Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Virulence ; 8(6): 719-740, 2017 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-27575017

RESUMO

The eukaryotic transcriptional regulator Nuclear Factor kappa B (NF-κB) plays a central role in the defense to pathogens. Despite this, few studies have analyzed NF-κB activity in single cells during infection. Here, we investigated at the single cell level how NF-κB nuclear localization - a proxy for NF-κB activity - oscillates in infected and uninfected fibroblasts co-existing in cultures exposed to Salmonella enterica serovar Typhimurium. Fibroblasts were used due to the capacity of S. Typhimurium to persist in this cell type. Real-time dynamics of NF-κB was examined in microfluidics, which prevents cytokine accumulation. In this condition, infected (ST+) cells translocate NF-κB to the nucleus at higher rate than the uninfected (ST-) cells. Surprisingly, in non-flow (static) culture conditions, ST- fibroblasts exhibited higher NF-κB nuclear translocation than the ST+ population, with these latter cells turning refractory to external stimuli such as TNF-α or a second infection. Sorting of ST+ and ST- cell populations confirmed enhanced expression of NF-κB target genes such as IL1B, NFKBIA, TNFAIP3, and TRAF1 in uninfected (ST-) fibroblasts. These observations proved that S. Typhimurium dampens the NF-κB response in the infected fibroblast. Higher expression of SOCS3, encoding a "suppressor of cytokine signaling," was also observed in the ST+ population. Intracellular S. Typhimurium subverts NF-κB activity using protein effectors translocated by the secretion systems encoded by pathogenicity islands 1 (T1) and 2 (T2). T1 is required for regulating expression of SOCS3 and all NF-κB target genes analyzed whereas T2 displayed no role in the control of SOCS3 and IL1B expression. Collectively, these data demonstrate that S. Typhimurium attenuates NF-κB signaling in fibroblasts, an effect only perceptible when ST+ and ST- populations are analyzed separately. This tune-down in a central host defense might be instrumental for S. Typhimurium to establish intracellular persistent infections.


Assuntos
Fibroblastos/microbiologia , NF-kappa B/metabolismo , Salmonella typhimurium/patogenicidade , Linhagem Celular , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Interleucina-1beta/genética , Microfluídica , Inibidor de NF-kappaB alfa/genética , NF-kappa B/genética , Salmonella typhimurium/efeitos dos fármacos , Salmonella typhimurium/metabolismo , Transdução de Sinais , Análise de Célula Única , Proteína 3 Supressora da Sinalização de Citocinas/genética , Fator 1 Associado a Receptor de TNF/genética , Imagem com Lapso de Tempo , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/farmacologia
2.
Infect Immun ; 81(1): 154-65, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23090959

RESUMO

Genome-wide expression analyses have provided clues on how Salmonella proliferates inside cultured macrophages and epithelial cells. However, in vivo studies show that Salmonella does not replicate massively within host cells, leaving the underlying mechanisms of such growth control largely undefined. In vitro infection models based on fibroblasts or dendritic cells reveal limited proliferation of the pathogen, but it is presently unknown whether these phenomena reflect events occurring in vivo. Fibroblasts are distinctive, since they represent a nonphagocytic cell type in which S. enterica serovar Typhimurium actively attenuates intracellular growth. Here, we show in the mouse model that S. Typhimurium restrains intracellular growth within nonphagocytic cells positioned in the intestinal lamina propria. This response requires a functional PhoP-PhoQ system and is reproduced in primary fibroblasts isolated from the mouse intestine. The fibroblast infection model was exploited to generate transcriptome data, which revealed that ∼2% (98 genes) of the S. Typhimurium genome is differentially expressed in nongrowing intracellular bacteria. Changes include metabolic reprogramming to microaerophilic conditions, induction of virulence plasmid genes, upregulation of the pathogenicity islands SPI-1 and SPI-2, and shutdown of flagella production and chemotaxis. Comparison of relative protein levels of several PhoP-PhoQ-regulated functions (PagN, PagP, and VirK) in nongrowing intracellular bacteria and extracellular bacteria exposed to diverse PhoP-PhoQ-inducing signals denoted a regulation responding to acidic pH. These data demonstrate that S. Typhimurium restrains intracellular growth in vivo and support a model in which dormant intracellular bacteria could sense vacuolar acidification to stimulate the PhoP-PhoQ system for preventing intracellular overgrowth.


Assuntos
Proteínas de Bactérias/biossíntese , Regulação Bacteriana da Expressão Gênica , Genoma Bacteriano , Salmonella typhimurium/citologia , Salmonella typhimurium/genética , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Feminino , Fibroblastos/metabolismo , Fibroblastos/microbiologia , Estudo de Associação Genômica Ampla/métodos , Ilhas Genômicas/genética , Concentração de Íons de Hidrogênio , Peptídeos e Proteínas de Sinalização Intercelular , Mucosa Intestinal/metabolismo , Intestinos/microbiologia , Rim/metabolismo , Rim/microbiologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mucosa/metabolismo , Mucosa/microbiologia , Peptídeos/genética , Peptídeos/metabolismo , Ratos , Infecções por Salmonella/genética , Infecções por Salmonella/metabolismo , Infecções por Salmonella/microbiologia , Salmonella typhimurium/metabolismo , Salmonella typhimurium/patogenicidade , Transcriptoma/genética , Virulência/genética
3.
Antioxid Redox Signal ; 15(2): 325-37, 2011 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21338289

RESUMO

Translocated in liposarcoma (TLS) is a poorly characterized multifunctional protein involved in the genotoxic response. TLS regulates gene expression at several steps, including splicing and mRNA transport, possibly connecting transcriptional and posttranscriptional events. AIMS: In this study we aimed to idenfity molecular targets and regulatory partners of TLS. RESULTS AND INNOVATION: Here we report that TLS transcriptionally regulates the expression of oxidative stress protection genes. This regulation requires interaction with the transcriptional coactivator peroxisome proliferator activated receptor γ-coactivator 1α (PGC-1α), a master regulator of mitochondrial function that coordinately induces the expression of genes involved in detoxification of mitochondrial reactive oxygen species (ROS). Microarray gene expression analysis showed that TLS transcriptional activity is impaired in the absence of PGC-1α, and is thus largely dependent on PGC-1α. CONCLUSION: These results suggest the existence of a regulatory circuit linking the control of ROS detoxification to the coordinated cross-talk between oxidative metabolism and the cellular response to genomic DNA damage.


Assuntos
Regulação da Expressão Gênica/fisiologia , Estresse Oxidativo/genética , Proteína FUS de Ligação a RNA/genética , Transativadores/fisiologia , Animais , Sequência de Bases , Células Cultivadas , Primers do DNA , Metabolismo Energético , Camundongos , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Fatores de Transcrição , Técnicas do Sistema de Duplo-Híbrido
4.
J Biol Chem ; 284(21): 14476-84, 2009 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-19324885

RESUMO

Oxidative stress is a hallmark of metabolism-related diseases and a risk factor for atherosclerosis. FoxO factors have been shown to play a key role in vascular endothelial development and homeostasis. Foxo3a can protect quiescent cells from oxidative stress through the regulation of detoxification genes such as sod2 and catalase. Here we show that Foxo3a is a direct transcriptional regulator of a group of oxidative stress protection genes in vascular endothelial cells. Importantly, Foxo3a activity requires the transcriptional co-activator PGC-1alpha, because it is severely curtailed in PGC-1alpha-deficient endothelial cells. Foxo3a and PGC-1alpha appear to interact directly, as shown by co-immunoprecipitation and in vitro interaction assays, and are recruited to the same promoter regions. The notion that Foxo3a and PGC-1alpha interact directly to regulate oxidative stress protection genes in the vascular endothelium is supported by the observation that PGC-1alpha transcriptional activity at the sod2 (manganese superoxide dismutase) promoter requires a functional FoxO site. We also demonstrate that Foxo3a is a direct transcriptional regulator of PGC-1alpha, suggesting that an auto-regulatory cycle regulates Foxo3a/PGC-1alpha control of the oxidative stress response.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica , Estresse Oxidativo/genética , Transativadores/metabolismo , Animais , Bovinos , Células Cultivadas , Células Endoteliais/metabolismo , Indução Enzimática , Proteína Forkhead Box O3 , Humanos , Camundongos , Modelos Biológicos , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Ligação Proteica , Superóxido Dismutase/biossíntese , Fatores de Transcrição , Veias Umbilicais/citologia
5.
Vet Microbiol ; 130(3-4): 298-311, 2008 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-18313237

RESUMO

Live attenuated Salmonella enterica strains have been extensively studied as potential vectors for the oral delivery of heterologous antigens. Due to its ability to target immune cells, its specific mechanism for crossing the intestinal barrier, and its swine-restricted tropism, S. enterica subspecies enterica serovar Choleraesuis (S. Choleraesuis) has attracted a great deal of interest for the production of bacterial-based oral carriers specifically adapted to swine. In this study, two mutants of S. Choleraesuis were constructed and their attenuation and intracellular fate analysed with the purpose of engineering new attenuated live strains with improved properties as oral vaccine carriers. Those strains harboured a specific deletion either within the phoP or rpoS genes, which encode virulence-related regulators in S. Typhimurium. In comparison to the wild-type parental S. Choleraesuis, the mutant strains, especially DeltaphoP, were extremely low in virulence in the murine model and in the natural host, the pig. Moreover, when compared with a commercial live vaccine strain, SC-54, the two mutants showed a higher level of attenuation in mice and DeltaphoP also in pigs. In addition, DeltarpoS and DeltaphoP presented a proliferation and survival phenotype within swine intestinal primary fibroblast and macrophage cell cultures, respectively. Collectively, the present results indicate that the DeltarpoS and DeltaphoP strains of S. Choleraesuis gather adequate features to be potential candidates for vaccine vectors for the specific delivery of heterologous antigens adapted to pigs.


Assuntos
Proteínas de Bactérias/genética , Fibroblastos/microbiologia , Intestinos/citologia , Macrófagos/microbiologia , Salmonella enterica/genética , Fator sigma/genética , Animais , Células Cultivadas , Deleção de Genes , Regulação Bacteriana da Expressão Gênica , Camundongos , Camundongos Endogâmicos BALB C , Salmonelose Animal/microbiologia , Salmonella enterica/classificação , Salmonella enterica/patogenicidade , Suínos , Doenças dos Suínos/microbiologia , Virulência
6.
Cell Microbiol ; 7(7): 901-9, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15953023

RESUMO

The literature refers to Salmonella enterica as an intracellular bacterial pathogen that proliferates within vacuoles of mammalian cells. However, recent in vivo studies have revealed that the vast majority of infected cells contain very few intracellular bacteria (three to four organisms). Salmonella intracellular growth is also limited in cultured dendritic cells and fibroblasts, two cell types abundant in tissues located underneath the intestinal epithelium. Recently, a Salmonella factor previously known for its role as a negative regulator of intracellular growth has been shown to tightly repress certain pathogen functions upon host colonization and to be critical for virulence. The connection between virulence and the negative control of intracellular growth is further sustained by the fact that some attenuated mutants overgrow in non-phagocytic cells located in the intestinal lamina propria. These findings are changing our classical view of Salmonella as a fast growing intracellular pathogen and suggest that this pathogen may trigger responses directed to reduce the growth rate within the infected cell. These responses could play a critical role in modulating the delicate balance between disease and persistence.


Assuntos
Regulação Bacteriana da Expressão Gênica , Salmonella enterica/crescimento & desenvolvimento , Salmonella enterica/patogenicidade , Animais , Células Cultivadas , Células Dendríticas/microbiologia , Fibroblastos/microbiologia , Humanos , Macrófagos/microbiologia , Salmonella enterica/genética , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA