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

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Cell Mol Life Sci ; 75(1): 161, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29022045

RESUMO

One of the author affiliations was missed to include in the original publication. The correct information is given below.

2.
Cell Mol Life Sci ; 74(9): 1625-1648, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-27866220

RESUMO

Macrophages play an essential role in the immune system by ingesting and degrading invading pathogens, initiating an inflammatory response and instructing adaptive immune cells, and resolving inflammation to restore homeostasis. More interesting is the fact that some bacteria have evolved to use macrophages as a natural habitat and tools of spread in the host, e.g., Mycobacterium tuberculosis (Mtb) and some non-tuberculous mycobacteria (NTM). Mtb is considered one of humanity's most successful pathogens and is the causal agent of tuberculosis, while NTMs cause opportunistic infections all of which are of significant public health concern. Here, we describe mechanisms by which intracellular pathogens, with an emphasis on mycobacteria, manipulate macrophage functions to circumvent killing and live inside these cells even under considerable immunological pressure. Such macrophage functions include the selective evasion or engagement of pattern recognition receptors, production of cytokines, reactive oxygen and nitrogen species, phagosome maturation, as well as other killing mechanisms like autophagy and cell death. A clear understanding of host responses elicited by a specific pathogen and strategies employed by the microbe to evade or exploit these is of significant importance for the development of effective vaccines and targeted immunotherapy against persistent intracellular infections like tuberculosis.


Assuntos
Macrófagos/microbiologia , Viabilidade Microbiana , Mycobacterium/citologia , Animais , Interações Hospedeiro-Patógeno , Humanos , Fagocitose , Fagossomos
3.
Proc Natl Acad Sci U S A ; 112(31): E4272-80, 2015 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-26195781

RESUMO

Several mechanisms are involved in controlling intracellular survival of pathogenic mycobacteria in host macrophages, but how these mechanisms are regulated remains poorly understood. We report a role for Kelch-like ECH-associated protein 1 (Keap1), an oxidative stress sensor, in regulating inflammation induced by infection with Mycobacterium avium in human primary macrophages. By using confocal microscopy, we found that Keap1 associated with mycobacterial phagosomes in a time-dependent manner, whereas siRNA-mediated knockdown of Keap1 increased M. avium-induced expression of inflammatory cytokines and type I interferons (IFNs). We show evidence of a mechanism whereby Keap1, as part of an E3 ubiquitin ligase complex with Cul3 and Rbx1, facilitates ubiquitination and degradation of IκB kinase (IKK)-ß thus terminating IKK activity. Keap1 knockdown led to increased nuclear translocation of transcription factors NF-κB, IFN regulatory factor (IRF) 1, and IRF5 driving the expression of inflammatory cytokines and IFN-ß. Furthermore, knockdown of other members of the Cul3 ubiquitin ligase complex also led to increased cytokine expression, further implicating this ligase complex in the regulation of the IKK family. Finally, increased inflammatory responses in Keap1-silenced cells contributed to decreased intracellular growth of M. avium in primary human macrophages that was reconstituted with inhibitors of IKKß or TANK-binding kinase 1 (TBK1). Taken together, we propose that Keap1 acts as a negative regulator for the control of inflammatory signaling in M. avium-infected human primary macrophages. Although this might be important to avoid sustained or overwhelming inflammation, our data suggest that a negative consequence could be facilitated growth of pathogens like M. avium inside macrophages.


Assuntos
Inflamação/patologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiologia , Mycobacterium avium/fisiologia , Transdução de Sinais , Proteínas de Transporte/metabolismo , Núcleo Celular/metabolismo , Células Cultivadas , Citocinas/biossíntese , Técnicas de Silenciamento de Genes , Humanos , Quinase I-kappa B/metabolismo , Fator Regulador 1 de Interferon/metabolismo , Fatores Reguladores de Interferon/metabolismo , Proteína 1 Associada a ECH Semelhante a Kelch , Mycobacterium avium/crescimento & desenvolvimento , NF-kappa B/metabolismo , Fagossomos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Estabilidade Proteica , Transporte Proteico , Proteólise , Espécies Reativas de Oxigênio/metabolismo , Transcrição Gênica , Tuberculose/imunologia , Tuberculose/metabolismo , Tuberculose/patologia , Ubiquitinação , Regulação para Cima
4.
J Biol Chem ; 285(29): 22576-91, 2010 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-20452972

RESUMO

The p62/SQSTM1 (sequestosome 1) protein, which acts as a cargo receptor for autophagic degradation of ubiquitinated targets, is up-regulated by various stressors. Induction of the p62 gene by oxidative stress is mediated by NF-E2-related factor 2 (NRF2) and, at the same time, p62 protein contributes to the activation of NRF2, but hitherto the mechanisms involved were not known. Herein, we have mapped an antioxidant response element (ARE) in the p62 promoter that is responsible for its induction by oxidative stress via NRF2. Chromatin immunoprecipitation and gel mobility-shift assays verified that NRF2 binds to this cis-element in vivo and in vitro. Also, p62 docks directly onto the Kelch-repeat domain of Kelch-like ECH-associated protein 1 (KEAP1), via a motif designated the KEAP1 interacting region (KIR), thereby blocking binding between KEAP1 and NRF2 that leads to ubiquitylation and degradation of the transcription factor. The KIR motif in p62 is located immediately C-terminal to the LC3-interacting region (LIR) and resembles the ETGE motif utilized by NRF2 for its interaction with KEAP1. KIR is required for p62 to stabilize NRF2, and inhibition of KEAP1 by p62 occurs from a cytoplasmic location within the cell. The LIR and KIR motifs cannot be engaged simultaneously by LC3 and KEAP1, but because p62 is polymeric the interaction between KEAP1 and p62 leads to accumulation of KEAP1 in p62 bodies, which is followed by autophagic degradation of KEAP1. Our data explain how p62 contributes to activation of NRF2 target genes in response to oxidative stress through creating a positive feedback loop.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Antioxidantes/metabolismo , Retroalimentação Fisiológica , Proteínas de Choque Térmico/genética , Fator 2 Relacionado a NF-E2/metabolismo , Elementos de Resposta/genética , Transcrição Gênica , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Sequência de Aminoácidos , Animais , Autofagia , Sequência de Bases , Sítios de Ligação , Ligação Competitiva , Regulação da Expressão Gênica , Células HeLa , Proteínas de Choque Térmico/química , Proteínas de Choque Térmico/metabolismo , Humanos , Corpos de Inclusão/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteína 1 Associada a ECH Semelhante a Kelch , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Dados de Sequência Molecular , Ligação Proteica , Processamento de Proteína Pós-Traducional , Proteína Sequestossoma-1 , Transdução de Sinais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA