Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 16(11): e0248668, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34767572

RESUMO

BACKGROUND: The NLRP3 inflammasome is a critical component of sterile inflammation, which is involved in many diseases. However, there is currently no known proximal biomarker for measuring NLRP3 activation in pathological conditions. Protein kinase D (PKD) has emerged as an important NLRP3 kinase that catalyzes the release of a phosphorylated NLRP3 species that is competent for inflammasome complex assembly. METHODS: To explore the potential for PKD activation to serve as a selective biomarker of the NLRP3 pathway, we tested various stimulatory conditions in THP-1 and U937 cell lines, probing the inflammasome space beyond NLRP3. We analyzed the correlation between PKD activation (monitored by its auto-phosphorylation) and functional inflammasome readouts. RESULTS: PKD activation/auto-phosphorylation always preceded cleavage of caspase-1 and gasdermin D, and treatment with the PKD inhibitor CRT0066101 could block NLRP3 inflammasome assembly and interleukin-1ß production. Conversely, blocking NLRP3 either genetically or using the MCC950 inhibitor prevented PKD auto-phosphorylation, indicating a bidirectional functional crosstalk between NLRP3 and PKD. Further assessments of the pyrin and NLRC4 pathways, however, revealed that PKD auto-phosphorylation can be triggered by a broad range of stimuli unrelated to NLRP3 inflammasome assembly. CONCLUSION: Although PKD and NLRP3 become functionally interconnected during NLRP3 activation, the promiscuous reactivity of PKD challenges its potential use for tracing the NLRP3 inflammasome pathway.


Assuntos
Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Proteína Quinase C/metabolismo , Biomarcadores/metabolismo , Caspase 1/metabolismo , Linhagem Celular Tumoral , Humanos , Inflamação/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Ligação a Fosfato/metabolismo , Fosforilação , Pirina/metabolismo , Células U937
2.
Proc Natl Acad Sci U S A ; 108(52): 21052-6, 2011 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-22160684

RESUMO

Hepatitis C virus (HCV) infection is a global health burden with over 170 million people infected worldwide. In a significant portion of patients chronic hepatitis C infection leads to serious liver diseases, including fibrosis, cirrhosis, and hepatocellular carcinoma. The HCV NS3 protein is essential for viral polyprotein processing and RNA replication and hence viral replication. It is composed of an N-terminal serine protease domain and a C-terminal helicase/NTPase domain. For full activity, the protease requires the NS4A protein as a cofactor. HCV NS3/4A protease is a prime target for developing direct-acting antiviral agents. First-generation NS3/4A protease inhibitors have recently been introduced into clinical practice, markedly changing HCV treatment options. To date, crystal structures of HCV NS3/4A protease inhibitors have only been reported in complex with the protease domain alone. Here, we present a unique structure of an inhibitor bound to the full-length, bifunctional protease-helicase NS3/4A and show that parts of the P4 capping and P2 moieties of the inhibitor interact with both protease and helicase residues. The structure sheds light on inhibitor binding to the more physiologically relevant form of the enzyme and supports exploring inhibitor-helicase interactions in the design of the next generation of HCV NS3/4A protease inhibitors. In addition, small angle X-ray scattering confirmed the observed protease-helicase domain assembly in solution.


Assuntos
Proteínas de Transporte/química , Hepatite C/tratamento farmacológico , Modelos Moleculares , Inibidores de Proteases/química , Conformação Proteica , Proteínas não Estruturais Virais/química , Proteínas de Transporte/metabolismo , Cromatografia em Gel , Cristalização , Cristalografia por Raios X , Escherichia coli , Concentração Inibidora 50 , Peptídeos e Proteínas de Sinalização Intracelular , Inibidores de Proteases/metabolismo , Inibidores de Proteases/uso terapêutico , Espalhamento a Baixo Ângulo , Proteínas não Estruturais Virais/metabolismo
3.
Anal Biochem ; 371(2): 201-7, 2007 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-17869210

RESUMO

The dynamic modification of proteins with ubiquitin is a key regulation paradigm in eukaryotic cells that controls stability, localization, and function of the vast majority of intracellular proteins. Here we describe a robust fluorescence intensity assay for monitoring the enzymatic activity of deubiquitinating proteases, which reverse ubiquitin modifications and comprise over 100 members in humans. The assay was developed for the catalytic domain of human ubiquitin-specific protease 2 (USP2) and human ubiquitin carboxyterminal hydrolase L3 (UCH-L3), and makes use of the novel substrate ubiquitin-rhodamine110-glycine. The latter combines the advantages of a high dynamic range and beneficial optical properties. Its enzymatic behavior is characterized by the kinetic constants K(m)=1.5 microM, k(cat) = 0.53s(-1) and k(cat)/K(m) = 3.5 x 10(5)M(-1) s(-1) for USP2 and K(m) = 34 nM, k(cat)=4.72s(-1), and k(cat)/K(m) = 1.4 x 10(8)M(-1) s(-1) for UCH-L3. This new assay is suitable for inhibitor screening and characterizations, and has been established for the 384-well plate format using protease concentrations of 120 pM for USP2 and 1 pM for UCH-L3 and substrate concentrations of 100 nM for both enzymes. Due to the low protease concentrations and high sensitivity, this assay would allow the determination of inhibitory constants in the subnanomolar range.


Assuntos
Cisteína Endopeptidases/metabolismo , Endopeptidases/metabolismo , Corantes Fluorescentes/química , Glicina/análogos & derivados , Glicina/química , Ubiquitina/química , Bioensaio/instrumentação , Bioensaio/métodos , Cisteína Endopeptidases/química , Endopeptidases/química , Corantes Fluorescentes/metabolismo , Glicina/metabolismo , Humanos , Inteínas , Cinética , Modelos Biológicos , Rodaminas/química , Rodaminas/metabolismo , Espectrometria de Fluorescência , Especificidade por Substrato , Ubiquitina/metabolismo , Ubiquitina Tiolesterase
4.
J Neurochem ; 80(6): 1079-88, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11953458

RESUMO

beta-Site APP-cleaving enzyme (BACE) initiates the processing of the amyloid precursor protein (APP) leading to the generation of beta-amyloid, the main component of Alzheimer's disease senile plaques. BACE (Asp2, memapsin 2) is a type I transmembrane aspartic protease responsible for the beta-secretase cleavage of APP producing a soluble form of the ectodomain (sAPPbeta) and the membrane-bound, carboxy-terminal intermediates C99 and C89. BACE maturation involves cysteine bridge formation, N -glycosylation and propeptide removal. We investigated variants of BACE in which the disulphide bonds of the catalytic domain spanning between Cys216/Cys420, Cys278/Cys443 and Cys330/Cys380 were removed by mutagenesis. When transfected in cultured cells, these mutants showed impaired maturation. Nevertheless, a fraction of mutated protein retained both the competence to mature as well as the activity to process APP. For the generation of a functional enzyme the conserved Cys330/Cys380 bond was the most critical, whereas the two bonds between Cys216/Cys420 and Cys278/Cys443, which are typical for the membrane-bound BACE, appeared to be less important.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Ácido Aspártico Endopeptidases/química , Ácido Aspártico Endopeptidases/metabolismo , Domínio Catalítico/fisiologia , Dissulfetos/química , Doença de Alzheimer/enzimologia , Secretases da Proteína Precursora do Amiloide , Precursor de Proteína beta-Amiloide/genética , Animais , Ácido Aspártico Endopeptidases/genética , Brefeldina A/farmacologia , Células COS/metabolismo , Linhagem Celular , Endopeptidases , Ativação Enzimática/efeitos dos fármacos , Glicosilação , Complexo de Golgi/metabolismo , Hexosaminidases/metabolismo , Humanos , Rim/metabolismo , Mutagênese Sítio-Dirigida , Dobramento de Proteína , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Inibidores da Síntese de Proteínas/farmacologia , Transporte Proteico , Relação Estrutura-Atividade , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA