Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Infect Immun ; 79(9): 3552-62, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21746856

RESUMO

Enteropathogenic Escherichia coli (EPEC) and enterohemorrhagic E. coli are noninvasive attaching and effacing (A/E) bacterial pathogens that cause intestinal inflammation and severe diarrheal disease. These pathogens utilize a type III secretion system to deliver effector proteins into host epithelial cells, modulating diverse cellular functions, including the release of the chemokine interleukin-8 (IL-8). While studies have implicated the effectors NleE (non-locus of enterocyte effacement [LEE]-encoded effector E) and NleH1 in suppressing IL-8 release, by preventing NF-κB nuclear translocation, the impact of these effectors only partially replicates the immunosuppressive actions of wild-type EPEC, suggesting another effector or effectors are involved. Testing an array of EPEC mutants, we identified the non-LEE-encoded effector C (NleC) as also suppressing IL-8 release. Infection by ΔnleC EPEC led to exaggerated IL-8 release from infected Caco-2 and HT-29 epithelial cells. NleC localized to EPEC-induced pedestals, with signaling studies revealing NleC inhibits both NF-κB and p38 mitogen-activated protein kinase (MAPK) activation. Using Citrobacter rodentium, a mouse-adapted A/E bacterium, we found that ΔnleC and wild-type C. rodentium-infected mice carried similar pathogen burdens, yet ΔnleC strain infection led to worsened colitis. Similarly, infection with ΔnleC C. rodentium in a cecal loop model induced significantly greater chemokine responses than infection with wild-type bacteria. These studies thus advance our understanding of how A/E pathogens subvert host inflammatory responses.


Assuntos
Citrobacter rodentium/patogenicidade , Infecções por Enterobacteriaceae/microbiologia , Escherichia coli Enteropatogênica/patogenicidade , Proteínas de Escherichia coli/metabolismo , NF-kappa B/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Aderência Bacteriana , Células CACO-2 , Quimiocinas/metabolismo , Citrobacter rodentium/genética , Colite/microbiologia , Infecções por Enterobacteriaceae/imunologia , Escherichia coli Enteropatogênica/genética , Células Epiteliais/imunologia , Proteínas de Escherichia coli/genética , Imunofluorescência , Células HT29 , Humanos , Interleucina-8/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Reação em Cadeia da Polimerase
3.
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
4.
Proc Natl Acad Sci U S A ; 106(10): 3982-7, 2009 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-19234126

RESUMO

The acquisition of DNA by horizontal gene transfer enables bacteria to adapt to previously unexploited ecological niches. Although horizontal gene transfer and mutation of protein-coding sequences are well-recognized forms of pathogen evolution, the evolutionary significance of cis-regulatory mutations in creating phenotypic diversity through altered transcriptional outputs is not known. We show the significance of regulatory mutation for pathogen evolution by mapping and then rewiring a cis-regulatory module controlling a gene required for murine typhoid. Acquisition of a binding site for the Salmonella pathogenicity island-2 regulator, SsrB, enabled the srfN gene, ancestral to the Salmonella genus, to play a role in pathoadaptation of S. typhimurium to a host animal. We identified the evolved cis-regulatory module and quantified the fitness gain that this regulatory output accrues for the bacterium using competitive infections of host animals. Our findings highlight a mechanism of pathogen evolution involving regulatory mutation that is selected because of the fitness advantage the new regulatory output provides the incipient clones.


Assuntos
Adaptação Fisiológica , Espaço Intracelular/microbiologia , Sequências Reguladoras de Ácido Nucleico/genética , Salmonella/genética , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sequência de Bases , Regulação Bacteriana da Expressão Gênica , Genes Bacterianos , Interações Hospedeiro-Patógeno , Camundongos , Dados de Sequência Molecular , Mutação/genética , Regiões Promotoras Genéticas/genética , Ligação Proteica , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Febre Tifoide/metabolismo
5.
Mol Microbiol ; 68(1): 124-38, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18333885

RESUMO

Antibodies from malaria-exposed individuals can agglutinate merozoites released from Plasmodium schizonts, thereby preventing them from invading new erythrocytes. Merozoite coat proteins attached to the plasma membrane are major targets for host antibodies and are therefore considered important malaria vaccine candidates. Prominent among these is the abundant glycosylphosphatidylinositol (GPI)-anchored merozoite surface protein 1 (MSP1) and particularly its C-terminal fragment (MSP1(19)) comprised of two epidermal growth factor (EGF)-like modules. In this paper, we revisit the role of agglutination and immunity using transgenic fluorescent marker proteins. We describe expression of heterologous MSP1(19)'miniproteins' on the surface of Plasmodium falciparum merozoites. To correctly express these proteins, we determined that GPI-anchoring and the presence of a signal sequence do not allow default export of proteins from the endoplasmic reticulum to merozoite surface and that extra sequence elements are required. The EGFs are insufficient for correct trafficking unless they are fused to additional residues that normally reside upstream of this fragment. Antibodies specifically targeting the surface-expressed miniprotein can inhibit erythrocyte invasion in vitro despite the presence of endogenous MSP1. Using a line expressing a green fluorescent protein-MSP1 fusion protein, we demonstrate that one mode of inhibition by antibodies targeting the MSP1(19) domain is the rapid agglutinating of merozoites prior to erythrocyte attachment.


Assuntos
Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/imunologia , Proteína 1 de Superfície de Merozoito/metabolismo , Plasmodium falciparum/metabolismo , Motivos de Aminoácidos , Animais , Antígenos de Protozoários/química , Antígenos de Protozoários/genética , Membrana Celular/metabolismo , Retículo Endoplasmático/metabolismo , Glicosilfosfatidilinositóis/metabolismo , Proteína 1 de Superfície de Merozoito/genética , Proteína 1 de Superfície de Merozoito/imunologia , Modelos Biológicos , Plasmodium falciparum/genética , Plasmodium falciparum/imunologia , Estrutura Terciária de Proteína , Transporte Proteico/fisiologia , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
6.
Appl Environ Microbiol ; 74(7): 2153-60, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18245257

RESUMO

Shiga toxin-producing Escherichia coli (STEC) strains are commensal bacteria in cattle with high potential for environmental and zoonotic transmission to humans. Although O157:H7 is the most common STEC serotype, there is growing concern over the emergence of more than 200 highly virulent non-O157 STEC serotypes that are globally distributed, several of which are associated with outbreaks and/or severe human illness such as hemolytic-uremic syndrome (HUS) and hemorrhagic colitis. At present, the underlying genetic basis of virulence potential in non-O157 STEC is unknown, although horizontal gene transfer and the acquisition of new pathogenicity islands are an expected origin. We used seropathotype classification as a framework to identify genetic elements that distinguish non-O157 STEC strains posing a serious risk to humans from STEC strains that are not associated with severe and epidemic disease. We report the identification of three genomic islands encoding non-LEE effector (nle) genes and 14 individual nle genes in non-O157 STEC strains that correlate independently with outbreak and HUS potential in humans. The implications for transmissible zoonotic spread and public health are discussed. These results and methods offer a molecular risk assessment strategy to rapidly recognize and respond to non-O157 STEC strains from environmental and animal sources that might pose serious public health risks to humans.


Assuntos
Infecções por Escherichia coli/diagnóstico , Ilhas Genômicas/genética , Síndrome Hemolítico-Urêmica/microbiologia , Escherichia coli Shiga Toxigênica/classificação , Fatores de Virulência/genética , Animais , Colo/microbiologia , Genes Bacterianos , Ilhas Genômicas/fisiologia , Síndrome Hemolítico-Urêmica/epidemiologia , Humanos , Saúde Pública , Toxinas Shiga/biossíntese , Escherichia coli Shiga Toxigênica/genética , Escherichia coli Shiga Toxigênica/isolamento & purificação
7.
FEMS Microbiol Lett ; 281(1): 98-107, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18279332

RESUMO

Enterohemorrhagic Escherichia coli (EHEC) is a water- and food-borne pathogen that causes hemorrhagic colitis. EHEC uses a type III secretion system (T3SS) to translocate effector proteins that subvert host cell function. T3SS-substrates encoded outside of the locus of enterocyte effacement are important to E. coli pathogenesis. We discovered an EHEC secreted protein, NleF, encoded by z6020 in O-island 71 of E. coli EDL933 that we hypothesized to be a T3SS substrate. Experiments are presented that probe the function of NleF and its role in virulence. Immunoblotting of secreted and translocated proteins suggest that NleF is secreted by the T3SS and is translocated into host cells in vitro where it localizes to the host cytoplasm. Infection of HeLa cells with E. coli possessing or lacking nleF and transient expression of NleF-GFP via transfection did not reveal a significant role for NleF in several assays of bacterial adherence, host cytoskeletal remodeling, or host protein secretion. However, competitive coinfection of mice with Citrobacter rodentium strains possessing or lacking nleF suggested a contribution of NleF to bacterial colonization. Challenge of gnotobiotic piglets also revealed a role for NleF in colonization of the piglet colon and rectoanal junction.


Assuntos
Citrobacter rodentium/genética , Citrobacter rodentium/patogenicidade , Escherichia coli O157/genética , Escherichia coli O157/patogenicidade , Proteínas de Escherichia coli/metabolismo , Fatores de Virulência/metabolismo , Animais , Células COS , Chlorocebus aethiops , Citoplasma/química , Infecções por Enterobacteriaceae/microbiologia , Células Epiteliais/química , Células Epiteliais/microbiologia , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Feminino , Trato Gastrointestinal/microbiologia , Células HeLa , Humanos , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Suínos , Virulência , Fatores de Virulência/química , Fatores de Virulência/genética
8.
Cell Host Microbe ; 2(2): 119-29, 2007 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-18005726

RESUMO

While the normal microbiota has been implicated as a critical defense against invading pathogens, the impact of enteropathogenic infection and host inflammation on intestinal microbial communities has not been elucidated. Using mouse models of Citrobacter rodentium, which closely mimics human diarrheal pathogens inducing host intestinal inflammation, and Campylobacter jejuni infection, as well as chemically and genetically induced models of intestinal inflammation, we demonstrate that host-mediated inflammation in response to an infecting agent, a chemical trigger, or genetic predisposition markedly alters the colonic microbial community. While eliminating a subset of indigenous microbiota, host-mediated inflammation supported the growth of either the resident or introduced aerobic bacteria, particularly of the Enterobacteriaceae family. Further, assault by an enteropathogen and host-mediated inflammation combined to significantly reduce the total numbers of resident colonic bacteria. These findings underscore the importance of intestinal microbial ecosystems in infectious colitis and noninfectious intestinal inflammatory conditions, such as inflammatory bowel disease.


Assuntos
Infecções por Enterobacteriaceae/patologia , Enterobacteriaceae/crescimento & desenvolvimento , Inflamação/microbiologia , Inflamação/fisiopatologia , Enteropatias/microbiologia , Intestinos/microbiologia , Animais , Infecções por Escherichia coli/fisiopatologia , Humanos , Hibridização in Situ Fluorescente , Interleucina-10/deficiência , Enteropatias/patologia , Mamíferos/microbiologia , Camundongos
9.
Cell Host Microbe ; 2(3): 204, 2007 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-18030708

RESUMO

While the normal microbiota has been implicated as a critical defense against invading pathogens, the impact of enteropathogenic infection and host inflammation on intestinal microbial communities has not been elucidated. Using mouse models of Citrobacter rodentium, which closely mimics human diarrheal pathogens inducing host intestinal inflammation, and Campylobacter jejuni infection, as well as chemically and genetically induced models of intestinal inflammation, we demonstrate that host-mediated inflammation in response to an infecting agent, a chemical trigger, or genetic predisposition markedly alters the colonic microbial community. While eliminating a subset of indigenous microbiota, host-mediated inflammation supported the growth of either the resident or introduced aerobic bacteria, particularly of the Enterobacteriaceae family. Further, assault by an enteropathogen and host-mediated inflammation combined to significantly reduce the total numbers of resident colonic bacteria. These findings underscore the importance of intestinal microbial ecosystems in infectious colitis and noninfectious intestinal inflammatory conditions,such as inflammatory bowel disease.


Assuntos
Infecções por Enterobacteriaceae/microbiologia , Enterobacteriaceae/crescimento & desenvolvimento , Trato Gastrointestinal/microbiologia , Trato Gastrointestinal/patologia , Inflamação/microbiologia , Animais , Biodiversidade , Infecções por Campylobacter/microbiologia , Infecções por Campylobacter/patologia , Campylobacter jejuni/crescimento & desenvolvimento , Citrobacter rodentium/crescimento & desenvolvimento , Contagem de Colônia Microbiana , Infecções por Enterobacteriaceae/patologia , Camundongos
10.
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
11.
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
12.
Microbes Infect ; 9(3): 400-7, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17317262

RESUMO

Severe disease caused by Shiga toxin-producing Escherichia coli (STEC) has been associated with a pathogenicity island, O-Island 122, which encodes the type III secretion system-effector NleE. Here we show that full virulence of the related attaching and effacing mouse pathogen Citrobacter rodentium requires NleE. Relative to wild-type bacteria, nleE-mutant C. rodentium are attenuated for colonisation in mice in both single and mixed infections. Examination of the ability of nleE-mutant bacteria to induce pathologic change in vivo revealed that nleE-mutant bacteria induce significantly less pathologic change than wild-type bacteria in susceptible mice. Consistent with these results, mice infected with nleE-mutant bacteria exhibit delayed mortality. These results suggested that pathologic change during attaching and effacing pathogen infection may associate with the degree of pathogen colonisation. Using mutants of 23 type III secretion genes, including the type III effectors nleC, nleD, nleE and nleF, the association of pathologic change with the ability of these mutants to colonise mice was examined. The induction of in vivo disease correlates strongly with the degree of colonisation, suggesting that the colonisation advantage type III secretion genes afford the bacteria, contribute to, and are required for, full virulence.


Assuntos
Proteínas de Bactérias , Citrobacter rodentium/fisiologia , Citrobacter rodentium/patogenicidade , Ilhas Genômicas , Fatores de Virulência , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Citrobacter rodentium/genética , Colo/microbiologia , Colo/patologia , Sequência Conservada , Infecções por Enterobacteriaceae/microbiologia , Infecções por Enterobacteriaceae/patologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Mutação , Virulência , Fatores de Virulência/química , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
13.
Microb Pathog ; 42(2-3): 62-71, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17174521

RESUMO

Verotoxin-producing Escherichia coli (VTEC) O157:H7 inhibits interferon-gamma-stimulated tyrosine phosphorylation of signal transducer and activator of transcription (Stat)-1 in epithelial cells, independent of Verotoxins and the locus of enterocyte effacement pathogenicity island. Although E. coli O157:H7 is the major cause of disease in humans, non-O157:H7 VTEC also cause human disease. However, the virulence properties of non-O157:H7 VTEC are less well characterized. The aims of this study were to define the ability of VTEC strains of differing seropathotypes (classified as A-E) to inhibit interferon-gamma stimulated Stat1-phosphorylation and to further characterize the bacterial-derived inhibitory factor. Confluent T84 and HEp-2 cells were infected with VTEC strains (MOI 100:1, 6h, 37 degrees C), and then stimulated with interferon-gamma (50 ng/mL) for 0.5h at 37 degrees C. Whole-cell protein extracts of infected cells were collected and prepared for immunoblotting to detect tyrosine phosphorylation of Stat1. The effects of E. coli O55 strains, the evolutionary precursors of VTEC, on Stat1-tyrosine phosphorylation were also determined. The effects of isogenic mutants of O-islands 47 and 122 were tested to determine the role of genes encoded on these putative pathogenicity islands in mediating VTEC inhibition of the interferon-gamma-Stat1 signaling cascade. To evaluate potential mechanism(s) of inhibition, VTEC O157:H7-infected cells were treated with pharmacological inhibitors, including, wortmannin and LY294002. Relative to uninfected cells, Stat1-tyrosine phosphorylation was significantly reduced after 6h infection of both T84 and HEp-2 cells by VTEC strains of all five seropathotypes. E. coli O55 strains, but not enteropathogenic E. coli (EPEC), also caused inhibition of Stat1-tyrosine phosphorylation, suggesting that this effect was acquired early in the evolution of VTEC. Stat1-activation did not recover in epithelial cells infected with isogenic mutants of O-islands 47 and 122, indicating that the inhibitory factor was not contained in these genomic regions. Stat1-phosphorylation remained intact when VTEC-infected cells were treated with wortmannin (0-100 nM), but not by treatment with the more specific PI3-kinase inhibitor, LY294002. Inhibition of interferon-gamma stimulated Stat1-tyrosine phosphorylation by VTEC of multiple seropathotypes indicates the presence of a common inhibitory factor that is independent of bacterial virulence in humans. The results of treatment with wortmannin suggest that the bacterial-derived inhibitory factor employs host cell signal transduction to mediate inhibition of Stat1-activation.


Assuntos
Infecções por Escherichia coli/metabolismo , Escherichia coli/metabolismo , Interferon gama/metabolismo , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais , Androstadienos/farmacologia , Linhagem Celular Tumoral , Cromonas/farmacologia , Inibidores Enzimáticos/farmacologia , Escherichia coli/classificação , Escherichia coli/patogenicidade , Ilhas Genômicas/genética , Humanos , Morfolinas/farmacologia , Mutação , Fosfatidilinositol 3-Quinases/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Toxinas Shiga/metabolismo , Especificidade da Espécie , Tirosina/metabolismo , Virulência , Wortmanina
14.
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
15.
J Infect Dis ; 194(6): 819-27, 2006 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-16941350

RESUMO

Although O157:H7 Shiga toxin-producing Escherichia coli (STEC) are the predominant cause of hemolytic-uremic syndrome (HUS) in the world, non-O157:H7 serotypes are a medically important cause of HUS that are underdetected by current diagnostic approaches. Because Shiga toxin is necessary but not sufficient to cause HUS, identifying the virulence determinants that predict severe disease after non-O157 STEC infection is of paramount importance. Disease caused by O157:H7 STEC has been associated with a 26-gene pathogenicity island known as O island (OI) 122. To assess the public-health significance of this pathogenicity island, we examined the association between OI122 genes and outbreaks and HUS after non-O157 STEC infection. We found that a subset of OI122 genes is independently associated with outbreaks and HUS after infection with non-O157 STEC. The presence of multiple virulence genes in non-O157 serotypes strengthened this association, which suggests that the additive effects of a variable repertoire of virulence genes contribute to disease severity. In vivo, Citrobacter rodentium mutants lacking outbreak- and HUS-associated genes were deficient for virulence in mice; in particular, nleB mutant bacteria were unable to cause mortality in mice. The present study shows that virulence genes associated epidemiologically with outbreaks and HUS after non-O157 STEC infection are pivotal to the initiation, progression, and outcome of in vivo disease.


Assuntos
Infecções por Escherichia coli/microbiologia , Escherichia coli/genética , Ilhas Genômicas/genética , Síndrome Hemolítico-Urêmica/microbiologia , Animais , Citrobacter rodentium/genética , Citrobacter rodentium/patogenicidade , Colo/microbiologia , Primers do DNA , Modelos Animais de Doenças , Surtos de Doenças , Escherichia coli/patogenicidade , Infecções por Escherichia coli/epidemiologia , Ilhas Genômicas/fisiologia , Síndrome Hemolítico-Urêmica/epidemiologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Prevalência , Toxinas Shiga/biossíntese , Análise de Sobrevida , Fatores de Tempo , Virulência/genética , Fatores de Virulência/genética
16.
Cell Microbiol ; 8(10): 1669-86, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16759225

RESUMO

Enteropathogenic Escherichia coli (EPEC) and enterohaemorrhagic E. coli are non-invasive attaching/effacing (A/E) bacterial pathogens that infect their host's intestinal epithelium, causing severe diarrhoeal disease. These bacteria utilize a type III secretion apparatus to deliver effector molecules into host cells, subverting cellular function. Mitochondrial associated protein (Map) is a multifunctional effector protein that targets host cell mitochondria and contributes to infection-induced epithelial barrier dysfunction in vitro. Unfortunately, the relevance of these actions to the pathogenesis of EPEC-induced disease is uncertain. Using Citrobacter rodentium, a mouse-adapted A/E bacterium, we found that Map colocalized with host cell mitochondria, and that in vivo infection led to a disruption of mitochondrial morphology in infected colonocytes as assessed by electron microscopy. Histochemical staining for the mitochondrial enzyme succinate dehydrogenase also revealed a significant loss of mitochondrial respiratory function in the infected intestinal epithelium; however, both pathologies were attenuated in mice infected with a Deltamap strain. C. rodentium Map was also implicated in the disruption of epithelial barrier function both in vitro and in vivo. These studies thus advance our understanding of how A/E pathogens subvert host cell functions and cause disease, demonstrating that Map contributes to the functional disruption of the intestinal epithelium during enteric infection by C. rodentium.


Assuntos
Proteínas de Bactérias/fisiologia , Citrobacter rodentium/fisiologia , Infecções por Enterobacteriaceae/patologia , Mucosa Intestinal/patologia , Mitocôndrias/metabolismo , Animais , Linhagem Celular Tumoral , Citrobacter rodentium/patogenicidade , Colo/microbiologia , Colo/patologia , Contagem de Colônia Microbiana , Infecções por Enterobacteriaceae/microbiologia , Infecções por Enterobacteriaceae/fisiopatologia , Deleção de Genes , Células HeLa , Humanos , Mucosa Intestinal/microbiologia , Potenciais da Membrana , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/enzimologia , Mitocôndrias/patologia , Membranas Mitocondriais , Succinato Desidrogenase/metabolismo
17.
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
18.
Cell Microbiol ; 8(4): 634-45, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16548889

RESUMO

Diarrhoea is a hallmark of infections by the human attaching and effacing (A/E) pathogens, enterohaemorrhagic Escherichia coli (EHEC) and enteropathogenic E. coli (EPEC). Although the mechanisms underlying diarrhoea induced by these pathogens remain unknown, cell culture results have suggested that these pathogens may target tight junctions. Tight junctions in the colon function as physical intercellular barriers that separate and prevent mixing of the luminal contents with adlumenal regions of the epithelium. Consequently, it is thought that the disruption of intestinal epithelial tight junctions by A/E pathogens could result in a loss of barrier function in the alimentary tract; however, this remains unexamined. Here we demonstrate for the first time that A/E pathogen infection results in the morphological alteration of tight junctions during natural disease. Tight junction alteration, characterized by relocalization of the transmembrane tight junction proteins claudin 1, 3 and 5, is a functional disruption; molecular tracers, which do not normally penetrate uninfected epithelia, pass across pathogen-infected epithelia. Functional junction disruption occurs with a concomitant increase in colon luminal water content. The effects on tissue are dependent upon the bacterial type III effector EspF (E. coli secreted protein F), because bacteria lacking EspF, while able to colonize, are defective for junction disruption and result in decreased proportions of water in the colon compared with wild-type infection. These results suggest that the diarrhoea induced by A/E pathogens occurs as part of functional tight junction disruption.


Assuntos
Aderência Bacteriana , Citrobacter rodentium/fisiologia , Infecções por Enterobacteriaceae/microbiologia , Junções Íntimas/fisiologia , Animais , Proteínas de Bactérias/metabolismo , Citrobacter rodentium/genética , Claudina-1 , Claudina-3 , Claudina-5 , Colo/microbiologia , Colo/ultraestrutura , Infecções por Enterobacteriaceae/patologia , Feminino , Mucosa Intestinal/microbiologia , Mucosa Intestinal/ultraestrutura , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão , Mutação , Junções Íntimas/microbiologia
19.
Proc Natl Acad Sci U S A ; 102(48): 17460-5, 2005 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-16301528

RESUMO

Salmonella enterica relies on a type III secretion system encoded in Salmonella pathogenicity island-2 (SPI-2) to survive and replicate within macrophages at systemic sites during typhoid. SPI-2 virulence is induced upon entry into macrophages, but the mechanisms of SPI-2 gene control in vivo remain unclear, particularly with regard to negative regulators that control the contextual activation of SPI-2. Here, we identified and characterized YdgT as a negative modulator of the SPI-2 pathogenicity island and established that this negative regulation is central to systemic pathogenesis because ydgT mutants overexpressing typhoid virulence genes were ultimately attenuated during infection. ydgT mutants displayed a biphasic virulence phenotype during in vivo competitive infections that consisted of an early "gain-of-virulence" dependent on SPI-2 activation, followed by attenuation later in infection indicating that proper contextual regulation of SPI-2 by YdgT is necessary for full virulence during systemic colonization. These data suggest that overexpression of virulence-associated type III secretion genes can have an adverse effect on bacterial pathogenesis in vivo.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação Bacteriana da Expressão Gênica/genética , Proteínas de Membrana/metabolismo , Salmonella typhi/patogenicidade , Febre Tifoide/metabolismo , Animais , Linhagem Celular , Proteínas de Ligação a DNA/genética , Perfilação da Expressão Gênica , Células HeLa , Humanos , Camundongos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Salmonella typhi/metabolismo , Virulência
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...