Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Mol Microbiol ; 90(5): 923-38, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24102920

RESUMO

Each Pseudomonas aeruginosa cell localizes two types of motility structures, a single flagellum and one or two clusters of type IV pili, to the cell poles. Previous studies suggested that these motility structures arrive at the pole through distinct mechanisms. Here we performed a swimming motility screen to identify polar flagellum localization factors and discovered three genes homologous to the TonB/ExbB/ExbD complex that have defects in both flagella-mediated swimming and pilus-mediated twitching motility. We found that deletion of tonB3, PA2983 or PA2982 led to non-polar localization of the flagellum and FlhF, which was thought to sit at the top of the flagellar localization hierarchy. Surprisingly, these mutants also exhibited pronounced changes in pilus formation or localization, indicating that these proteins may co-ordinate both the pilus and flagellum motility systems. Thus, we have renamed PA2983 and PA2982, pocA and pocB, respectively, for polar organelle co-ordinator to reflect this function. Our results suggest that TonB3, PocA and PocB may form a membrane-associated complex, which we term the Poc complex. These proteins do not exhibit polar localization themselves, but are required for increased expression of pilus genes upon surface association, indicating that they regulate motility structures through either localization or transcriptional mechanisms.


Assuntos
Proteínas de Bactérias/metabolismo , Fímbrias Bacterianas/fisiologia , Flagelos/fisiologia , Proteínas de Membrana/metabolismo , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Pseudomonas aeruginosa/fisiologia , Proteínas de Bactérias/genética , Fímbrias Bacterianas/genética , Flagelos/genética , Regulação Bacteriana da Expressão Gênica , Proteínas de Membrana/genética , Microscopia Eletrônica de Transmissão , Proteínas Monoméricas de Ligação ao GTP/genética , Movimento , Pseudomonas aeruginosa/genética , Deleção de Sequência
2.
PLoS Pathog ; 8(4): e1002661, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22532801

RESUMO

The cell intrinsic innate immune responses provide a first line of defense against viral infection, and often function by targeting cellular pathways usurped by the virus during infection. In particular, many viruses manipulate cellular lipids to form complex structures required for viral replication, many of which are dependent on de novo fatty acid synthesis. We found that the energy regulator AMPK, which potently inhibits fatty acid synthesis, restricts infection of the Bunyavirus, Rift Valley Fever Virus (RVFV), an important re-emerging arthropod-borne human pathogen for which there are no effective vaccines or therapeutics. We show restriction of RVFV both by AMPK and its upstream activator LKB1, indicating an antiviral role for this signaling pathway. Furthermore, we found that AMPK is activated during RVFV infection, leading to the phosphorylation and inhibition of acetyl-CoA carboxylase, the first rate-limiting enzyme in fatty acid synthesis. Activating AMPK pharmacologically both restricted infection and reduced lipid levels. This restriction could be bypassed by treatment with the fatty acid palmitate, demonstrating that AMPK restricts RVFV infection through its inhibition of fatty acid biosynthesis. Lastly, we found that this pathway plays a broad role in antiviral defense since additional viruses from disparate families were also restricted by AMPK and LKB1. Therefore, AMPK is an important component of the cell intrinsic immune response that restricts infection through a novel mechanism involving the inhibition of fatty acid metabolism.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Ácidos Graxos/biossíntese , Febre do Vale de Rift/enzimologia , Vírus da Febre do Vale do Rift/metabolismo , Quinases Proteína-Quinases Ativadas por AMP , Proteínas Quinases Ativadas por AMP/imunologia , Acetil-CoA Carboxilase/metabolismo , Animais , Linhagem Celular , Cricetinae , Humanos , Imunidade Inata/fisiologia , Camundongos , Proteínas Serina-Treonina Quinases/metabolismo , Febre do Vale de Rift/imunologia , Vírus da Febre do Vale do Rift/imunologia
3.
PLoS Pathog ; 6(6): e1000954, 2010 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-20585561

RESUMO

Poxviruses include medically important human pathogens, yet little is known about the specific cellular factors essential for their replication. To identify genes essential for poxvirus infection, we used high-throughput RNA interference to screen the Drosophila kinome for factors required for vaccinia infection. We identified seven genes including the three subunits of AMPK as promoting vaccinia infection. AMPK not only facilitated infection in insect cells, but also in mammalian cells. Moreover, we found that AMPK is required for macropinocytosis, a major endocytic entry pathway for vaccinia. Furthermore, we show that AMPK contributes to other virus-independent actin-dependent processes including lamellipodia formation and wound healing, independent of the known AMPK activators LKB1 and CaMKK. Therefore, AMPK plays a highly conserved role in poxvirus infection and actin dynamics independent of its role as an energy regulator.


Assuntos
Actinas/metabolismo , Drosophila melanogaster/genética , Genoma de Inseto , Proteínas Quinases/fisiologia , Vaccinia virus/fisiologia , Vacínia/genética , Internalização do Vírus , Quinases Proteína-Quinases Ativadas por AMP , Proteínas Quinases Ativadas por AMP , Animais , Northern Blotting , Movimento Celular , Células Cultivadas , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/metabolismo , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Imunofluorescência , Immunoblotting , Camundongos , Camundongos Knockout , Fosforilação , Pinocitose , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Pseudópodes , Interferência de RNA , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Vacínia/metabolismo , Vacínia/virologia , Replicação Viral , Cicatrização
4.
Bio Protoc ; 2(23)2012 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-27453902

RESUMO

Immunofluorescence is a method to detect viral infection in multiple types of host cells. This procedure can be adapted for both high-throughput and low-throughput assays for any virus for which there are antibodies available. Time of infection and virus multiplicity of infection (MOI) vary and should be optimized for each virus and host cell type. Here we give an example of viral immunofluorescence in a 96 well plate with a Rift Valley fever virus (RVFV, strain MP12) infection in mouse embryonic fibroblasts (MEF).

5.
J Vis Exp ; (42)2010 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-20834214

RESUMO

Viral pathogens represent a significant public health threat; not only can viruses cause natural epidemics of human disease, but their potential use in bioterrorism is also a concern. A better understanding of the cellular factors that impact infection would facilitate the development of much-needed therapeutics. Recent advances in RNA interference (RNAi) technology coupled with complete genome sequencing of several organisms has led to the optimization of genome-wide, cell-based loss-of-function screens. Drosophila cells are particularly amenable to genome-scale screens because of the ease and efficiency of RNAi in this system (1). Importantly, a wide variety of viruses can infect Drosophila cells, including a number of mammalian viruses of medical and agricultural importance (2,3,4). Previous RNAi screens in Drosophila have identified host factors that are required for various steps in virus infection including entry, translation and RNA replication (5). Moreover, many of the cellular factors required for viral replication in Drosophila cell culture are also limiting in human cells infected with these viruses (4,6,7,8, 9). Therefore, the identification of host factors co-opted during viral infection presents novel targets for antiviral therapeutics. Here we present a generalized protocol for a high-throughput RNAi screen to identify cellular factors involved in viral infection, using vaccinia virus as an example.


Assuntos
Drosophila/genética , Drosophila/virologia , Interferência de RNA , Vaccinia virus/patogenicidade , Vacínia/genética , Animais , Células Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA