Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
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.

2.
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.

3.
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
4.
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.

5.
Comput Intell Neurosci ; 2022: 5019384, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36177309

RESUMEN

In this paper, we do research on cross-corpus speech emotion recognition (SER), in which the training and testing speech signals come from different speech corpus. The mismatched feature distribution between the training and testing sets makes many classical algorithms unable to achieve better results. To deal with this issue, a transfer learning and multi-loss dynamic adjustment (TLMLDA) algorithm is initiatively proposed in this paper. The proposed algorithm first builds a novel deep network model based on a deep auto-encoder and fully connected layers to improve the representation ability of features. Subsequently, global domain and subdomain adaptive algorithms are jointly adopted to implement features transfer. Finally, dynamic weighting factors are constructed to adjust the contribution of different loss functions to prevent optimization offset of model training, which effectively improve the generalization ability of the whole system. The results of simulation experiments on Berlin, eNTERFACE, and CASIA speech corpora show that the proposed algorithm can achieve excellent recognition results, and it is competitive with most of the state-of-the-art algorithms.


Asunto(s)
Emociones , Habla , Algoritmos , Aprendizaje , Aprendizaje Automático
6.
RSC Chem Biol ; 2(2): 450-467, 2021 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-34381999

RESUMEN

Ubiquitination is an important protein post-translational modification regulating many cellular processes in eukaryotes. Ubiquitination is catalyzed by a three-enzyme cascade resulting in the conjugation of the C-terminal carboxylate of ubiquitin (Ub) to the ε-amino group of a lysine residue in the acceptor protein via an isopeptide bond. In vitro enzymatic ubiquitination utilizing Ub ligases has been successfully employed to generate Ub dimers and polymers. However, limitations of the enzymatic approach exist, particularly due to the requirement of specific Ub ligase for any given target protein and the low catalytic efficiency of the Ub ligase. To achieve an in-depth understanding of the molecular mechanism of Ub signaling, new methods are needed to generate mono- and poly-ubiquitinated proteins at a specific site with defined polyubiquitin chain linkage and length. Chemical methods offer an attractive solution to the above-described challenges. In this review, we summarize the recently developed chemical methods for generating ubiquitinated proteins using synthetic and semisynthetic approaches. These new tools and approaches, as an important part of the Ub toolbox, are crucial to our understanding and exploitation of the Ub system for novel therapeutics.

7.
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
8.
Burns ; 47(5): 987-998, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33814214

RESUMEN

BACKGROUND: Triamcinolone acetonide (TAC) is widely used for hypertrophic scars and keloids; however, TAC has variable efficacy and safety in different individuals. PURPOSE: To evaluate the efficacy and safety of intralesional TAC for treatment of hypertrophic scars and keloids. DATA SOURCES: Searches of PubMed, EMBASE, the Cochrane Library, and ClinicalTrials.gov prior to 25 March 2020. STUDY SELECTION: Randomized controlled trials in English that compared TAC with a placebo or other medications that are commonly used for intralesional injection in hypertrophic scars and keloids. DATA EXTRACTION: Primary outcomes were reduction in scar height, vascularity, pliability, pigmentation, total scores on the Vancouver Scar Scale (VSS) or patient and observer scar assessment scale (POSAS), telangiectasia, and skin atrophy. Secondary outcomes included overall scar improvement. DATA SYNTHESIS: Fifteen trials met the inclusion criteria. In the short term, TAC was associated with a significant improvement in vascularity (MD: -0.22, 95% CI: -0.42 to -0.02) and pliability (MD: -0.25, 95% CI: -0.44 to -0.06) compared to verapamil. In the medium term, compared to TAC, 5-FU showed a significant improvement in scar height (SMD: 0.95, 95% CI: 0.15-1.75), while TAC led to a significant improvement in vascularity compared to 5-FU (MD: -0.45, 95% CI: -0.76 to -0.14). Compared to TAC, TAC+5-FU showed a significant improvement in pliability (SMD: 0.98, 95% CI: 0.17-1.78) and pigmentation (MD: 0.45, 95% CI: 0.12-0.78). Botulinum toxin type A resulted in significantly better pliability (SMD: 1.99, 95% CI: 0.98-3.00) compared to TAC. In the long term, compared to TAC, 5-FU led to a significant improvement in scar height (MD: 0.55, 95% CI: 0.17-0.93), but significantly less vascularity (MD: -0.35, 95% CI: -0.65 to -0.05). Compared to TAC, TAC+5-FU produced a significant improvement in scar height (MD: 1.50, 95% CI: 1.12-1.88), pliability (MD: 0.45, 95% CI: 0.10-0.80), and pigmentation (MD: 0.55, 95% CI: 0.24-0.86). CONCLUSION: TAC may be beneficial for the short-term treatment of hypertrophic scars and keloids; however, 5-FU, 5-FU+TAC, and verapamil may produce superior results for medium- and long-term treatments. TAC injections at concentrations of 20 mg/ml or 40 mg/ml are more likely to result in skin atrophy compared to 5-FU or verapamil, and are more likely to cause telangiectasia than 5-FU, 5-FU+TAC, or bleomycin.


Asunto(s)
Quemaduras , Cicatriz Hipertrófica , Queloide , Triamcinolona Acetonida , Atrofia , Cicatriz Hipertrófica/tratamiento farmacológico , Cicatriz Hipertrófica/patología , Medios de Cultivo , Fluorouracilo/uso terapéutico , Humanos , Inyecciones Intralesiones , Queloide/tratamiento farmacológico , Queloide/patología , Ensayos Clínicos Controlados Aleatorios como Asunto , Telangiectasia , Resultado del Tratamiento , Triamcinolona Acetonida/uso terapéutico , Verapamilo/uso terapéutico
9.
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
11.
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
12.
Chem Sci ; 9(40): 7859-7865, 2018 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-30429995

RESUMEN

Ubiquitination is an essential eukaryotic post-translational modification that regulates various cellular processes. The removal of ubiquitin from its target protein is catalyzed by deubiquitinating enzymes (DUBs). Although it was proposed that many DUBs specifically interact and recognize ubiquitinated proteins as substrates, more direct evidence is needed to support this notion. Here we report protein-targeting activity-based DUB probes that allowed the identification of DUBs recognizing monoubiquitinated proliferating cell nuclear antigen (PCNA) in Saccharomyces cerevisiae. This new class of DUB probes contain a Michael acceptor as a warhead between ubiquitin and the target protein PCNA through a linkage that mimics the native isopeptide bond. We selected two known and biologically relevant ubiquitination sites on PCNA to generate the DUB probes. This allowed us to interrogate the site-specific deubiquitination of a target protein by DUBs. DUBs were profiled in yeast cell lysates using the two Ub-PCNA DUB probes in conjunction with two control probes that contain a noncleavable linkage but no warhead. We identified yeast DUBs through pulldown coupled with quantitative mass spectrometry analysis of the pulled down proteins. Our results showed that specific yeast DUBs recognize monoubiquitinated PCNA and corroborated previous genetic study. We also identified DUBs as potential new deubiquitinase of PCNA. Remarkably, identified DUBs clearly distinguish the different modification sites on PCNA, thus supporting a high level of DUB specificity beyond the target protein identity.

13.
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
14.
ACS Chem Biol ; 12(9): 2399-2407, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28836754

RESUMEN

The deubiquitinases, or DUBs, are associated with various human diseases, including neurological disorders, cancer, and viral infection, making them excellent candidates for pharmacological intervention. Drug discovery campaigns against DUBs require enzymatic deubiquitination assays amenable for high-throughput screening (HTS). Although several DUB substrates and assays have been developed in recent years, they are largely limited to recombinantly purified DUBs. Many DUBs are large multidomain proteins that are difficult to obtain recombinantly in sufficient quantities for HTS. Therefore, an assay that obviates the need of recombinant protein generation and also recapitulates a physiologically relevant environment is highly desirable. Such an assay will open doors for drug discovery against many therapeutically relevant, but currently inaccessible, DUBs. Here, we report a cell lysate DUB assay based on AlphaLISA technology for high throughput screening. This assay platform uses a biotin-tagged ubiquitin probe and a HA-tagged DUB expressed in human cells. The assay was validated and adapted to a 1536-well format, which enabled a screening against UCHL1 as proof of principle using a library of 15 000 compounds. We expect that the new platform can be readily adapted to other DUBs to allow the identification of more potent and selective small molecule inhibitors and chemical probes.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Pruebas de Enzimas/métodos , Inhibidores Enzimáticos/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Ubiquitina Tiolesterasa/antagonistas & inhibidores , Células HEK293 , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Ubiquitina Tiolesterasa/metabolismo
15.
Methods Mol Biol ; 1513: 223-232, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27807841

RESUMEN

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.


Asunto(s)
Derivados del Benceno/química , Técnicas de Química Sintética , Enzimas Desubicuitinizantes/análisis , Sondas Moleculares/síntesis química , Ubiquitina/metabolismo , Dominio Catalítico , Cisteína/química , Enzimas Desubicuitinizantes/metabolismo , Humanos , Mesna/química , Sondas Moleculares/química , Plásmidos/química , Plásmidos/metabolismo , Ubiquitinación
16.
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
17.
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
18.
Structure ; 23(4): 724-733, 2015 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-25773143

RESUMEN

Proliferating cell nuclear antigen (PCNA) is a pivotal replication protein, which also controls cellular responses to DNA damage. Posttranslational modification of PCNA by SUMO and ubiquitin modulate these responses. How the modifiers alter PCNA-dependent DNA repair and damage tolerance pathways is largely unknown. We used hybrid methods to identify atomic models of PCNAK107-Ub and PCNAK164-SUMO consistent with small-angle X-ray scattering data of these complexes in solution. We show that SUMO and ubiquitin have distinct modes of association to PCNA. Ubiquitin adopts discrete docked binding positions. By contrast, SUMO associates by simple tethering and adopts extended flexible conformations. These structural differences are the result of the opposite electrostatic potentials of SUMO and Ub. The unexpected contrast in conformational behavior of Ub-PCNA and SUMO-PCNA has implications for interactions with partner proteins, interacting surfaces accessibility, and access points for pathway regulation.


Asunto(s)
Antígeno Nuclear de Célula en Proliferación/química , Proteína SUMO-1/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Sumoilación , Secuencia de Aminoácidos , Daño del ADN , Reparación del ADN , Simulación de Dinámica Molecular , Datos de Secuencia Molecular , Antígeno Nuclear de Célula en Proliferación/metabolismo , Unión Proteica , Proteína SUMO-1/química , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/enzimología , Proteínas de Saccharomyces cerevisiae/metabolismo
19.
J Med Chem ; 57(19): 8099-110, 2014 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-25229643

RESUMEN

Deregulation of ubiquitin conjugation or deconjugation has been implicated in the pathogenesis of many human diseases including cancer. The deubiquitinating enzyme USP1 (ubiquitin-specific protease 1), in association with UAF1 (USP1-associated factor 1), is a known regulator of DNA damage response and has been shown as a promising anticancer target. To further evaluate USP1/UAF1 as a therapeutic target, we conducted a quantitative high throughput screen of >400000 compounds and subsequent medicinal chemistry optimization of small molecules that inhibit the deubiquitinating activity of USP1/UAF1. Ultimately, these efforts led to the identification of ML323 (70) and related N-benzyl-2-phenylpyrimidin-4-amine derivatives, which possess nanomolar USP1/UAF1 inhibitory potency. Moreover, we demonstrate a strong correlation between compound IC50 values for USP1/UAF1 inhibition and activity in nonsmall cell lung cancer cells, specifically increased monoubiquitinated PCNA (Ub-PCNA) levels and decreased cell survival. Our results establish the druggability of the USP1/UAF1 deubiquitinase complex and its potential as a molecular target for anticancer therapies.


Asunto(s)
Antineoplásicos/síntesis química , Proteínas de Arabidopsis/antagonistas & inhibidores , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Proteínas Nucleares/antagonistas & inhibidores , Pirimidinas/síntesis química , Proteasas Ubiquitina-Específicas/antagonistas & inhibidores , Antineoplásicos/farmacología , Línea Celular Tumoral , Humanos , Antígeno Nuclear de Célula en Proliferación/metabolismo , Pirimidinas/farmacología , Relación Estructura-Actividad , Ubiquitinación
20.
ACS Chem Biol ; 9(8): 1685-91, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24918305

RESUMEN

Polyubiquitination of proteins regulates a variety of cellular processes, including protein degradation, NF-κB pathway activation, apoptosis, and DNA damage tolerance. Methods for generating polyubiquitinated protein with defined ubiquitin chain linkage and length are needed for an in-depth molecular understanding of protein polyubiquitination. However, enzymatic protein polyubiquitination usually generates polyubiquitinated proteins with mixed chain lengths in a low yield. We report herein a new chemical approach for protein polyubiquitination with a defined ubiquitin chain length and linkage under a mild condition that preserves the native fold of the target protein. In DNA damage tolerance, K63-polyubiquitinated proliferating cell nuclear antigen (PCNA) plays an important yet unclear role in regulating the selection of the error-free over error-prone lesion bypass pathways. Using the chemically polyubiquitinated PCNA, we revealed a mechanism of the K63 polyubiquitin chain on PCNA in promoting the error-free lesion bypass by suppressing the DNA translesion synthesis (TLS).


Asunto(s)
Daño del ADN , Poliubiquitina/química , Proteínas/química , Ubiquitinación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...