Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Mol Syst Biol ; 13(1): 906, 2017 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-28082348

RESUMO

Protein-protein interaction networks (interactomes) define the functionality of all biological systems. In apoptosis, proteolysis by caspases is thought to initiate disassembly of protein complexes and cell death. Here we used a quantitative proteomics approach, protein correlation profiling (PCP), to explore changes in cytoplasmic and mitochondrial interactomes in response to apoptosis initiation as a function of caspase activity. We measured the response to initiation of Fas-mediated apoptosis in 17,991 interactions among 2,779 proteins, comprising the largest dynamic interactome to date. The majority of interactions were unaffected early in apoptosis, but multiple complexes containing known caspase targets were disassembled. Nonetheless, proteome-wide analysis of proteolytic processing by terminal amine isotopic labeling of substrates (TAILS) revealed little correlation between proteolytic and interactome changes. Our findings show that, in apoptosis, significant interactome alterations occur before and independently of caspase activity. Thus, apoptosis initiation includes a tight program of interactome rearrangement, leading to disassembly of relatively few, select complexes. These early interactome alterations occur independently of cleavage of these protein by caspases.


Assuntos
Caspases/metabolismo , Citoplasma/metabolismo , Mitocôndrias/metabolismo , Proteômica/métodos , Receptor fas/metabolismo , Apoptose , Cromatografia Líquida , Humanos , Marcação por Isótopo , Células Jurkat , Espectrometria de Massas , Mapas de Interação de Proteínas , Proteólise
2.
PLoS Pathog ; 9(7): e1003518, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23935490

RESUMO

To further its pathogenesis, S. Typhimurium delivers effector proteins into host cells, including the novel E3 ubiquitin ligase (NEL) effector SspH2. Using model systems in a cross-kingdom approach we gained further insight into the molecular function of this effector. Here, we show that SspH2 modulates innate immunity in both mammalian and plant cells. In mammalian cell culture, SspH2 significantly enhanced Nod1-mediated IL-8 secretion when transiently expressed or bacterially delivered. In addition, SspH2 also enhanced an Rx-dependent hypersensitive response in planta. In both of these nucleotide-binding leucine rich repeat receptor (NLR) model systems, SspH2-mediated phenotypes required its catalytic E3 ubiquitin ligase activity and interaction with the conserved host protein SGT1. SGT1 has an essential cell cycle function and an additional function as an NLR co-chaperone in animal and plant cells. Interaction between SspH2 and SGT1 was restricted to SGT1 proteins that have NLR co-chaperone function and accordingly, SspH2 did not affect SGT1 cell cycle functions. Mechanistic studies revealed that SspH2 interacted with, and ubiquitinated Nod1 and could induce Nod1 activity in an agonist-independent manner if catalytically active. Interestingly, SspH2 in vitro ubiquitination activity and protein stability were enhanced by SGT1. Overall, this work adds to our understanding of the sophisticated mechanisms used by bacterial effectors to co-opt host pathways by demonstrating that SspH2 can subvert immune responses by selectively exploiting the functions of a conserved host co-chaperone.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas de Ciclo Celular/metabolismo , Imunidade Inata , Proteínas de Membrana/metabolismo , Chaperonas Moleculares/metabolismo , Proteínas Adaptadoras de Sinalização NOD/metabolismo , Salmonella typhimurium/imunologia , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Ciclo Celular/química , Linhagem Celular , Deleção de Genes , Interações Hospedeiro-Patógeno , Humanos , Interleucina-8/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/genética , Proteínas Mutantes/metabolismo , Imunidade Vegetal , Folhas de Planta/genética , Folhas de Planta/imunologia , Folhas de Planta/metabolismo , Folhas de Planta/microbiologia , Proteínas de Plantas/metabolismo , Plantas Geneticamente Modificadas , Estabilidade Proteica , Proteínas Recombinantes/metabolismo , Salmonella typhimurium/metabolismo , Nicotiana/genética , Nicotiana/imunologia , Nicotiana/metabolismo , Nicotiana/microbiologia , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Regulação para Cima
3.
Mol Cell Proteomics ; 12(6): 1632-43, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23459991

RESUMO

During the late stages of infection, Salmonella secretes numerous effectors through a type III secretion system that is encoded within Salmonella pathogenicity island 2 (SPI2). Despite the importance of SPI2 as a major virulence factor leading to the systemic spread of the bacteria and diseases, a global view of its effects on host responses is still lacking. Here, we measured global impacts of SPI2 effectors on the host phosphorylation and protein expression levels in RAW264.7 and in HeLa cells, as macrophage and nonphagocytic models of infection. We observe that SPI2 effectors differentially modulate the host phosphoproteome and cellular processes (e.g. protein trafficking, cytoskeletal regulation, and immune signaling) in a host cell-dependent manner. Our unbiased approach reveals the involvement of many previously unrecognized proteins, including E3 ligases (HERC4, RanBP2, and RAD18), kinases (CDK, SIK3, and WNK1), and histones (H2B1F, H4, and H15), in late stages of Salmonella infection. Furthermore, from this phosphoproteome analysis and other quantitative screens, we identified HSP27 as a direct in vitro and in vivo molecular target of the only type III secreted kinase, SteC. Using biochemical and cell biological assays, we demonstrate that SteC phosphorylates multiple sites in HSP27 and induces actin rearrangement through this protein. Together, these results provide a broader landscape of host players contributing to specific processes/pathways mediated by SPI2 effectors than was previously appreciated.


Assuntos
Proteínas de Bactérias/genética , Ilhas Genômicas , Proteínas de Choque Térmico HSP27/genética , Macrófagos/metabolismo , Fosfoproteínas/genética , Proteínas Quinases/genética , Proteoma/genética , Salmonella typhimurium/genética , Salmonella typhimurium/patogenicidade , Actinas/genética , Actinas/metabolismo , Animais , Proteínas de Bactérias/metabolismo , Linhagem Celular , Regulação da Expressão Gênica , Proteínas de Choque Térmico HSP27/metabolismo , Células HeLa , Proteínas de Choque Térmico , Interações Hospedeiro-Patógeno , Humanos , Macrófagos/citologia , Macrófagos/microbiologia , Camundongos , Chaperonas Moleculares , Fosfoproteínas/metabolismo , Mapeamento de Interação de Proteínas , Proteínas Quinases/metabolismo , Proteoma/isolamento & purificação , Proteoma/metabolismo , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
4.
EMBO J ; 29(8): 1331-47, 2010 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-20300065

RESUMO

3-phosphorylated phosphoinositides (3-PtdIns) orchestrate endocytic trafficking pathways exploited by intracellular pathogens such as Salmonella to gain entry into the cell. To infect the host, Salmonellae subvert its normal macropinocytic activity, manipulating the process to generate an intracellular replicative niche. Disruption of the PtdIns(5) kinase, PIKfyve, be it by interfering mutant, siRNA-mediated knockdown or pharmacological means, inhibits the intracellular replication of Salmonella enterica serovar typhimurium in epithelial cells. Monitoring the dynamics of macropinocytosis by time-lapse 3D (4D) videomicroscopy revealed a new and essential role for PI(3,5)P(2) in macropinosome-late endosome/lysosome fusion, which is distinct from that of the small GTPase Rab7. This PI(3,5)P(2)-dependent step is required for the proper maturation of the Salmonella-containing vacuole (SCV) through the formation of Salmonella-induced filaments (SIFs) and for the engagement of the Salmonella pathogenicity island 2-encoded type 3 secretion system (SPI2-T3SS). Finally, although inhibition of PIKfyve in macrophages did inhibit Salmonella replication, it also appears to disrupt the macrophage's bactericidal response.


Assuntos
Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Salmonella typhimurium/patogenicidade , Aminopiridinas/farmacologia , Animais , Proteínas de Bactérias/metabolismo , Linhagem Celular , Endocitose , Endossomos/metabolismo , Compostos Heterocíclicos com 3 Anéis/farmacologia , Humanos , Lisossomos/metabolismo , Macrófagos/microbiologia , Proteínas de Membrana/metabolismo , Mutação , Fosfatidilinositol 3-Quinases/genética , Pinocitose , Interferência de RNA , Salmonella typhimurium/crescimento & desenvolvimento , Vacúolos/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , proteínas de unión al GTP Rab7
5.
Curr Biol ; 17(9): 783-8, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17442572

RESUMO

Virulence, defined as damage to the host, is a trait of pathogens that evolutionary theory suggests benefits the pathogen in the "struggle for existence". Pathogens employ virulence mechanisms that contribute to disease. Central to the evolution of virulence of the infectious agents causing an array of bacterial disease is the evolutionary acquisition of type III secretion, a macromolecular complex that creates a syringe-like apparatus extending from the bacterial cytosol to the eukaryotic cytosol and delivers secreted bacterial virulence factors (effectors) into host cells. In this work, we quantify the contribution of virulence determinants to the evolutionary success of a pathogen. Using a natural pathogen of mice, we show that virulence factors provide a selective advantage by enhancing transmission between hosts. Virulence factors that have a major contribution to disease were absolutely required for transmission of the pathogen to naive hosts. Virulence-factor mutants with more subtle defects in pathogenesis had quantifiable roles in the time required to transmit the pathogen between mice. Virulence-factor mutants were also found to lose in competition with wild-type bacteria when iteratively transmitted from infected to uninfected mice. These results directly demonstrate that virulence is selected via the fitness advantage it provides to the host-to-host cycle of pathogenic species.


Assuntos
Evolução Biológica , Citrobacter rodentium/patogenicidade , Transmissão de Doença Infecciosa , Infecções por Enterobacteriaceae/transmissão , Fatores de Virulência/metabolismo , Animais , Citrobacter rodentium/genética , Camundongos , Seleção Genética , Fatores de Tempo , Virulência , Fatores de Virulência/genética
6.
Cell Microbiol ; 10(8): 1646-61, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18397382

RESUMO

Host resistance against Salmonella enterica serovar Typhimurium (S. Typhimurium) is mediated by natural resistance-associated macrophage protein 1 (Nramp1/Slc11a1). Nramp1 is critical to host defence, as mice lacking Nramp1 fail to control bacterial replication and succumb to low doses of S. Typhimurium. Despite this crucial role, the mechanisms underlying Nramp1's protective effects are unclear. Dendritic cells (DCs) that sample the intestinal lumen are among the first cells encountered by S. Typhimurium following oral infection and act as a conduit for S. Typhimurium to cross the intestinal epithelial barrier. We report that DCs, including intestinal, splenic and bone marrow-derived DCs (BMDCs), express Nramp1 protein. In the small intestine, Nramp1 expression is greater in a subset of DCs (CD11c(+)CD103(-)) characterized by the elevated expression of pro-inflammatory cytokines in response to bacterial products. While Nramp1 expression did not affect S. Typhimurium replication in BMDCs, infected Nramp1+/+ BMDCs and intestinal CD11c(+)CD103(-) DCs secreted more inflammatory cytokines (IL-6, IL-12 and TNF-alpha) than Nramp1-/-, suggesting that Nramp1 expression may promote a more rapid inflammatory response following infection. Collectively, these findings reveal a new role for DCs and Nramp1 in modulating the host inflammatory response to S. Typhimurium.


Assuntos
Proteínas de Transporte de Cátions/imunologia , Células Dendríticas/imunologia , Salmonelose Animal/imunologia , Salmonella typhimurium/imunologia , Animais , Citocinas/imunologia , Intestino Delgado/microbiologia , Camundongos , Mucosa/imunologia
7.
PLoS Pathog ; 2(5): e42, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16733541

RESUMO

The evolution of pathogens presents a paradox. Pathogenic species are often absolutely dependent on their host species for their propagation through evolutionary time, yet the pathogenic lifestyle requires that the host be damaged during this dependence. It is clear that pathogenic strategies are successful in evolutionary terms because a diverse array of pathogens exists in nature. Pathogens also evolve using a broad range of molecular mechanisms to acquire and modulate existing virulence traits in order to achieve this success. Detailing the benefit of enhanced selection derived through virulence and understanding the mechanisms through which virulence evolves are important to understanding the natural world and both have implications for human health.


Assuntos
Evolução Biológica , Seleção Genética , Fatores de Virulência/genética , Animais , Humanos
8.
BMC Infect Dis ; 7: 65, 2007 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-17597539

RESUMO

BACKGROUND: Salmonella meningitis is a rare and serious infection of the central nervous system following acute Salmonella enterica sepsis. For this pathogen, no appropriate model has been reported in which to examine infection kinetics and natural dissemination to the brain. METHODS: Five mouse lines including C57BL/6, Balb/c, 129S6-Slc11a1tm1Mcg, 129S1/SvImJ, B6.129-Inpp5dtm1Rkh were used in the murine typhoid model to examine the dissemination of systemic Salmonella enterica serovar Typhimurium following oral infection. RESULTS: We report data on spontaneous meningitis and brain infection following oral infection of mice with Salmonella enterica serovar Typhimurium. CONCLUSION: This model may provide a system in which dissemination of bacteria through the central nervous system and the influence of host and bacterial genetics can be queried.


Assuntos
Modelos Animais de Doenças , Meningites Bacterianas/microbiologia , Salmonelose Animal/fisiopatologia , Salmonella typhimurium/patogenicidade , Administração Oral , Animais , Encefalite/microbiologia , Encefalite/patologia , Feminino , Meningites Bacterianas/patologia , Camundongos , Camundongos Endogâmicos
9.
J Mol Biol ; 348(4): 817-30, 2005 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-15843015

RESUMO

Salmonella enterica serovar Typhimurium is lysogenized by several temperate bacteriophages that encode lysogenic conversion genes, which can act as virulence factors during infection and contribute to the genetic diversity and pathogenic potential of the lysogen. We have investigated the temperate bacteriophage called Gifsy-1 in S.enterica serovar Typhimurium and show here that the product of the gogB gene encoded within this phage shares similarity with proteins from other Gram-negative pathogens. The amino-terminal portion of GogB shares similarity with leucine-rich repeat-containing virulence-associated proteins from other Gram-negative pathogens, whereas the carboxyl-terminal portion of GogB shares similarity with uncharacterized proteins in other pathogens. We show that GogB is secreted by both type III secretion systems encoded in Salmonella Pathogenicity Island-1 (SPI-1) and SPI-2 but translocation into host cells is a SPI-2-mediated process. Once translocated, GogB localizes to the cytoplasm of infected host cells. The genetic regulation of gogB in Salmonella is influenced by the transcriptional activator, SsrB, under SPI-2-inducing conditions, but the modular nature of the gogB gene allows for autonomous expression and type III secretion following horizontal gene transfer into a heterologous pathogen. These data define the first autonomously expressed lysogenic conversion gene within Gifsy-1 that acts as a modular and promiscuous type III-secreted substrate of the infection process.


Assuntos
Regulação Viral da Expressão Gênica , Fagos de Salmonella/genética , Fagos de Salmonella/metabolismo , Salmonella typhimurium/metabolismo , Salmonella typhimurium/virologia , Proteínas Virais/genética , Proteínas Virais/metabolismo , Sequência de Aminoácidos , Células HeLa , Humanos , Dados de Sequência Molecular , Mutação/genética , Prófagos/genética , Prófagos/metabolismo , Prófagos/patogenicidade , Transporte Proteico , Fagos de Salmonella/patogenicidade , Salmonella typhimurium/química , Salmonella typhimurium/genética , Alinhamento de Sequência , Especificidade por Substrato , Proteínas Virais/química , Fatores de Virulência/química , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
10.
Microbes Infect ; 5(7): 561-70, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12787732

RESUMO

The Salmonella pathogenicity island-2 (SPI2) is a virulence locus on the bacterial chromosome required for intracellular proliferation and systemic infection in mice. Cell culture models and a murine model of systemic infection were used to address the role of an uncharacterized SPI2 open reading frame, designated as sseA, in Salmonella virulence. A Salmonella strain with an unmarked internal deletion of sseA displayed a phenotype that was similar to an SPI2-encoded type III secretion system apparatus mutant. Moreover, SseA was required for survival and replication within epithelial cells and macrophages. Murine infection studies confirmed that the DeltasseA strain was severely attenuated for virulence. Using immunofluorescence microscopy, the virulence defect in the DeltasseA strain was attributed to an inability to translocate SPI2 effector proteins into host cells. These data demonstrate that SseA is essential for SPI2-mediated translocation of effector proteins.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas de Membrana/metabolismo , Chaperonas Moleculares/fisiologia , Salmonella/patogenicidade , Animais , Proteínas de Bactérias/genética , Feminino , Genes Bacterianos , Células HeLa , Humanos , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Chaperonas Moleculares/genética , Mutação , Transporte Proteico , Salmonella/metabolismo , Salmonelose Animal/microbiologia , Fatores de Virulência/fisiologia
11.
J Med Microbiol ; 49(11): 993-1001, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11073153

RESUMO

Melioidosis is an infectious disease caused by Burkholderia pseudomallei. Genomic subtractive hybridisation was performed with the closely related avirulent species B. thailandensis to identify virulence genes of B. pseudomallei. The subtractive hybridisation products were highly specific for B. pseudomallei. Sequence analysis revealed a number of putative virulence factors, as well as apparently novel sequences of unknown function. The subtracted library contained DNA regions of relatively low G + C mol% content, which were distributed throughout the B. pseudomallei genome. The distribution of subtracted sequences amongst a collection of 22 B. pseudomallei isolates was found to be variable, with the exception of three strains which almost universally lacked the subtracted sequences. These three strains also differed in that they were highly haemolytic, indicating a possible separate virotype.


Assuntos
Burkholderia pseudomallei/genética , Burkholderia/genética , DNA Bacteriano/química , Burkholderia/classificação , Burkholderia/patogenicidade , Burkholderia pseudomallei/classificação , Burkholderia pseudomallei/patogenicidade , Clonagem Molecular , DNA Ribossômico/química , Eletroforese em Gel de Campo Pulsado , Genoma Bacteriano , Humanos , Hibridização de Ácido Nucleico , Fenótipo , Filogenia , Reação em Cadeia da Polimerase , RNA Ribossômico 16S/genética , Análise de Sequência de DNA , Virulência/genética
12.
Artigo em Inglês | MEDLINE | ID: mdl-15115093

RESUMO

The lambdaZAP II expressed genomic library of B. pseudomallei was screened with pooled melioidosis serum preabsorbed with E. coli host cell. The positive clones were detected by using protein A-CDP-star chemiluminescence. All of 14 positive clones reacted with only the pooled absorbed melioidosis serum and not the pooled absorbed normal serum when tested with the plaque dot blot analysis. The expressed genes were detected by using a combination of immunoscreening, bioinformatics and molecular biology. At least six in vivo expressed genes were identified by this approach. Two were well known virulent genes, gmhA (a capsule biosynthetic gene) and bipD (type III secretion protein gene). Another two were genes coded for conserved hypothetical protein. The last two isolated genes were groEL (a chaperonine protein gene), and a gene encoding transmembrane protein.


Assuntos
Proteínas de Bactérias/genética , Burkholderia pseudomallei/genética , Burkholderia pseudomallei/patogenicidade , Melioidose/microbiologia , Bacteriófago lambda , Clonagem Molecular , Perfilação da Expressão Gênica , Biblioteca Genômica , Humanos , Imunoensaio , Medições Luminescentes , Virulência/genética
13.
mBio ; 5(5): e01727-14, 2014 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-25249283

RESUMO

UNLABELLED: The transcription factors HilA and SsrB activate expression of two type III secretion systems (T3SSs) and cognate effectors that reprogram host cell functions to benefit infecting Salmonella in the host. These transcription factors, the secretion systems, and the effectors are all encoded by horizontally acquired genes. Using quantitative proteomics, we quantified the abundance of 2,149 proteins from hilA or ssrB Salmonella in vitro. Our results suggest that the HilA regulon does not extend significantly beyond proteins known to be involved in direct interactions with intestinal epithelium. On the other hand, SsrB influences the expression of a diverse range of proteins, many of which are ancestral to the acquisition of ssrB. In addition to the known regulon of T3SS-related proteins, we show that, through SodCI and bacterioferritin, SsrB controls resistance to reactive oxygen species and that SsrB down-regulates flagella and motility. This indicates that SsrB-controlled proteins not only redirect host cell membrane traffic to establish a supportive niche within host cells but also have adapted to the chemistry and physical constraints of that niche. IMPORTANCE: Expression of T3SSs typically requires a transcription factor that is linked in a genomic island. Studies of the targets of HilA and SsrB have focused on almost exclusively on T3SS substrates that are either linked or encoded in distinct genomic islands. By broadening our focus, we found that the regulon of SsrB extended considerably beyond T3SS-2 and its substrates, while that of HilA did not. That at least two SsrB-regulated processes streamline existence in the intracellular niche afforded by T3SS-2 seems to be a predictable outcome of evolution and natural selection. However, and importantly, these are the first such functions to be implicated as being SsrB dependent. The concept of T3SS-associated transcription factors coordinating manipulations of host cells together with distinct bacterial processes for increased efficiency has unrealized implications for numerous host-pathogen systems.


Assuntos
Adaptação Fisiológica/genética , Proteínas de Bactérias/metabolismo , Sistemas de Secreção Bacterianos/genética , Salmonella/genética , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Proteínas de Bactérias/genética , Biologia Computacional , Grupo dos Citocromos b/genética , Grupo dos Citocromos b/metabolismo , Ferritinas/genética , Ferritinas/metabolismo , Regulação Bacteriana da Expressão Gênica , Transferência Genética Horizontal , Ilhas Genômicas , Família Multigênica , Fenótipo , Proteômica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Regulon , Salmonella/crescimento & desenvolvimento , Transativadores/genética , Fatores de Transcrição/genética , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
14.
Mob Genet Elements ; 1(2): 118-121, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22016859

RESUMO

A major virulence mechanism used by pathogenic Gram-negative bacteria is the delivery of effector proteins from the bacterial cytoplasm into host cells by type III secretion. Typically, genes encoding type III secretion systems (T3SS) and effectors have been horizontally acquired by the bacteria that employ them. In proteobacteria, and especially Salmonella, and attaching and effacing (A/E) pathogens, the genetic structure of these systems presents as a large locus encoding a T3SS with a small number of effectors, plus numerous small unlinked loci encoding additional individual effectors. We discuss the generation of novel effectors, and the evolution of G+C content following acquisition. We also consider the currently held view that each locus has been acquired individually, as well as propose an alternative where recombination may have redistributed and broken up clusters of effectors. It is clear that the evolution of this virulence strategy is highly complex and challenging to analyze.

15.
Sci Signal ; 4(191): rs9, 2011 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-21934108

RESUMO

Salmonella enterica is a bacterial pathogen that causes gastroenteritis and typhoid fever. Virulence is achieved by two type III secretion systems that translocate effector proteins into host cells, where they mimic or block host protein function. Effectors translocated into host cells by the first type III secretion system facilitate invasion and stimulate intracellular signaling cascades leading to inflammation. Here, we performed global temporal analysis of host signaling events induced during the initial stages of Salmonella infection and identified the dynamics of host protein phosphorylation as well as differences between growth factor-stimulated and bacteria-induced signaling. Informatics analysis predicted that sites with altered phosphorylation in infected cells were targeted by the serine-threonine kinases Akt, protein kinase C, and Pim and that up to half of the host phosphorylation events detected after Salmonella infection required the effector protein SopB. Our data reveal extensive manipulation of host phosphorylation cascades by this Salmonella effector and provide a detailed map of the events leading to intestinal inflammation, which is the crucial host response that enables Salmonella to proliferate in the intestine.


Assuntos
Proteínas de Bactérias/metabolismo , Interações Hospedeiro-Patógeno/fisiologia , Proteínas Quinases/metabolismo , Infecções por Salmonella/enzimologia , Salmonella enterica/fisiologia , Transdução de Sinais , Células HeLa , Humanos , Fosforilação , Proteômica/métodos , Infecções por Salmonella/microbiologia
16.
PLoS One ; 6(3): e17824, 2011 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-21445262

RESUMO

Salmonella enterica is a species of bacteria that is a major cause of enteritis across the globe, while certain serovars cause typhoid, a more serious disease associated with a significant mortality rate. Type III secreted effectors are major contributors to the pathogenesis of Salmonella infections. Genes encoding effectors are acquired via horizontal gene transfer, and a subset are encoded within active phage lysogens. Because the acquisition of effectors is in flux, the complement of effectors possessed by various Salmonella strains frequently differs. By comparing the genome sequences of S. enterica serovar Typhimurium strain SL1344 with LT2, we identified a gene with significant similarity to SseK/NleB type III secreted effector proteins within a phage ST64B lysogen that is absent from LT2. We have named this gene sseK3. SseK3 was co-regulated with the SPI-2 type III secretion system in vitro and inside host cells, and was also injected into infected host cells. While no role for SseK3 in virulence could be identified, a role for the other family members in murine typhoid was found. SseK3 and other phage-encoded effectors were found to have a significant but sparse distribution in the available Salmonella genome sequences, indicating the potential for more uncharacterised effectors to be present in less studied serovars. These phage-encoded effectors may be principle subjects of contemporary selective processes shaping Salmonella-host interactions.


Assuntos
Proteínas de Bactérias/genética , Fagos de Salmonella/genética , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Sequência de Bases , Linhagem Celular , Primers do DNA , Citometria de Fluxo , Genes Virais , Genoma Viral , Camundongos , Dados de Sequência Molecular , Transporte Proteico , Salmonella/patogenicidade , Homologia de Sequência de Aminoácidos , Virulência
17.
Cell Microbiol ; 9(12): 2839-55, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17696999

RESUMO

The intracellular bacterial pathogen, Salmonella enterica serovar Typhimurium (S. typhimurium), causes disease in a variety of hosts. To invade and replicate in host cells, these bacteria subvert host molecular machinery using bacterial proteins, called effectors, which they translocate into host cells using specialized protein delivery systems. One of these effectors, SopD, contributes to gastroenteritis, systemic virulence and persistence of S. typhimurium in animal models of infection. Recently, SopD has been implicated in invasion of polarized epithelial cells and here we investigate the features of SopD-mediated invasion. We show that SopD plays a role in membrane fission and macropinosome formation during S. typhimurium invasion, events previously shown to be mediated by the SopB effector. We further demonstrate that SopD acts cooperatively with SopB to promote these events during invasion. Using live cell imaging we show that a SopD-GFP fusion does not localize to HeLa cell cytosol as previously described, but instead is membrane associated. Upon S. typhimurium infection of these cells, SopD-GFP is recruited to the invasion site, and this recruitment required the phosphatase activity of SopB. Our findings demonstrate a role for SopD in manipulation of host-cell membrane during S. typhimurium invasion and reveal the nature of its cooperative action with SopB.


Assuntos
Proteínas de Bactérias/fisiologia , Infecções por Salmonella/microbiologia , Salmonella typhimurium/fisiologia , Proteínas de Bactérias/análise , Proteínas de Bactérias/genética , Membrana Celular/química , Endocitose/fisiologia , Células Epiteliais/microbiologia , Deleção de Genes , Genes Reporter , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Células HeLa , Humanos , Proteínas Recombinantes de Fusão/análise , Proteínas Recombinantes de Fusão/genética , Fatores de Virulência/fisiologia
18.
Microbiology (Reading) ; 153(Pt 10): 3517-3526, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17906149

RESUMO

The ability of Salmonella enterica serovar Typhimurium (S. Typhimurium) to penetrate the intestinal epithelium is key to its pathogenesis. Bacterial invasion can be seen as a two-step process initially requiring adherence to the host cell surface followed by internalization into the host cell. Evidence suggests that adherence of S. Typhimurium to host cells is receptor-mediated; however, the host cell receptor(s) has/have not been identified. Internalization of S. Typhimurium absolutely requires the actin cytoskeleton yet only a few of the cytoskeletal components involved in this process have been identified. In order to identify host proteins that may play a role in S. Typhimurium invasion, the recruitment of actin-associated proteins was investigated. The contribution of recruited Src homology 2 adaptor proteins to invasion was further investigated and it was found that, while not involved in bacterial internalization itself, the adaptors Nck and ShcA influenced adherence of S. Typhimurium to non-phagocytic cells.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Aderência Bacteriana/fisiologia , Proteínas Oncogênicas/fisiologia , Salmonella typhimurium/fisiologia , Aderência Bacteriana/genética , Células HeLa , Humanos , Salmonella typhimurium/genética , Proteínas Adaptadoras da Sinalização Shc , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src
19.
Infect Immun ; 75(2): 574-80, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17158898

RESUMO

Bacterial pathogens use horizontal gene transfer to acquire virulence factors that influence host colonization, alter virulence traits, and ultimately shape the outcome of disease following infection. One hallmark of the host-pathogen interaction is the prokaryotic type III secretion system that translocates virulence factors into host cells during infection. Salmonella enterica possesses two type III secretion systems that are utilized during host colonization and intracellular replication. Salmonella pathogenicity island 2 (SPI2) is a genomic island containing approximately 30 contiguous genes required to assemble a functional secretion system including the two-component regulatory system called SsrA-SsrB that positively regulates transcription of the secretion apparatus. We used transcriptional profiling with DNA microarrays to search for genes that coregulate with the SPI2 type III secretion machinery in an SsrB-dependent manner. Here we report the identification of a Salmonella-specific translocated effector called SseL that is required for full virulence during murine typhoid-like disease. Analysis of infected macrophages using fluorescence-activated cell sorting revealed that sseL is induced inside cells and requires SsrB for expression. SseL is retained predominantly in the cytoplasm of infected cells following translocation by the type III system encoded in SPI2. Animal infection experiments with sseL mutant bacteria indicate that integration of SseL into the SsrB response regulatory system contributes to systemic virulence of this pathogen.


Assuntos
Proteínas de Bactérias/fisiologia , Ilhas Genômicas/genética , Transporte Proteico , Salmonelose Animal/microbiologia , Salmonella typhimurium/fisiologia , Salmonella typhimurium/patogenicidade , Fatores de Virulência/fisiologia , Animais , Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Linhagem Celular , Citoplasma/química , Modelos Animais de Doenças , Células Epiteliais/microbiologia , Citometria de Fluxo , Deleção de Genes , Regulação Bacteriana da Expressão Gênica , Humanos , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Mutagênese Insercional , Análise de Sequência com Séries de Oligonucleotídeos , Transporte Proteico/genética , Salmonella typhimurium/genética , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia , Fatores de Virulência/biossíntese , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
20.
Cell Microbiol ; 8(12): 1946-57, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16869830

RESUMO

Salmonella enterica serovar Typhimurium is a major cause of human gastroenteritis. Infection of epithelial monolayers by S. Typhimurium disrupts tight junctions that normally maintain the intestinal barrier and regulate cell polarity. Tight junction disruption is dependent upon the Salmonella pathogenicity island-1 (SPI-1) type 3 secretion system but the specific effectors involved have not been identified. In this study we demonstrate that SopB, SopE, SopE2 and SipA are the SPI-1-secreted effectors responsible for disruption of tight junction structure and function. Tight junction disruption by S. Typhimurium was prevented by inhibiting host protein geranylgeranylation but was not dependent on host protein synthesis or secretion of host-derived products. Unlike wild-type S. Typhimurium, DeltasopB, DeltasopE/E2, DeltasipA, or DeltasipA/sopB mutants, DeltasopB/E/E2 and DeltasipA/sopE/E2 mutants were unable to increase the permeability of polarized epithelial monolayers, did not disrupt the distribution or levels of ZO-1 and occludin, and did not alter cell polarity. These data suggest that SPI-1-secreted effectors utilize their ability to stimulate Rho family GTPases to disrupt tight junction structure and function.


Assuntos
Proteínas de Bactérias/fisiologia , Células Epiteliais/microbiologia , Fatores de Troca do Nucleotídeo Guanina/fisiologia , Mucosa Intestinal/microbiologia , Proteínas dos Microfilamentos/fisiologia , Salmonella typhimurium/patogenicidade , Junções Íntimas/ultraestrutura , Animais , Células CACO-2 , Linhagem Celular , Cães , Células Epiteliais/fisiologia , Células Epiteliais/ultraestrutura , Humanos , Mucosa Intestinal/fisiologia , Mucosa Intestinal/ultraestrutura , Salmonella typhimurium/fisiologia , Junções Íntimas/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA