Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Microbiol ; 37(2): 287-99, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10931325

RESUMO

ExoS is a type III cytotoxin of Pseudomonas aeruginosa, which modulates two eukaryotic signalling pathways. The N-terminus (residues 1-234) is a GTPase activating protein (GAP) for RhoGTPases, while the C-terminus (residues 232-453) encodes an ADP-ribosyltransferase. Utilizing a series of N-terminal deletion peptides of ExoS and an epitope-tagged full-length ExoS, two independent domains have been identified within the N-terminus of ExoS that are involved in intracellular localization and expression of GAP activity. N-terminal peptides of ExoS localized to the perinuclear region of CHO cells, and a membrane localization domain was localized between residues 36 and 78 of ExoS. The capacity to elicit CHO cell rounding and express GAP activity resided within residues 90-234 of ExoS, which showed that membrane localization was not required to elicit actin reorganization. ExoS was present in CHO cells as a full-length form, which fractionated with membranes, and as an N-terminally processed fragment, which localized to the cytosol. Thus, ExoS localizes in eukaryotic cells to the perinuclear region and is processed to a soluble fragment, which possesses both the GAP and ADP-ribosyltransferase activities.


Assuntos
ADP Ribose Transferases , Toxinas Bacterianas , Proteínas Ativadoras de GTPase/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Proteínas Quinases/metabolismo , Pseudomonas aeruginosa/metabolismo , Sequência de Aminoácidos , Animais , Western Blotting , Células CHO , Núcleo Celular/metabolismo , Núcleo Celular/microbiologia , Cricetinae , Imunofluorescência , Proteínas Ativadoras de GTPase/genética , Histidina Quinase , Dados de Sequência Molecular , Sinais de Localização Nuclear , Poli(ADP-Ribose) Polimerases/genética , Proteínas Quinases/genética , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , Deleção de Sequência , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Solubilidade
2.
Infect Immun ; 68(1): 403-6, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10603417

RESUMO

The presence of invasion-inhibitory activity that is regulated by the transcriptional activator ExsA of cytotoxic Pseudomonas aeruginosa has previously been proposed. The results of this study show that both ExoT and ExoS, known type III secreted effector proteins of P. aeruginosa that are regulated by ExsA, possess this activity. Invasion was reduced 94.4% by ExoT and 96.0% by ExoS. Invasion-inhibitory activity is not linked to ADP-ribosylation activity, at least for ExoS, since a noncatalytic mutant also inhibits uptake by an epithelial cell line (invasion was reduced 96. 0% by ExoSE381A).


Assuntos
Proteínas de Bactérias/fisiologia , Epitélio Corneano/microbiologia , Pseudomonas aeruginosa/patogenicidade , Animais , Proteínas de Bactérias/genética , Linhagem Celular , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Genes Bacterianos , Mutação , Pseudomonas aeruginosa/genética , Coelhos , Transativadores/genética , Transativadores/fisiologia , Virulência/genética , Virulência/fisiologia
3.
Mol Microbiol ; 32(2): 393-401, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10231494

RESUMO

Pseudomonas aeruginosa delivers exoenzyme S (ExoS) into the intracellular compartment of eukaryotic cells via a type III secretion pathway. Intracellular delivery of ExoS is cytotoxic for eukaryotic cells and has been shown to ADP-ribosylate Ras in vivo and uncouple a Ras-mediated signal transduction pathway. Functional mapping has localized the FAS-dependent ADP-ribosyltransferase domain to the carboxyl-terminus of ExoS. A transient transfection system was used to examine cellular responses to the amino-terminal 234 amino acids of ExoS (DeltaC234). Intracellular expression of DeltaC234 elicited the rounding of Chinese hamster ovary (CHO) cells and the disruption of actin filaments in a dose-dependent manner. Expression of DeltaC234 did not inhibit the expression of two independent reporter proteins, GFP and luciferase, or induce trypan blue uptake, which indicated that expression of DeltaC234 was not cytotoxic to CHO cells. Carboxyl-terminal deletion proteins of DeltaC234 were less efficient in the elicitation of CHO cell rounding than DeltaC234. Cytoskeleton rearrangement elicited by DeltaC234 was blocked and reversed by the addition of cytotoxic necrotizing factor 1 (CNF-1). CNF-1 catalyses the deamidation of Gln-63 of members of the Rho subfamily of small-molecular-weight GTP-binding proteins, resulting in protein activation. This implies a role for small-molecular-weight GTP-binding proteins in the disruption of actin by DeltaC234. Together, these data identify ExoS as a cytotoxin that possesses two functional domains. Intracellular expression of the amino-terminal domain of ExoS elicits the disruption of actin, while expression of the carboxyl-terminal domain of ExoS possesses FAS-dependent ADP-ribosyltransferase activity and is cytotoxic to eukaryotic cells.


Assuntos
ADP Ribose Transferases/química , Actinas/metabolismo , Citoesqueleto/metabolismo , Proteínas de Escherichia coli , Proteínas de Ligação ao GTP/metabolismo , Pseudomonas aeruginosa/metabolismo , ADP Ribose Transferases/genética , ADP Ribose Transferases/metabolismo , Animais , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/farmacologia , Western Blotting , Células CHO , Tamanho Celular , Cricetinae , Citoesqueleto/ultraestrutura , Citotoxinas/metabolismo , Citotoxinas/farmacologia , Pseudomonas aeruginosa/crescimento & desenvolvimento , Transfecção
4.
Infect Immun ; 67(4): 2040-4, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10085057

RESUMO

A strain of Pseudomonas aeruginosa that fails to express known type III-secreted effector proteins was constructed as an expression host. Individual effectors were expressed in trans, and their biological effects on CHO cells were assessed in an acute cellular infection model. Intoxication with ExoS, ExoT, or ExoY resulted in alterations in cell morphology. As shown in previous genetic studies, ExoU expression was linked to acute cytotoxicity.


Assuntos
Toxinas Bacterianas/metabolismo , Exotoxinas/metabolismo , Pseudomonas aeruginosa/metabolismo , Animais , Transporte Biológico , Células CHO , Cricetinae
5.
Infect Immun ; 67(2): 914-20, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9916108

RESUMO

This study was initiated to characterize the regulation and secretion of ExoS by Pseudomonas aeruginosa during contact with eukaryotic cells. The production of ExoS was monitored by a sensitive ADP-ribosyltransferase activity assay, and specific activities were calculated for supernatant and cell-associated fractions. Time course analysis indicated that ExoS was produced after a lag period, suggesting that induction of the regulon is necessary for the expression of detectable amounts of enzyme activity. Under tissue culture growth conditions, ExoS was induced when P. aeruginosa was in contact with Chinese hamster ovary (CHO) cells or after growth in tissue culture medium with serum. The serum induction of ExoS appeared to result in generalized type III secretion, while induction by contact with CHO cells appeared to result in polarized type III secretion. Mutants in the type III secretory system that express a null phenotype for ExoS production in bacteriological medium produced but did not secrete the enzyme when P. aeruginosa was grown under inducing conditions in tissue culture medium. These results suggest that both induction and secretion of ExoS may differ when the bacteria are exposed to different growth environments. The putative type III translocation proteins and secretion apparatus of P. aeruginosa were required for translocation of bacterial factors that mediate changes in CHO cell morphology during infection.


Assuntos
Proteínas Quinases/biossíntese , Pseudomonas aeruginosa/metabolismo , Adaptação Biológica , Animais , Transporte Biológico , Células CHO , Proteínas de Transporte/metabolismo , Cricetinae , Técnicas de Cultura , Histidina Quinase , Mutação , Proteínas Quinases/metabolismo , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/crescimento & desenvolvimento
6.
Proc Natl Acad Sci U S A ; 95(23): 13899-904, 1998 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-9811898

RESUMO

The exoenzyme S regulon is a set of coordinately regulated virulence genes of Pseudomonas aeruginosa. Proteins encoded by the regulon include a type III secretion and translocation apparatus, regulators of gene expression, and effector proteins. The effector proteins include two enzymes with ADP-ribosyltransferase activity (ExoS and ExoT) and an acute cytotoxin (ExoU). In this study, we identified ExoY as a fourth effector protein of the regulon. ExoY is homologous to the extracellular adenylate cyclases of Bordetella pertussis (CyaA) and Bacillus anthracis (EF). The homology among the three adenylate cyclases is limited to two short regions, one of which possesses an ATP-binding motif. In assays for adenylate cyclase activity, recombinant ExoY (rExoY) catalyzed the formation of cAMP with a specific activity similar to the basal activity of CyaA. In contrast to CyaA and EF, rExoY activity was not stimulated or activated by calmodulin. A 500-fold stimulation of activity was detected following the addition of a cytosolic extract from Chinese hamster ovary (CHO) cells. These results indicate that a eukaryotic factor, distinct from calmodulin, enhances rExoY catalysis. Site-directed mutagenesis of residues within the putative active site of ExoY abolished adenylate cyclase activity. Infection of CHO cells with ExoY-producing strains of P. aeruginosa resulted in the intracellular accumulation of cAMP. cAMP accumulation within CHO cells depended on an intact type III translocation apparatus, demonstrating that ExoY is directly translocated into the eukaryotic cytosol.


Assuntos
Adenilil Ciclases/genética , Glucosiltransferases/genética , Pseudomonas aeruginosa/enzimologia , Adenilil Ciclases/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Cricetinae , Genes Bacterianos , Glucosiltransferases/metabolismo , Dados de Sequência Molecular
7.
J Cell Sci ; 109 ( Pt 6): 1437-47, 1996 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8799831

RESUMO

Histone H1 is highly phosphorylated in mitotic HeLa cells, but is quickly dephosphorylated in vivo at the end of mitosis and in vitro following cell lysis. We show here that okadaic acid and microcystin-LR block the in vitro dephosphorylation of H1 and that they do so directly by inhibiting the histone H1 phosphatase rather than by some indirect mechanism. The concentrations of microcystin and okadaic acid required for inhibition strongly suggest that the histone H1 phosphatase is either PP1 or an unknown protein phosphatase with okadaic acid-sensitivity similar to PP1. The histone H1 phosphatase is predominantly located in chromosomes with at most one copy for every 86 nucleosomes. This tends to support its identification as PP1, since localization in mitotic chromosomes is a characteristic of PP1 but not of the other known okadaic acid-sensitive protein phosphatases. We also show that treatment of metaphase-arrested HeLa cells with staurosporine and olomoucine, inhibitors of p34cdc2 and other protein kinases, rapidly induces reassembly of interphase nuclei and dephosphorylation of histone H1 without chromosome segregation. This result indicates that protein kinase activity must remain elevated to maintain a mitotic block. Using this as a model system for the M- to G1-phase transition, we present evidence from inhibitor studies suggesting that the in vivo histone H1 phosphatase may be either PP1 or another phosphatase with similar okadaic acid-sensitivity, but not PP2A.


Assuntos
Histonas/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Núcleo Celular/efeitos dos fármacos , Cromatina/enzimologia , Inibidores Enzimáticos/farmacologia , Células HeLa , Humanos , Cinetina , Toxinas Marinhas , Metáfase , Microcistinas , Mitose , Ácido Okadáico/farmacologia , Oxazóis/farmacologia , Peptídeos Cíclicos/farmacologia , Fosfoproteínas Fosfatases/antagonistas & inibidores , Fosforilação , Inibidores de Proteínas Quinases , Proteína Fosfatase 1 , Proteína Fosfatase 2 , Purinas/farmacologia , Estaurosporina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...