Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(44)2021 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-34706941

RESUMO

Type III secretion systems are multiprotein molecular machines required for the virulence of several important bacterial pathogens. The central element of these machines is the injectisome, a ∼5-Md multiprotein structure that mediates the delivery of bacterially encoded proteins into eukaryotic target cells. The injectisome is composed of a cytoplasmic sorting platform, and a membrane-embedded needle complex, which is made up of a multiring base and a needle-like filament that extends several nanometers from the bacterial surface. The needle filament is capped at its distal end by another substructure known as the tip complex, which is crucial for the translocation of effector proteins through the eukaryotic cell plasma membrane. Here we report the cryo-EM structure of the Salmonella Typhimurium needle tip complex docked onto the needle filament tip. Combined with a detailed analysis of structurally guided mutants, this study provides major insight into the assembly and function of this essential component of the type III secretion protein injection machine.


Assuntos
Salmonella typhimurium/ultraestrutura , Sistemas de Secreção Tipo III/ultraestrutura , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/ultraestrutura , Sistemas de Secreção Bacterianos/metabolismo , Sistemas de Secreção Bacterianos/ultraestrutura , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Microscopia Crioeletrônica/métodos , Citoesqueleto/metabolismo , Citosol/metabolismo , Transporte Proteico/fisiologia , Salmonella typhimurium/metabolismo , Salmonella typhimurium/patogenicidade , Sistemas de Secreção Tipo III/metabolismo , Sistemas de Secreção Tipo III/fisiologia
2.
Proc Natl Acad Sci U S A ; 118(2)2021 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-33397726

RESUMO

Enteropathogenic Escherichia coli (EPEC) and enterohemorrhagic Escherichia coli (EHEC) utilize a macromolecular type III secretion system (T3SS) to inject effector proteins into eukaryotic cells. This apparatus spans the inner and outer bacterial membranes and includes a helical needle protruding into the extracellular space. Thus far observed only in EPEC and EHEC and not found in other pathogenic Gram-negative bacteria that have a T3SS is an additional helical filament made by the EspA protein that forms a long extension to the needle, mediating both attachment to eukaryotic cells and transport of effector proteins through the intestinal mucus layer. Here, we present the structure of the EspA filament from EPEC at 3.4 Å resolution. The structure reveals that the EspA filament is a right-handed 1-start helical assembly with a conserved lumen architecture with respect to the needle to ensure the seamless transport of unfolded cargos en route to the target cell. This functional conservation is despite the fact that there is little apparent overall conservation at the level of sequence or structure with the needle. We also unveil the molecular details of the immunodominant EspA epitope that can now be exploited for the rational design of epitope display systems.


Assuntos
Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/ultraestrutura , Sistemas de Secreção Tipo III/metabolismo , Microscopia Crioeletrônica/métodos , Citoesqueleto/metabolismo , Escherichia coli Êntero-Hemorrágica/metabolismo , Escherichia coli Enteropatogênica/metabolismo , Escherichia coli/metabolismo , Proteínas de Escherichia coli/fisiologia , Humanos , Transporte Proteico/fisiologia , Sistemas de Secreção Tipo III/fisiologia
3.
Mol Microbiol ; 115(3): 395-411, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33251695

RESUMO

The type III secretion system is the common core of two bacterial molecular machines: the flagellum and the injectisome. The flagellum is the most widely distributed prokaryotic locomotion device, whereas the injectisome is a syringe-like apparatus for inter-kingdom protein translocation, which is essential for virulence in important human pathogens. The successful concept of the type III secretion system has been modified for different bacterial needs. It can be adapted to changing conditions, and was found to be a dynamic complex constantly exchanging components. In this review, we highlight the flexibility, adaptivity, and dynamic nature of the type III secretion system.


Assuntos
Adaptação Fisiológica , Fenômenos Fisiológicos Bacterianos , Proteínas de Bactérias/fisiologia , Flagelos/fisiologia , Sistemas de Translocação de Proteínas/fisiologia , Sistemas de Secreção Tipo III/fisiologia , Animais , Evolução Biológica , Humanos , Fatores de Virulência/metabolismo
4.
Mol Microbiol ; 115(2): 305-319, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33012037

RESUMO

Many Gram-negative pathogens use a type III secretion system (T3SS) to promote disease by injecting effector proteins into host cells. Common to many T3SSs is that injection of effector proteins is feedback inhibited. The mechanism of feedback inhibition and its role in pathogenesis are unclear. In the case of P. aeruginosa, the effector protein ExoS is central to limiting effector injection. ExoS is bifunctional, with an amino-terminal RhoGAP and a carboxy-terminal ADP-ribosyltransferase domain. We demonstrate that both domains are required to fully feedback inhibit effector injection. The RhoGAP-, but not the ADP-ribosyltransferase domain of the related effector protein ExoT also participates. Feedback inhibition does not involve translocator insertion nor pore-formation. Instead, feedback inhibition is due, in part, to a loss of the activating trigger for effector injection, and likely also decreased translocon stability. Surprisingly, feedback inhibition is abrogated in phagocytic cells. The lack of feedback inhibition in these cells requires phagocytic uptake of the bacteria, but cannot be explained through acidification of the phagosome or calcium limitation. Given that phagocytes are crucial for controlling P. aeruginosa infections, our data suggest that feedback inhibition allows P. aeruginosa to direct its effector arsenal against the cell types most damaging to its survival.


Assuntos
ADP Ribose Transferases/metabolismo , Toxinas Bacterianas/metabolismo , Pseudomonas aeruginosa/metabolismo , Sistemas de Secreção Tipo III/metabolismo , ADP Ribose Transferases/genética , ADP Ribose Transferases/fisiologia , Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/genética , Células Epiteliais/microbiologia , Retroalimentação Fisiológica/fisiologia , Proteínas Ativadoras de GTPase , Infecções por Pseudomonas/microbiologia , Sistemas de Secreção Tipo III/fisiologia
5.
PLoS Pathog ; 16(7): e1008680, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32673374

RESUMO

Pathogenic bacteria frequently acquire virulence traits via horizontal gene transfer, yet additional evolutionary innovations may be necessary to integrate newly acquired genes into existing regulatory pathways. The plant bacterial pathogen Pseudomonas syringae relies on a horizontally acquired type III secretion system (T3SS) to cause disease. T3SS-encoding genes are induced by plant-derived metabolites, yet how this regulation occurs, and how it evolved, is poorly understood. Here we report that the two-component system AauS-AauR and substrate-binding protein AatJ, proteins encoded by an acidic amino acid-transport (aat) and -utilization (aau) locus in P. syringae, directly regulate T3SS-encoding genes in response to host aspartate and glutamate signals. Mutants of P. syringae strain DC3000 lacking aauS, aauR or aatJ expressed lower levels of T3SS genes in response to aspartate and glutamate, and had decreased T3SS deployment and virulence during infection of Arabidopsis. We identified an AauR-binding motif (Rbm) upstream of genes encoding T3SS regulators HrpR and HrpS, and demonstrated that this Rbm is required for maximal T3SS deployment and virulence of DC3000. The Rbm upstream of hrpRS is conserved in all P. syringae strains with a canonical T3SS, suggesting AauR regulation of hrpRS is ancient. Consistent with a model of conserved function, an aauR deletion mutant of P. syringae strain B728a, a bean pathogen, had decreased T3SS expression and growth in host plants. Together, our data suggest that, upon acquisition of T3SS-encoding genes, a strain ancestral to P. syringae co-opted an existing AatJ-AauS-AauR pathway to regulate T3SS deployment in response to specific host metabolite signals.


Assuntos
Arabidopsis/microbiologia , Regulação Bacteriana da Expressão Gênica/fisiologia , Pseudomonas syringae/patogenicidade , Sistemas de Secreção Tipo III/fisiologia , Virulência/fisiologia , Doenças das Plantas/microbiologia
6.
Plant Cell Environ ; 45(1): 236-247, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34708407

RESUMO

Upon sensing attack by pathogens and insect herbivores, plants release complex mixtures of volatile compounds. Here, we show that the infection of lima bean (Phaseolus lunatus L.) plants with the non-host bacterial pathogen Pseudomonas syringae pv. tomato led to the production of microbe-induced plant volatiles (MIPVs). Surprisingly, the bacterial type III secretion system, which injects effector proteins directly into the plant cytosol to subvert host functions, was found to prime both intra- and inter-specific defense responses in neighbouring wild tobacco (Nicotiana benthamiana) plants. Screening of each of 16 effectors using the Pseudomonas fluorescens effector-to-host analyser revealed that an effector, HopP1, was responsible for immune activation in receiver tobacco plants. Further study demonstrated that 1-octen-3-ol, 3-octanone and 3-octanol are novel MIPVs emitted by the lima bean plant in a HopP1-dependent manner. Exposure to synthetic 1-octen-3-ol activated immunity in tobacco plants against a virulent pathogen Pseudomonas syringae pv. tabaci. Our results show for the first time that a bacterial type III effector can trigger the emission of C8 plant volatiles that mediate defense priming via plant-plant interactions. These results provide novel insights into the role of airborne chemicals in bacterial pathogen-induced inter-specific plant-plant interactions.


Assuntos
Interações Hospedeiro-Patógeno/fisiologia , Imunidade Vegetal , Pseudomonas syringae/patogenicidade , Sistemas de Secreção Tipo III/fisiologia , Compostos Orgânicos Voláteis/metabolismo , Ar , Capsicum/fisiologia , Cucumis sativus/fisiologia , Regulação da Expressão Gênica de Plantas , Octanóis/farmacologia , Phaseolus/fisiologia , Imunidade Vegetal/efeitos dos fármacos , Transdução de Sinais , Nicotiana/fisiologia , Compostos Orgânicos Voláteis/farmacologia
7.
PLoS Pathog ; 15(1): e1007565, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30668610

RESUMO

Many bacterial pathogens and symbionts use type III secretion machines to interact with their hosts by injecting bacterial effector proteins into host target cells. A central component of this complex machine is the cytoplasmic sorting platform, which orchestrates the engagement and preparation of type III secreted proteins for their delivery to the needle complex, the substructure of the type III secretion system that mediates their passage through the bacterial envelope. The sorting platform is thought to be a dynamic structure whose components alternate between assembled and disassembled states. However, how this dynamic behavior is controlled is not understood. In S. Typhimurium a core component of the sorting platform is SpaO, which is synthesized in two tandemly translated products, a full length (SpaOL) and a short form (SpaOS) composed of the C-terminal 101 amino acids. Here we show that in the absence of SpaOS the assembly of the needle substructure of the needle complex, which requires a functional sorting platform, can still occur although with reduced efficiency. Consistent with this observation, in the absence of SpaOS secretion of effectors proteins, which requires a fully assembled injectisome, is only slightly compromised. In the absence of SpaOS we detect a significant number of fully assembled needle complexes that are not associated with fully assembled sorting platforms. We also find that although binding of SpaOL to SpaOS can be detected in the absence of other components of the sorting platform, this interaction is not detected in the context of a fully assembled sorting platform suggesting that SpaOS may not be a core structural component of the sorting platform. Consistent with this observation we find that SpaOS and OrgB, a component of the sorting platform, share the same binding surface on SpaOL. We conclude that SpaOS regulates the assembly of the sorting platform during type III secretion.


Assuntos
Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Sistemas de Secreção Tipo III/metabolismo , Proteínas de Bactérias/fisiologia , Proteínas de Membrana/fisiologia , Isoformas de Proteínas , Transporte Proteico/fisiologia , Salmonella/metabolismo , Salmonella/patogenicidade , Salmonella typhimurium/metabolismo , Sistemas de Secreção Tipo III/fisiologia
8.
PLoS Pathog ; 15(7): e1007959, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31339948

RESUMO

The enteric bacterial pathogen Salmonella enterica serovar Typhimurium (S. Typhimurium), utilizes two type III secretion systems (T3SSs) to invade host cells, survive and replicate intracellularly. T3SS1 and its dedicated effector proteins are required for bacterial entry into non-phagocytic cells and establishment and trafficking of the nascent Salmonella-containing vacuole (SCV). Here we identify the first T3SS1 effector required to maintain the integrity of the nascent SCV as SopF. SopF associates with host cell membranes, either when translocated by bacteria or ectopically expressed. Recombinant SopF binds to multiple phosphoinositides in protein-lipid overlays, suggesting that it targets eukaryotic cell membranes via phospholipid interactions. In yeast, the subcellular localization of SopF is dependent on the activity of Mss4, a phosphatidylinositol 4-phosphate 5-kinase that generates PI(4,5)P2 from PI(4)P, indicating that membrane recruitment of SopF requires specific phospholipids. Ectopically expressed SopF partially colocalizes with specific phosphoinositide pools present on the plasma membrane in mammalian cells and with cytoskeletal-associated markers at the leading edge of cells. Translocated SopF concentrates on plasma membrane ruffles and around intracellular bacteria, presumably on the SCV. SopF is not required for bacterial invasion of non-phagocytic cells but is required for maintenance of the internalization vacuole membrane as infection with a S. Typhimurium ΔsopF mutant led to increased lysis of the SCV compared to wild type bacteria. Our structure-function analysis shows that the carboxy-terminal seven amino acids of SopF are essential for its membrane association in host cells and to promote SCV membrane stability. We also describe that SopF and another T3SS1 effector, SopB, act antagonistically to modulate nascent SCV membrane dynamics. In summary, our study highlights that a delicate balance of type III effector activities regulates the stability of the Salmonella internalization vacuole.


Assuntos
Salmonella typhimurium/fisiologia , Sistemas de Secreção Tipo III/fisiologia , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , Membrana Celular/microbiologia , Células HeLa , Interações entre Hospedeiro e Microrganismos/fisiologia , Humanos , Camundongos , Fosfatidilinositóis/metabolismo , Ligação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Salmonella typhimurium/genética , Salmonella typhimurium/patogenicidade , Sistemas de Secreção Tipo III/química , Sistemas de Secreção Tipo III/genética , Vacúolos/metabolismo , Vacúolos/microbiologia
9.
Int J Mol Sci ; 22(11)2021 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-34063760

RESUMO

Type III Secretion Systems (T3SSs) are multicomponent nanomachines located at the cell envelope of Gram-negative bacteria. Their main function is to transport bacterial proteins either extracellularly or directly into the eukaryotic host cell cytoplasm. Type III Secretion effectors (T3SEs), latest to be secreted T3S substrates, are destined to act at the eukaryotic host cell cytoplasm and occasionally at the nucleus, hijacking cellular processes through mimicking eukaryotic proteins. A broad range of functions is attributed to T3SEs, ranging from the manipulation of the host cell's metabolism for the benefit of the bacterium to bypassing the host's defense mechanisms. To perform this broad range of manipulations, T3SEs have evolved numerous novel folds that are compatible with some basic requirements: they should be able to easily unfold, pass through the narrow T3SS channel, and refold to an active form when on the other side. In this review, the various folds of T3SEs are presented with the emphasis placed on the functional and structural importance of α-helices and helical domains.


Assuntos
Conformação Proteica em alfa-Hélice/fisiologia , Sistemas de Secreção Tipo III/fisiologia , Animais , Proteínas de Bactérias/metabolismo , Células Eucarióticas/metabolismo , Bactérias Gram-Negativas/metabolismo , Bactérias Gram-Negativas/fisiologia , Sistemas de Secreção Tipo III/metabolismo
10.
J Bacteriol ; 202(17)2020 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-32571967

RESUMO

Salmonella enterica serovar Typhimurium uses a type three secretion system (T3SS) encoded on the Salmonella pathogenicity island 1 (SPI1) to invade intestinal epithelial cells and induce inflammatory diarrhea. The SPI1 T3SS is regulated by numerous environmental and physiological signals, integrated to either activate or repress invasion. Transcription of hilA, encoding the transcriptional activator of the SPI1 structural genes, is activated by three AraC-like regulators, HilD, HilC, and RtsA, that act in a complex feed-forward loop. Deletion of bamB, encoding a component of the ß-barrel assembly machinery, causes a dramatic repression of SPI1, but the mechanism was unknown. Here, we show that partially defective ß-barrel assembly activates the RcsCDB regulon, leading to decreased hilA transcription. This regulation is independent of RpoE activation. Though Rcs has been previously shown to repress SPI1 when disulfide bond formation is impaired, we show that activation of Rcs in a bamB background is dependent on the sensor protein RcsF, whereas disulfide bond status is sensed independently. Rcs decreases transcription of the flagellar regulon, including fliZ, the product of which indirectly activates HilD protein activity. Rcs also represses hilD, hilC, and rtsA promoters by an unknown mechanism. Both dsbA and bamB mutants have motility defects, though this is simply regulatory in a bamB background; motility is restored in the absence of Rcs. Effector secretion assays show that repression of SPI1 in a bamB background is also regulatory; if expressed, the SPI1 T3SS is functional in a bamB background. This emphasizes the sensitivity of SPI1 regulation to overall envelope homeostasis.IMPORTANCESalmonella causes worldwide foodborne illness, leading to massive disease burden and an estimated 600,000 deaths per year. Salmonella infects orally and invades intestinal epithelial cells using a type 3 secretion system that directly injects effector proteins into host cells. This first step in invasion is tightly regulated by a variety of inputs. In this work, we demonstrate that Salmonella senses the functionality of outer membrane assembly in determining regulation of invasion machinery, and we show that Salmonella uses distinct mechanisms to detect specific perturbations in envelope assembly.


Assuntos
Ilhas Genômicas/fisiologia , Salmonella typhimurium/fisiologia , Estresse Fisiológico , Sistemas de Secreção Tipo III/fisiologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , RNA Bacteriano/genética , RNA Bacteriano/metabolismo
11.
Mol Microbiol ; 112(4): 1270-1283, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31370104

RESUMO

Salmonella enterica serovar Typhimurium is a facultative intracellular pathogen that invades the intestinal epithelium. Following invasion of epithelial cells, Salmonella survives and replicates within two distinct intracellular niches. While all of the bacteria are initially taken up into a membrane bound vacuole, the Salmonella-containing vacuole or SCV, a significant proportion of them promptly escape into the cytosol. Cytosolic Salmonella replicates more rapidly compared to the vacuolar population, although the reasons for this are not well understood. SipA, a multi-function effector protein, has been shown to affect intracellular replication and is secreted by cytosolic Salmonella via the invasion-associated Type III Secretion System 1 (T3SS1). Here, we have used a multipronged microscopy approach to show that SipA does not affect bacterial replication rates per se, but rather mediates intra-cytosolic survival and/or initiation of replication following bacterial egress from the SCV. Altogether, our findings reveal an important role for SipA in the early survival of cytosolic Salmonella.


Assuntos
Proteínas de Bactérias/metabolismo , Células Epiteliais/metabolismo , Proteínas dos Microfilamentos/metabolismo , Sistemas de Secreção Tipo III/metabolismo , Adaptação Fisiológica/fisiologia , Bactérias/metabolismo , Proteínas de Bactérias/fisiologia , Citoplasma/metabolismo , Citosol/metabolismo , Citosol/fisiologia , Células Epiteliais/fisiologia , Células HeLa , Humanos , Proteínas dos Microfilamentos/fisiologia , Infecções por Salmonella/microbiologia , Salmonella enterica/metabolismo , Salmonella typhimurium/metabolismo , Sistemas de Secreção Tipo III/fisiologia , Vacúolos/fisiologia
12.
PLoS Pathog ; 14(8): e1007248, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30102745

RESUMO

Salmonella enterica serotype Typhimurium (S. Typhimurium) is one of the most frequent causes of food-borne illness in humans and usually associated with acute self-limiting gastroenteritis. However, in immunocompromised patients, the pathogen can disseminate and lead to severe systemic diseases. S. Typhimurium are facultative intracellular bacteria. For uptake and intracellular life, Salmonella translocate numerous effector proteins into host cells using two type-III secretion systems (T3SS), which are encoded within Salmonella pathogenicity islands 1 (SPI-1) and 2 (SPI-2). While SPI-1 effectors mainly promote initial invasion, SPI-2 effectors control intracellular survival and proliferation. Here, we elucidate the mode of action of Salmonella SPI-2 effector SseI, which is involved in control of systemic dissemination of S. Typhimurium. SseI deamidates a specific glutamine residue of heterotrimeric G proteins of the Gαi family, resulting in persistent activation of the G protein. Gi activation inhibits cAMP production and stimulates PI3-kinase γ by Gαi-released Gßγ subunits, resulting in activation of survival pathways by phosphorylation of Akt and mTOR. Moreover, SseI-induced deamidation leads to non-polarized activation of Gαi and, thereby, to loss of directed migration of dendritic cells.


Assuntos
Proteínas de Bactérias/fisiologia , Quimiotaxia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Salmonella typhimurium , Sistemas de Secreção Tipo III/fisiologia , Animais , Proteínas de Bactérias/genética , Sobrevivência Celular/genética , Quimiotaxia/genética , Desaminação/genética , Feminino , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/química , Células HEK293 , Células HeLa , Interações Hospedeiro-Patógeno/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Multimerização Proteica/genética , Processamento de Proteína Pós-Traducional/genética , Células RAW 264.7 , Infecções por Salmonella/metabolismo , Infecções por Salmonella/patologia , Salmonella typhimurium/genética , Salmonella typhimurium/metabolismo , Sistemas de Secreção Tipo III/genética , Sistemas de Secreção Tipo III/metabolismo
13.
Proc Natl Acad Sci U S A ; 114(23): 6098-6103, 2017 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-28533372

RESUMO

Type III protein secretion machines have evolved to deliver bacterially encoded effector proteins into eukaryotic cells. Although electron microscopy has provided a detailed view of these machines in isolation or fixed samples, little is known about their organization in live bacteria. Here we report the visualization and characterization of the Salmonella type III secretion machine in live bacteria by 2D and 3D single-molecule switching superresolution microscopy. This approach provided access to transient components of this machine, which previously could not be analyzed. We determined the subcellular distribution of individual machines, the stoichiometry of the different components of this machine in situ, and the spatial distribution of the substrates of this machine before secretion. Furthermore, by visualizing this machine in Salmonella mutants we obtained major insights into the machine's assembly. This study bridges a major resolution gap in the visualization of this nanomachine and may serve as a paradigm for the examination of other bacterially encoded molecular machines.


Assuntos
Imagem Individual de Molécula/métodos , Sistemas de Secreção Tipo III/fisiologia , Sistemas de Secreção Tipo III/ultraestrutura , Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , Sistemas de Secreção Bacterianos/metabolismo , Análise por Conglomerados , Modelos Moleculares , Transporte Proteico , Salmonella typhimurium/metabolismo , Sistemas de Secreção Tipo III/química
14.
Infect Immun ; 87(9)2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31235639

RESUMO

Salmonella enterica serovar Typhimurium (S. Typhimurium) induces inflammatory changes in the ceca of streptomycin-pretreated mice. In this mouse model of colitis, the type III secretion system 1 (T3SS-1) has been shown to induce rapid inflammatory change in the cecum at early points, 10 to 24 h after infection. Five proteins, SipA, SopA, SopB, SopD, and SopE2, have been identified as effectors involved in eliciting intestinal inflammation within this time range. In contrast, a T3SS-1-deficient strain was shown to exhibit inflammatory changes in the cecum at 72 to 120 h postinfection. However, the effectors eliciting T3SS-1-independent inflammation remain to be clarified. In this study, we focused on two T3SS-2 phenotypes, macrophage proliferation and cytotoxicity, to identify the T3SS-2 effectors involved in T3SS-1-independent inflammation. We identified a mutant strain that could not induce cytotoxicity in a macrophage-like cell line and that reduced intestinal inflammation in streptomycin-pretreated mice. We also identified five T3SS-2 effectors, SifA, SpvB, SseF, SseJ, and SteA, associated with T3SS-1-independent macrophage cytotoxicity. We then constructed a strain lacking T3SS-1 and all the five T3SS-2 effectors, termed T1S5. The S. Typhimurium T1S5 strain significantly reduced cytotoxicity in macrophages in the same manner as a mutant invA spiB strain (T1T2). Finally, the T1S5 strain elicited no inflammatory changes in the ceca of streptomycin-pretreated mice. We conclude that these five T3SS-2 effectors contribute to T3SS-1-independent inflammation.


Assuntos
Antibacterianos/farmacologia , Proteínas de Bactérias/fisiologia , Colite/microbiologia , Salmonella enterica/patogenicidade , Estreptomicina/farmacologia , Sistemas de Secreção Tipo III/fisiologia , Animais , Ceco/patologia , Colite/patologia , Modelos Animais de Doenças , Macrófagos/patologia , Camundongos , Proteínas dos Microfilamentos/fisiologia , Salmonella enterica/metabolismo
15.
Mol Microbiol ; 104(2): 234-249, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28106310

RESUMO

The bacterial flagellum contains a specialized secretion apparatus in its base that pumps certain protein subunits through the growing structure to their sites of installation beyond the membrane. A related apparatus functions in the injectisomes of gram-negative pathogens to export virulence factors into host cells. This mode of protein export is termed type-III secretion (T3S). Details of the T3S mechanism are unclear. It is energized by the proton gradient; here, a mutational approach was used to identify proton-binding groups that might function in transport. Conserved proton-binding residues in all the membrane components were tested. The results identify residues R147, R154 and D158 of FlhA as most critical. These lie in a small, well-conserved cytoplasmic domain of FlhA, located between transmembrane segments 4 and 5. Two-hybrid experiments demonstrate self-interaction of the domain, and targeted cross-linking indicates that it forms a multimeric array. A mutation that mimics protonation of the key acidic residue (D158N) was shown to trigger a global conformational change that affects the other, larger cytoplasmic domain that interacts with the export cargo. The results are discussed in the framework of a transport model based on proton-actuated movements in the cytoplasmic domains of FlhA.


Assuntos
Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Sistemas de Secreção Tipo III/metabolismo , Sequência de Aminoácidos , Flagelos/metabolismo , Mutação , Conformação Proteica , Estrutura Terciária de Proteína , Transporte Proteico/fisiologia , Relação Estrutura-Atividade , Sistemas de Secreção Tipo III/fisiologia
16.
Int J Med Microbiol ; 308(7): 872-881, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29936031

RESUMO

Microbial pathogens have developed intriguing molecular strategies to modulate and/or control host cell functions to ensure their own survival and replication. During this molecular interplay between microbes and their respective hosts especially secreted virulence factors play a major role. These factors not only include a plethora of cytotoxins but also sophisticated effector proteins targeting intracellular decision points leading to inhibition of defense responses - and/or even to cell death. To be effective, most of these secreted factors have to get across the cytoplasmic membrane and reach their targets in the cytoplasm. Apparently, pathogens use multiple mechanisms to deliver virulence factors to their cytoplasmic destination. Here, we exemplarily discuss the recently emerging scenario of parallel strategies for the intracellular deployment of toxins and effector proteins of Gram-negative pathogens with a special focus on pathogenic Escherichia coli. These pathogens employ various nanomachines such as the type III secretion system (T3SS), cell-penetrating effector proteins (CPE), and the wrapping of virulence factors in outer membrane vesicles (OMV) for protection and parallel delivery. As intracellular delivery remains a major problem in drug development, potential therapeutic applications based on these bacterial strategies will be briefly discussed.


Assuntos
Toxinas Bacterianas/metabolismo , Escherichia coli Enteropatogênica/patogenicidade , Infecções por Escherichia coli/transmissão , Interações Hospedeiro-Patógeno/fisiologia , Sistemas de Secreção Tipo III/fisiologia , Sistemas de Secreção Tipo IV/fisiologia , Sistemas de Secreção Tipo VI/fisiologia , Infecções por Escherichia coli/microbiologia , Humanos , Transporte Proteico/fisiologia , Fatores de Virulência/metabolismo
17.
Dis Aquat Organ ; 130(2): 117-129, 2018 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-30198487

RESUMO

EseN is a type III secretion system (T3SS) effector that is encoded on the Edwardsiella ictaluri chromosome and is homologous to a family of T3SS effector proteins with phosphothreonine lyase (PTL) activity, including OspF from Shigella and SpvC from Salmonella. A yeast-2-hybrid system was used to identify the major vault protein (MVP) as a specific host-cell binding partner for EseN, and the proximity ligation assay (PLA) confirmed the interaction. Similar to other pathogens, E. ictaluri invasion activates extracellular signal-regulated kinases 1 and 2 (ERK1/2) early in the infection, which are subsequently inactivated by EseN. Structurally, EseN contains a highly conserved docking motif that is required for specific binding to mitogen-activated protein kinases, such as ERK1/2, and a motif that is essential for PTL activity. Immunoblotting and immunofluorescence analyses indicate that EseN inactivates ERK1/2 by dephosphorylation in vivo in the head kidney of infected fish and ex vivo in head kidney derived macrophages. Interaction of EseN with phosphorylated ERK1/2 (pERK1/2) was also confirmed using PLA, suggesting that MVP serves as a signaling scaffold for ERK1/2 and EseN. Channel catfish Ictalurus punctatus infected with E. ictaluri strains lacking the eseN gene had reduced numbers of E. ictaluri in the tissues following infection and reduced mortality compared to fish infected with the wild-type. Our results indicate that eseN encodes a PTL domain that interacts with MVP as a possible scaffold protein and inactivates pERK1/2 to ERK1/2, resulting in increased proliferation of E. ictaluri and, ultimately, death of the host.


Assuntos
Edwardsiella ictaluri , Doenças dos Peixes , Ictaluridae , Sistemas de Secreção Tipo III , Animais , Edwardsiella ictaluri/fisiologia , Infecções por Enterobacteriaceae/veterinária , Ictaluridae/fisiologia , Ictaluridae/virologia , Liases , Fosfotreonina , Sistemas de Secreção Tipo III/fisiologia
18.
PLoS Genet ; 11(8): e1005443, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26244495

RESUMO

Flagella are assembled sequentially from the inside-out with morphogenetic checkpoints that enforce the temporal order of subunit addition. Here we show that flagellar basal bodies fail to proceed to hook assembly at high frequency in the absence of the monotopic protein SwrB of Bacillus subtilis. Genetic suppressor analysis indicates that SwrB activates the flagellar type III secretion export apparatus by the membrane protein FliP. Furthermore, mutants defective in the flagellar C-ring phenocopy the absence of SwrB for reduced hook frequency and C-ring defects may be bypassed either by SwrB overexpression or by a gain-of-function allele in the polymerization domain of FliG. We conclude that SwrB enhances the probability that the flagellar basal body adopts a conformation proficient for secretion to ensure that rod and hook subunits are not secreted in the absence of a suitable platform on which to polymerize.


Assuntos
Bacillus subtilis/metabolismo , Sistemas de Secreção Tipo III/fisiologia , Alelos , Bacillus subtilis/genética , Bacillus subtilis/ultraestrutura , Expressão Gênica , Genes Bacterianos , Óperon , Multimerização Proteica , Estrutura Quaternária de Proteína
19.
Acta Med Okayama ; 72(3): 231-239, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29926000

RESUMO

The bacterium Vibrio alginolyticus, an opportunistic pathogen in humans, has a type III secretion system (T3SS) that is responsible for its cytotoxicity toward eukaryotic cells. The effector of T3SS that is responsible for the cytotoxicity had not been identified. Here we demonstrate that VepA, a homolog of the T3SS effector in V. parahaemolyticus, is required for cytotoxicity in V. alginolyticus. VepA induces lysosomal membrane permeabilization, and it allows the leakage of only small molecules into the cytosol. Our findings revealed that VepA induces cathepsin-independent cell death in mammalian cells. The ferrous ion, one of the small molecules in the lysosome contents, appears to be involved in the cell death caused by V. alginolyticus VepA.


Assuntos
Catepsinas/fisiologia , Lisossomos/patologia , Sistemas de Secreção Tipo III/fisiologia , Vibrio alginolyticus/patogenicidade , Morte Celular , Permeabilidade da Membrana Celular , Células HeLa , Humanos
20.
J Bacteriol ; 199(1)2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27795324

RESUMO

The type III secretion system (T3SS) is a supramolecular machine used by many bacterial pathogens to translocate effector proteins directly into the eukaryotic host cell cytoplasm. Enteropathogenic Escherichia coli (EPEC) is an important cause of infantile diarrheal disease in underdeveloped countries. EPEC virulence relies on a T3SS encoded within a chromosomal pathogenicity island known as the locus of enterocyte effacement (LEE). In this work, we pursued the functional characterization of the LEE-encoded protein EscK (previously known as Orf4). We provide evidence indicating that EscK is crucial for efficient T3S and belongs to the SctK (OrgA/YscK/MxiK) protein family, whose members have been implicated in the formation of a sorting platform for secretion of T3S substrates. Bacterial fractionation studies showed that EscK localizes to the inner membrane independently of the presence of any other T3SS component. Combining yeast two-hybrid screening and pulldown assays, we identified an interaction between EscK and the C-ring/sorting platform component EscQ. Site-directed mutagenesis of conserved residues revealed amino acids that are critical for EscK function and for its interaction with EscQ. In addition, we found that T3S substrate overproduction is capable of compensating for the absence of EscK. Overall, our data suggest that EscK is a structural component of the EPEC T3SS sorting platform, playing a central role in the recruitment of T3S substrates for boosting the efficiency of the protein translocation process. IMPORTANCE: The type III secretion system (T3SS) is an essential virulence determinant for enteropathogenic Escherichia coli (EPEC) colonization of intestinal epithelial cells. Multiple EPEC effector proteins are injected via the T3SS into enterocyte cells, leading to diarrheal disease. The T3SS is encoded within a genomic pathogenicity island termed the locus of enterocyte effacement (LEE). Here we unravel the function of EscK, a previously uncharacterized LEE-encoded protein. We show that EscK is central for T3SS biogenesis and function. EscK forms a protein complex with EscQ, the main component of the cytoplasmic sorting platform, serving as a docking site for T3S substrates. Our results provide a comprehensive functional analysis of an understudied component of T3SSs.


Assuntos
Proteínas de Transporte/metabolismo , Escherichia coli Enteropatogênica/metabolismo , Proteínas de Escherichia coli/metabolismo , Regulação Bacteriana da Expressão Gênica/fisiologia , Sistemas de Secreção Tipo III/fisiologia , Proteínas de Transporte/genética , Escherichia coli Enteropatogênica/genética , Proteínas de Escherichia coli/genética , Mutação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA