regulator of the DNA damage response and a target for overcoming resistance to the platinum-based anticancer drugs.


Asunto(s)
Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Proteínas de Arabidopsis/antagonistas & inhibidores , Daño del ADN/fisiología , Proteínas Nucleares/antagonistas & inhibidores , Proteasas Ubiquitina-Específicas/antagonistas & inhibidores , Ubiquitinación/efectos de los fármacos , Algoritmos , Butiratos/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cisplatino/farmacología , Ensayo de Unidades Formadoras de Colonias , Daño del ADN/genética , ADN de Neoplasias/antagonistas & inhibidores , ADN de Neoplasias/biosíntesis , Resistencia a Antineoplásicos , Electroforesis en Gel de Poliacrilamida , Anemia de Fanconi/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/antagonistas & inhibidores , Ensayos Analíticos de Alto Rendimiento , Humanos , Indicadores y Reactivos , Compuestos de Fenilurea/farmacología , Pimozida/farmacología , Antígeno Nuclear de Célula en Proliferación/efectos de los fármacos , Antígeno Nuclear de Célula en Proliferación/metabolismo , ARN Interferente Pequeño/genética , Proteínas Recombinantes/química , Recombinación Genética/efectos de los fármacos , Intercambio de Cromátides Hermanas/efectos de los fármacos
12.
Nucleic Acids Res ; 40(21): 10795-808, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22987070

RESUMEN

Unrepaired DNA damage may arrest ongoing replication forks, potentially resulting in fork collapse, increased mutagenesis and genomic instability. Replication through DNA lesions depends on mono- and polyubiquitylation of proliferating cell nuclear antigen (PCNA), which enable translesion synthesis (TLS) and template switching, respectively. A proper replication fork rescue is ensured by the dynamic ubiquitylation and deubiquitylation of PCNA; however, as yet, little is known about its regulation. Here, we show that human Spartan/C1orf124 protein provides a higher cellular level of ubiquitylated-PCNA by which it regulates the choice of DNA damage tolerance pathways. We find that Spartan is recruited to sites of replication stress, a process that depends on its PCNA- and ubiquitin-interacting domains and the RAD18 PCNA ubiquitin ligase. Preferential association of Spartan with ubiquitin-modified PCNA protects against PCNA deubiquitylation by ubiquitin-specific protease 1 and facilitates the access of a TLS polymerase to the replication fork. In concert, depletion of Spartan leads to increased sensitivity to DNA damaging agents and causes elevated levels of sister chromatid exchanges. We propose that Spartan promotes genomic stability by regulating the choice of rescue of stalled replication fork, whose mechanism includes its interaction with ubiquitin-conjugated PCNA and protection against PCNA deubiquitylation.


Asunto(s)
Daño del ADN , Proteínas de Unión al ADN/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Ubiquitina/metabolismo , Proteínas de Arabidopsis , Línea Celular , Replicación del ADN , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/fisiología , ADN Polimerasa Dirigida por ADN/metabolismo , Endopeptidasas/metabolismo , Humanos , Estructura Terciaria de Proteína , Intercambio de Cromátides Hermanas , Ubiquitina-Proteína Ligasas/fisiología , Proteasas Ubiquitina-Específicas
13.
Proc Natl Acad Sci U S A ; 108(43): 17672-7, 2011 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-22006297

RESUMEN

PCNA ubiquitination in response to DNA damage leads to the recruitment of specialized translesion polymerases to the damage locus. This constitutes one of the initial steps in translesion synthesis (TLS)--a critical pathway for cell survival and for maintenance of genome stability. The recent crystal structure of ubiquitinated PCNA (Ub-PCNA) sheds light on the mode of association between the two proteins but also revealed that paradoxically, the ubiquitin surface engaged in PCNA interactions was the same as the surface implicated in translesion polymerase binding. This finding implied a degree of flexibility inherent in the Ub-PCNA complex that would allow it to transition into a conformation competent to bind the TLS polymerase. To address the issue of segmental flexibility, we combined multiscale computational modeling and small angle X-ray scattering. This combined strategy revealed alternative positions for ubiquitin to reside on the surface of the PCNA homotrimer, distinct from the position identified in the crystal structure. Two mutations originally identified in genetic screens and known to interfere with TLS are positioned directly beneath the bound ubiquitin in the alternative models. These computationally derived positions, in an ensemble with the crystallographic and flexible positions, provided the best fit to the solution scattering, indicating that ubiquitin dynamically associated with PCNA and is capable of transitioning between a few discrete sites on the PCNA surface. The finding of new docking sites and the positional equilibrium of PCNA-Ub occurring in solution provide unexpected insight into previously unexplained biological observations.


Asunto(s)
Sustancias Macromoleculares/química , Modelos Moleculares , Antígeno Nuclear de Célula en Proliferación/química , Unión Proteica , Conformación Proteica , Ubiquitina/química , Biología Computacional , Humanos , Dispersión del Ángulo Pequeño
14.
ACS Pharmacol Transl Sci ; 7(9): 2827-2839, 2024 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-39296272

RESUMEN

Ubiquitination is a post-translational modification that elicits a variety of cellular responses. Deubiquitinases (DUBs) remove ubiquitin moieties from proteins and modulate cellular processes by counteracting the ubiquitin ligase activities. Ubiquitination and deubiquitination processes are tightly regulated by different mechanisms and their dysregulation is associated with many diseases. Discovery of DUB inhibitors could not only lead to therapeutics but also facilitate the understanding of ubiquitination/deubiquitination processes and their regulatory mechanisms. To enable the inhibitor discovery against DUBs, we developed a cell-based DUB assay that utilizes a cell-permeable ubiquitin probe, Biotin-cR10-Ub-PA, to covalently label DUBs in their native cellular environment. Amplified luminescent proximity homogeneous assay (Alpha, specifically AlphaLISA) is utilized to quantitatively assess the capture of the target DUB by the Biotin-cR10-Ub-PA probe. We demonstrated that this new cell-based DUB assay is robust and amenable to high-throughput screening. Human USP15 was selected as a DUB of interest and screened against a library of protease inhibitors as a proof of concept. In addition to the widely adopted pan-DUB inhibitor PR-619, several other DUB inhibitors from the library were also identified as hits. This new DUB assay can be readily adapted for inhibitor discovery against many other human DUBs to identify potent and cell-permeable inhibitors.

15.
Cell Rep Methods ; 4(6): 100796, 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38866007

RESUMEN

We present an efficient, effective, and economical approach, named E3technology, for proteomics sample preparation. By immobilizing silica microparticles into the polytetrafluoroethylene matrix, we develop a robust membrane medium, which could serve as a reliable platform to generate proteomics-friendly samples in a rapid and low-cost fashion. We benchmark its performance using different formats and demonstrate them with a variety of sample types of varied complexity, quantity, and volume. Our data suggest that E3technology provides proteome-wide identification and quantitation performance equivalent or superior to many existing methods. We further propose an enhanced single-vessel approach, named E4technology, which performs on-filter in-cell digestion with minimal sample loss and high sensitivity, enabling low-input and low-cell proteomics. Lastly, we utilized the above technologies to investigate RNA-binding proteins and profile the intact bacterial cell proteome.


Asunto(s)
Proteoma , Proteómica , Proteómica/métodos , Proteoma/análisis , Proteoma/metabolismo , Dióxido de Silicio/química , Politetrafluoroetileno
16.
Biochemistry ; 52(19): 3217-28, 2013 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-23634825

RESUMEN

Although often associated with proteasome-mediated protein degradation, ubiquitin plays essential nondegradative roles in a myriad of cellular processes, including chromatin dynamics, membrane trafficking, innate immunity, and DNA damage response. The recent progress in understanding DNA translesion synthesis (TLS), an important branch of DNA damage response, has largely been stimulated by the finding that ubiquitination of an essential nuclear protein, proliferating cell nuclear antigen (PCNA), controls precisely how eukaryotic cells respond to DNA damage. Despite the remarkable activity of the TLS polymerases in synthesizing past the damaged nucleotides, they are intrinsically error-prone on the normal DNA template. Therefore, a stringent regulation of the TLS polymerases is essential for the faithful replication of the DNA genome. Here we review the structure and function of the Y-family TLS polymerases and their interactions with ubiquitin and monoubiquitinated PCNA (Ub-PCNA). Driven by the need for monoubiquitinated PCNA in a sufficient quantity and purity, researchers developed both chemical and enzymatic methods for PCNA monoubiquitination, which have propelled our understanding of the structure of Ub-PCNA by X-ray crystallography and small-angle X-ray scattering. Together with studies using a reconstituted polymerase switching assay, these investigations revealed a surprising conformational flexibility of ubiquitin as a modifier on PCNA. Although the molecular details of TLS in cells still need to be deciphered, two working models, polymerase switching and postreplicative gap filling, have been proposed and tested in both in vitro and cellular systems. Evidence for both models is discussed herein. Compared to PCNA monoubiquitination, polyubiquitination of PCNA in DNA damage response is much less well understood and will be the subject of a future investigation. Given the close connection of DNA damage response and anticancer therapy, an in-depth understanding of the eukaryotic translesion synthesis and its regulation by ubiquitin will likely provide new opportunities for therapeutic intervention.


Asunto(s)
Daño del ADN , Reparación del ADN/fisiología , Ubiquitina/metabolismo , Secuencia de Aminoácidos , Animales , ADN Polimerasa Dirigida por ADN/química , ADN Polimerasa Dirigida por ADN/genética , ADN Polimerasa Dirigida por ADN/metabolismo , Humanos , Modelos Biológicos , Modelos Moleculares , Datos de Secuencia Molecular , Antígeno Nuclear de Célula en Proliferación/química , Antígeno Nuclear de Célula en Proliferación/metabolismo , Conformación Proteica , Procesamiento Proteico-Postraduccional , Homología de Secuencia de Aminoácido , Ubiquitinación
17.
ACS Chem Biol ; 18(4): 837-847, 2023 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-36972492

RESUMEN

The important roles played by branched polyubiquitin chains were recently uncovered in proteasomal protein degradation, mitotic regulation, and NF-κB signaling. With the new realization of a wide presence of branched ubiquitin chains in mammalian cells, there is an urgent need of identifying the reader and eraser proteins of the various branched ubiquitin chains. In this work, we report the generation of noncleavable branched triubiquitin probes with combinations of K11-, K48-, and K63-linkages. Through a pulldown approach using the branched triUb probes, we identified human proteins that recognize branched triubiquitin structures including ubiquitin-binding proteins and deubiquitinases (DUBs). Proteomics analysis of the identified proteins enriched by the branched triubiquitin probes points to possible roles of branched ubiquitin chains in cellular processes including DNA damage response, autophagy, and receptor endocytosis. In vitro characterization of several identified UIM-containing proteins demonstrated their binding to branch triubiquitin chains with moderate to high affinities. Availability of this new class of branched triubiquitin probes will enable future investigation into the roles of branched polyubiquitin chains through identification of specific reader and eraser proteins, and the modes of branched ubiquitin chain recognition and processing using biochemical and biophysical methods.


Asunto(s)
Sondas Moleculares , Poliubiquitina , Ubiquitina , Humanos , Proteínas Portadoras/metabolismo , Poliubiquitina/química , Poliubiquitina/metabolismo , Unión Proteica , Transducción de Señal , Ubiquitina/química , Ubiquitina/metabolismo , Ubiquitinación , Sondas Moleculares/química
18.
Res Sq ; 2023 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-37502920

RESUMEN

Proteomics experiments have typically high economic and technical barriers to broad biomedical scientists, which not only result in costly supplies and accessories for sample preparation but also the reluctance to adapt new techniques. In the present study, we present an effective and efficient, yet economical technology, which we call E3technology, for proteomics sample preparation. By immobilizing silica microparticles into a polytetrafluoroethylene (PTFE) matrix, we developed a novel medium, which could be used as a robust and reliable proteomics platform to generate LCMS-friendly samples in a rapid and low-cost fashion. Using different formats of E3technology, including E3tip, E3filter, E3cartridge, and E3plate, we explored a variety of sample types in varied complexity, quantity, volume, and size, including bacterial, fungi, mammalian cells, mouse tissue, and human body fluids. We benchmark their performance against several established approaches. Our data suggest that E3technology outperforms many of the currently available techniques in terms of proteome identification and quantitation. It is widely applicable, highly reproducible, readily scalable and automatable, and is user-friendly and stress-free to non-expert proteomics laboratories. It does not require specialized expertise and equipment, and significantly lowers the technical and economical barrier to proteomics experiments. An enhanced version, E4technology, also opens new avenues to sample preparation for low input and/or low-cell proteomics analysis. The presented technologies by our study represent a breakthrough innovation in biomedical science, and we anticipate widespread adoption by the proteomics community.

19.
Biochemistry ; 51(13): 2829-39, 2012 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-22439892

RESUMEN

Ubiquitin-specific proteases (USPs) constitute the largest family of the human deubiquitinating enzymes. USP1 belongs to the cysteine protease family and contains a catalytic triad comprised of C90, H593, and D751. Notably, the catalytic activity of USP1 is stimulated through the formation of a tight complex with a WD40 repeat protein UAF1 (USP1-associated factor 1). Our kinetic analyses revealed a general base catalysis in USP1/UAF1, in contrast to an ion-pair mechanism as demonstrated for papain and cathepsin. The pK(a) value of the catalytic cysteine was determined to be 8.67 ± 0.07 in a pH-dependent inactivation study of USP1/UAF1 by iodoacetamide. A normal solvent kinetic isotope effect of 2.8 for k(cat) and 3.0 for k(cat)/K(m) was observed in the USP1/UAF1-catalyzed hydrolysis of ubiquitin-AMC substrate. Moreover, proton inventory analysis supported the transfer of a single solvent-derived proton in the transition state. Our study also revealed the molecular basis for the activation of USP1 by UAF1. Although the pK(a) of the catalytic cysteine in USP1 and USP1/UAF1 was almost identical, the pK(a) of the catalytic histidine in USP1/UAF1 was 0.43 pH unit lower than that in USP1, which facilitates general base catalysis at a neutral pH and contributes to the elevated catalytic efficiency. We ruled out that the higher catalytic efficiency is due to a tighter binding of ubiquitin. Our results support a regulatory mechanism in which UAF1 activates USP1 by modulating its active site conformation. This finding has a general implication for the regulation of USPs that form complex with partner proteins.


Asunto(s)
Proteínas Nucleares/metabolismo , Biocatálisis , Dominio Catalítico , Concentración de Iones de Hidrógeno , Hidrólisis , Cinética , Proteínas Nucleares/antagonistas & inhibidores , Cloruro de Sodio/química , Ubiquitina/farmacología
20.
Biochemistry ; 51(50): 10075-86, 2012 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-23211065

RESUMEN

Deubiquitination has emerged as an essential regulatory mechanism of a number of cellular processes. An in-depth understanding of deubiquitinating enzyme (DUB) catalysis, particularly the mode of ubiquitin binding and the individual steps in the DUB catalytic turnover, is imperative for exploiting DUBs for therapeutic intervention. In this work, we present a transient kinetic study of USP2 in hydrolyzing a model substrate Ub-AMC and a physiological substrate K48-linked diubiquitin. We conducted stopped-flow fluorescence analyses of the binding of mono- and diubiquitin to an inactive USP2 mutant and unveiled interesting differences in the binding kinetics between the two substrates. While a simple one-step binding of monoubiquitin to USP2 was observed, a biphasic binding was evident for diubiquitin. We further followed the deubiquitination reaction of Ub-AMC and K48-linked IQF-diubiquitin by USP2 using stopped-flow florescence under a single-turnover condition. Global fitting of the reaction traces revealed differences in the microscopic rate constants between Ub-AMC and the physiological diubiquitin substrate. Our binding and single-turnover data support a conformational rearrangement of the diubiquitin substrate in USP2-catalyzed deubiquitination. This finding is significant given the recent finding that the K48-linked diubiquitin is dynamic in its conformation. Our results provide useful insights into the mechanism of how USP recognizes ubiquitin moieties in a chain structure, which is important for understanding USP catalysis and developing inhibitors against USPs.


Asunto(s)
Endopeptidasas/metabolismo , Ubiquitinas/metabolismo , Cumarinas/metabolismo , Humanos , Cinética , Conformación Proteica , Ubiquitina Tiolesterasa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA