Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Mol Microbiol ; 115(3): 383-394, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33217073

RESUMO

The bacterial type VI secretion system (T6SS) system is a contractile secretion apparatus that delivers proteins to neighboring bacterial or eukaryotic cells. Antibacterial effectors are mostly toxins that inhibit the growth of other species and help to dominate the niche. A broad variety of these toxins cause cell lysis of the prey cell by disrupting the cell envelope. Other effectors are delivered into the cytoplasm where they affect DNA integrity, cell division or exhaust energy resources. The modular nature of T6SS machinery allows different means of recruitment of toxic effectors to secreted inner tube and spike components that act as carriers. Toxic effectors can be translationally fused to the secreted components or interact with them through specialized structural domains. These interactions can also be assisted by dedicated chaperone proteins. Moreover, conserved sequence motifs in effector-associated domains are subject to genetic rearrangements and therefore engage in the diversification of the arsenal of toxic effectors. This review discusses the diversity of T6SS secreted toxins and presents current knowledge about their loading on the T6SS machinery.


Assuntos
Proteínas de Bactérias/fisiologia , Chaperonas Moleculares/fisiologia , Sistemas de Secreção Tipo VI/fisiologia , Antibacterianos/metabolismo , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/farmacologia , Sequência Conservada , Citoplasma/efeitos dos fármacos , Interações Microbianas , Periplasma/efeitos dos fármacos , Domínios Proteicos
2.
Proc Natl Acad Sci U S A ; 116(46): 23292-23298, 2019 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-31659021

RESUMO

The type VI secretion system (T6SS) is a lethal yet energetically costly weapon in gram-negative bacteria. Through contraction of a long sheath, the T6SS ejects a few copies of effectors accompanied by hundreds of structural carrier proteins per delivery. The few ejected effectors, however, dictate T6SS functions. It remains elusive how the T6SS ensures effector loading and avoids futile ejection. Here, by systemically mutating the active sites of 3 Vibrio cholerae effectors, TseL, VasX, and VgrG3, we show that the physical presence but not their activities is crucial for T6SS assembly. We constructed catalytic mutants of TseL and VgrG3 and truncated VasX mutants. These mutations abolished the killing of the effector-cognate immunity mutants. We determined that the VasX-mediated antimicrobial activity is solely dependent on the C-terminal colicin domain. Removal of the colicin domain abolished VasX secretion and reduced T6SS assembly, while deletion of the colicin internal loop abolished its toxicity but had little effect on secretion and assembly. The triple effector-inactive mutant maintains an active T6SS that is capable of delivering chimeric VgrG, PAAR, and TseL proteins fused with a cargo nuclease, indicating effector activities are not required for T6SS assembly or penetration into the cytosol of recipient cells. Therefore, by recruiting effectors as critical components for T6SS assembly, it represents an effective onboard checking mechanism that ensures effectors are loaded in place to prevent futile secretion. Our study also demonstrates a detoxified secretion platform by inactivating native effector activities that could translocate engineered cargo proteins via multiple routes.


Assuntos
Sistemas de Secreção Tipo VI/fisiologia , Vibrio cholerae/fisiologia , Proteínas de Bactérias/metabolismo
3.
Proc Natl Acad Sci U S A ; 116(4): 1378-1383, 2019 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-30626645

RESUMO

Acinetobacter baumannii (Ab) is a nosocomial pathogen with one of the highest rates of multidrug resistance (MDR). This is partially due to transmissible plasmids. Many Ab strains harbor a constitutively active type VI secretion system (T6SS) that is employed to kill nonkin bacteria. T6SS and plasmid conjugation both involve cell-to-cell contact. Paradoxically, successful conjugation requires the survival of the recipient, which is the target of the T6SS. Thus, an active T6SS in either the donor or the recipient poses a challenge to plasmid conjugation. Here, we show that large conjugative MDR plasmids heavily rely on their distinctive ability to repress the T6SS of their hosts to enable their own dissemination and the conjugation of other plasmids, contributing to the propagation of MDR among Acinetobacter isolates.


Assuntos
Acinetobacter baumannii/metabolismo , Acinetobacter baumannii/fisiologia , Farmacorresistência Bacteriana Múltipla/fisiologia , Sistemas de Secreção Tipo VI/fisiologia , Infecções por Acinetobacter/microbiologia , Proteínas de Bactérias/metabolismo , Plasmídeos/metabolismo
4.
Infect Immun ; 89(9): e0015121, 2021 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-34097462

RESUMO

Zebrafish (Danio rerio) are an attractive model organism for a variety of scientific studies, including host-microbe interactions. The organism is particularly useful for the study of aquatic microbes that can colonize vertebrate hosts, including Vibrio cholerae, an intestinal pathogen. V. cholerae must colonize the intestine of an exposed host for pathogenicity to occur. While numerous studies have explored various aspects of the pathogenic effects of V. cholerae on zebrafish and other model organisms, few, if any, have examined how a V. cholerae infection alters the resident intestinal microbiome and the role of the type six secretion system (T6SS) in that process. In this study, 16S rRNA gene sequencing was utilized to investigate how strains of V. cholerae both with and without the T6SS alter the aforementioned microbial profiles following an infection. V. cholerae infection induced significant changes in the zebrafish intestinal microbiome, and while not necessary for colonization, the T6SS was important for inducing mucin secretion, a marker for diarrhea. Additional salient differences to the microbiome were observed based on the presence or absence of the T6SS in the V. cholerae utilized for challenging the zebrafish hosts. We conclude that V. cholerae significantly modulates the zebrafish intestinal microbiome to enable colonization and that the T6SS is important for pathogenesis induced by the examined V. cholerae strains. Furthermore, the presence or absence of T6SS differentially and significantly affected the composition and structure of the intestinal microbiome, with an increased abundance of other Vibrio bacteria observed in the absence of V. cholerae T6SS.


Assuntos
Cólera/microbiologia , Microbioma Gastrointestinal , Interações entre Hospedeiro e Microrganismos , Interações Hospedeiro-Patógeno , Sistemas de Secreção Tipo VI/fisiologia , Vibrio cholerae/fisiologia , Animais , Modelos Animais de Doenças , Suscetibilidade a Doenças , Metagenômica/métodos , RNA Ribossômico 16S , Peixe-Zebra
5.
Infect Immun ; 89(7): e0057920, 2021 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-33875476

RESUMO

Francisella tularensis causes the deadly zoonotic disease tularemia in humans and is able to infect a broad range of organisms including arthropods, which are thought to play a major role in Francisella transmission. However, while mammalian in vitro and in vivo infection models are widely used to investigate Francisella pathogenicity, a detailed characterization of the major Francisella virulence factor, a noncanonical type VI secretion system (T6SS), in an arthropod in vivo infection model is missing. Here, we use Galleria mellonella larvae to analyze the role of the Francisella T6SS and its corresponding effectors in F. tularensis subsp. novicida virulence. We report that G. mellonella larvae killing depends on the functional T6SS and infectious dose. In contrast to other mammalian in vivo infection models, even one of the T6SS effectors PdpC, PdpD, or OpiA is sufficient to kill G. mellonella larvae, while sheath recycling by ClpB is dispensable. We further demonstrate that treatment by polyethylene glycol (PEG) activates Francisella T6SS in liquid culture and that this is independent of the response regulator PmrA. PEG-activated IglC secretion is dependent on T6SS structural component PdpB but independent of putative effectors PdpC, PdpD, AnmK, OpiB1, OpiB2, and OpiB3. The results of larvae infection and secretion assay suggest that AnmK, a putative T6SS component with unknown function, interferes with OpiA-mediated toxicity but not with general T6SS activity. We establish that the easy-to-use G. mellonella larvae infection model provides new insights into the function of T6SS and pathogenesis of Francisella.


Assuntos
Proteínas de Bactérias/genética , Francisella tularensis/fisiologia , Larva/microbiologia , Mariposas/microbiologia , Sistemas de Secreção Tipo VI/fisiologia , Animais , Proteínas de Bactérias/metabolismo , Modelos Animais de Doenças , Francisella tularensis/efeitos dos fármacos , Polietilenoglicóis/farmacologia , Tularemia , Sistemas de Secreção Tipo VI/efeitos dos fármacos , Virulência/genética , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
6.
Mol Microbiol ; 114(2): 308-321, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32279364

RESUMO

The human pathogen Pseudomonas aeruginosa harbors three paralogous zinc proteases annotated as AmpD, AmpDh2, and AmpDh3, which turn over the cell wall and cell wall-derived muropeptides. AmpD is cytoplasmic and plays a role in the recycling of cell wall muropeptides, with a link to antibiotic resistance. AmpDh2 is a periplasmic soluble enzyme with the former anchored to the inner leaflet of the outer membrane. We document, herein, that the type VI secretion system locus II (H2-T6SS) of P. aeruginosa delivers AmpDh3 (but not AmpD or AmpDh2) to the periplasm of a prey bacterium upon contact. AmpDh3 hydrolyzes the cell wall peptidoglycan of the prey bacterium, which leads to its killing, thereby providing a growth advantage for P. aeruginosa in bacterial competition. We also document that the periplasmic protein PA0808, heretofore of unknown function, affords self-protection from lysis by AmpDh3. Cognates of the AmpDh3-PA0808 pair are widely distributed across Gram-negative bacteria. Taken together, these findings underscore the importance of their function as an evolutionary advantage and that of the H2-T6SS as the means for the manifestation of the effect.


Assuntos
Pseudomonas aeruginosa/metabolismo , Sistemas de Secreção Tipo VI/metabolismo , Sistemas de Secreção Tipo VI/fisiologia , Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , Parede Celular/metabolismo , Bactérias Gram-Negativas/metabolismo , Metaloproteases/metabolismo , N-Acetil-Muramil-L-Alanina Amidase/metabolismo , Peptidoglicano/metabolismo , Periplasma/metabolismo , Fatores de Virulência/metabolismo , beta-Lactamases/metabolismo
7.
PLoS Pathog ; 15(7): e1007885, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31323074

RESUMO

Colonies of the opportunistic pathogen Proteus mirabilis can distinguish self from non-self: in swarming colonies of two different strains, one strain excludes the other from the expanding colony edge. Predominant models characterize bacterial kin discrimination as immediate antagonism towards non-kin cells, typically through delivery of toxin effector molecules from one cell into its neighbor. Upon effector delivery, receiving cells must either neutralize it by presenting a cognate anti-toxin as would a clonal sibling, or suffer cell death or irreversible growth inhibition as would a non-kin cell. Here we expand this paradigm to explain the non-lethal Ids self-recognition system, which stops access to a social behavior in P. mirabilis by selectively and transiently inducing non-self cells into a growth-arrested lifestyle incompatible with cooperative swarming. This state is characterized by reduced expression of genes associated with protein synthesis, virulence, and motility, and also causes non-self cells to tolerate previously lethal concentrations of antibiotics. We show that temporary activation of the stringent response is necessary for entry into this state, ultimately resulting in the iterative exclusion of non-self cells as a swarm colony migrates outwards. These data clarify the intricate connection between non-lethal recognition and the lifecycle of P. mirabilis swarm colonies.


Assuntos
Interações Microbianas/fisiologia , Proteus mirabilis/fisiologia , Proteus mirabilis/patogenicidade , Animais , Farmacorresistência Bacteriana/genética , Farmacorresistência Bacteriana/fisiologia , Genes Bacterianos , Humanos , Interações Microbianas/genética , Modelos Biológicos , Infecções por Proteus/microbiologia , Proteus mirabilis/genética , Transcrição Gênica , Sistemas de Secreção Tipo VI/genética , Sistemas de Secreção Tipo VI/fisiologia , Infecções Urinárias/microbiologia , Virulência/genética , Virulência/fisiologia
8.
Cell Microbiol ; 22(3): e13153, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31872954

RESUMO

Microbiota niches have space and/or nutrient restrictions, which has led to the coevolution of cooperation, specialisation, and competition within the population. Different animal and environmental niches contain defined resident microbiota that tend to be stable over time and offer protection against undesired intruders. Yet fluxes can occur, which alter the composition of a bacterial population. In humans, the microbiota are now considered a key contributor to maintenance of health and homeostasis, and its alteration leads to dysbiosis. The bacterial type VI secretion system (T6SS) transports proteins into the environment, directly into host cells or can function as an antibacterial weapon by killing surrounding competitors. Upon contact with neighbouring cells, the T6SS fires, delivering a payload of effector proteins. In the absence of an immunity protein, this results in growth inhibition or death of prey leading to a competitive advantage for the attacker. It is becoming apparent that the T6SS has a role in modulating and shaping the microbiota at multiple levels, which is the focus of this review. Discussed here is the T6SS, its role in competition, key examples of its effect upon the microbiota, and future avenues of research.


Assuntos
Microbiota , Sistemas de Secreção Tipo VI/fisiologia , Animais , Antibiose , Proteínas de Bactérias/fisiologia , Homeostase , Interações entre Hospedeiro e Microrganismos , Humanos
9.
Arch Microbiol ; 203(1): 45-58, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32929595

RESUMO

Shigella sonnei is the emerging pathogen globally, as it is the second common infectious species of shigellosis (bloody diarrhoea) in low- and middle-income countries (LMICs) and the leading one in developed world. The multifactorial processes and novel mechanisms have been identified in S. sonnei, that are collectively playing apart a substantial role in increasing its prevalence, while replacing the S. flexneri and other Gram-negative gut pathogens niche occupancy. Recently, studies suggest that due to improvement in sanitation S. sonnei has reduced cross-immunization from Plesiomonas shigelliodes (having same O-antigen as S. sonnei) and also found to outcompete the two major species of Enterobacteriaceae family (Shigella flexneri and Escherichia coli), due to encoding of type VI secretion system (T6SS). This review aimed to highlight S. sonnei as an emerging pathogen in the light of recent research with pondering aspects on its epidemiology, transmission, and pathogenic mechanisms. Additionally, this paper aimed to review S. sonnei disease pattern and related complications, symptoms, and laboratory diagnostic techniques. Furthermore, the available treatment reigns and antibiotic-resistance patterns of S. sonnei are also discussed, as the ciprofloxacin and fluoroquinolone-resistant S. sonnei has already intensified the global spread and burden of antimicrobial resistance. In last, prevention and controlling strategies are briefed to limit and tackle S. sonnei and possible future areas are also explored that needed more research to unravel the hidden mysteries surrounding S. sonnei.


Assuntos
Farmacorresistência Bacteriana , Disenteria Bacilar/tratamento farmacológico , Disenteria Bacilar/microbiologia , Shigella sonnei/efeitos dos fármacos , Shigella sonnei/patogenicidade , Antibacterianos/farmacologia , Ciprofloxacina/farmacologia , Disenteria Bacilar/diagnóstico , Disenteria Bacilar/patologia , Escherichia coli/efeitos dos fármacos , Fluoroquinolonas/farmacologia , Humanos , Shigella flexneri/efeitos dos fármacos , Sistemas de Secreção Tipo VI/fisiologia
10.
Proc Natl Acad Sci U S A ; 115(49): 12519-12524, 2018 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-30455305

RESUMO

The type VI secretion system (T6SS) is a supramolecular complex involved in the delivery of potent toxins during bacterial competition. Pseudomonas aeruginosa possesses three T6SS gene clusters and several hcp and vgrG gene islands, the latter encoding the spike at the T6SS tip. The vgrG1b cluster encompasses seven genes whose organization and sequences are highly conserved in P. aeruginosa genomes, except for two genes that we called tse7 and tsi7 We show that Tse7 is a Tox-GHH2 domain nuclease which is distinct from other T6SS nucleases identified thus far. Expression of this toxin induces the SOS response, causes growth arrest and ultimately results in DNA degradation. The cytotoxic domain of Tse7 lies at its C terminus, while the N terminus is a predicted PAAR domain. We find that Tse7 sits on the tip of the VgrG1b spike and that specific residues at the PAAR-VgrG1b interface are essential for VgrG1b-dependent delivery of Tse7 into bacterial prey. We also show that the delivery of Tse7 is dependent on the H1-T6SS cluster, and injection of the nuclease into bacterial competitors is deployed for interbacterial competition. Tsi7, the cognate immunity protein, protects the producer from the deleterious effect of Tse7 through a direct protein-protein interaction so specific that toxin/immunity pairs are effective only if they originate from the same P. aeruginosa isolate. Overall, our study highlights the diversity of T6SS effectors, the exquisite fitting of toxins on the tip of the T6SS, and the specificity in Tsi7-dependent protection, suggesting a role in interstrain competition.


Assuntos
Proteínas de Bactérias/metabolismo , Dano ao DNA/fisiologia , Regulação Bacteriana da Expressão Gênica/fisiologia , Pseudomonas aeruginosa/metabolismo , Sistemas de Secreção Tipo VI/fisiologia , Proteínas de Bactérias/genética , Modelos Moleculares , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Pseudomonas aeruginosa/genética
11.
Proc Natl Acad Sci U S A ; 115(16): E3779-E3787, 2018 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-29610339

RESUMO

Host-associated microbiota help defend against bacterial pathogens; however, the mechanisms by which pathogens overcome this defense remain largely unknown. We developed a zebrafish model and used live imaging to directly study how the human pathogen Vibrio cholerae invades the intestine. The gut microbiota of fish monocolonized by symbiotic strain Aeromonas veronii was displaced by V. cholerae expressing its type VI secretion system (T6SS), a syringe-like apparatus that deploys effector proteins into target cells. Surprisingly, displacement was independent of T6SS-mediated killing of A. veronii, driven instead by T6SS-induced enhancement of zebrafish intestinal movements that led to expulsion of the resident microbiota by the host. Deleting an actin cross-linking domain from the T6SS apparatus returned intestinal motility to normal and thwarted expulsion, without weakening V. cholerae's ability to kill A. veronii in vitro. Our finding that bacteria can manipulate host physiology to influence intermicrobial competition has implications for both pathogenesis and microbiome engineering.


Assuntos
Antibiose/fisiologia , Microbioma Gastrointestinal , Sistemas de Secreção Tipo VI/fisiologia , Vibrio cholerae/fisiologia , Peixe-Zebra/microbiologia , Actinas/fisiologia , Aeromonas veronii , Animais , Proteínas de Bactérias/fisiologia , Motilidade Gastrointestinal , Vida Livre de Germes , Interações Hospedeiro-Patógeno , Simbiose , Vibrio cholerae/patogenicidade
12.
J Fish Dis ; 44(11): 1669-1679, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34431107

RESUMO

Pseudomonas plecoglossicida, the causative agent of visceral granulomas in the large yellow croaker (Larimichthys crocea) in China, encodes three sets of type Ⅵ secretion systems (T6SS1-3). The purpose of this study was to characterize the different roles of T6SSs involved in infection. In-frame deletion of T6SSs was constructed, which resulted in 8 mutants. Competition against E. coli DH5α, virulence against the croaker and in vivo survival ability of the mutants were tested. The expression and secretion of Hcp by P. plecoglossicida NB2011 were investigated. The results showed T6SS2 mutant failed to inhibit the growth of E. coli, which is an indication of T6SS2 acting against environmental bacteria. The LD50 value of T6SS1 mutant strongly increased; T6SS2 and T6SS3 mutants were similar to that of the wild type; and the virulence of double deletion or triple deletion mutant was drastically alleviated, indicating that T6SS1 being one of the major virulence factors, and T6SS2 and T6SS3 directly or indirectly being involved in the pathogenicity. T6SS1 mutant disappeared in the fish spleen in 3 days, while other strains kept increasing, indicating the T6SS1 stimulation bacteria replication in vivo. Hcp1 secreted at 12-28°C and Hcp2 secreted at 12-35°C, while Hcp3 secretion not detected in vitro. This study has thrown some insights on the understanding of pathogenicity mechanisms of this pathogen.


Assuntos
Doenças dos Peixes/microbiologia , Perciformes/microbiologia , Pseudomonas/patogenicidade , Sistemas de Secreção Tipo VI , Virulência , Animais , Pseudomonas/genética , Infecções por Pseudomonas/veterinária , Sistemas de Secreção Tipo VI/fisiologia , Fatores de Virulência
13.
Proc Natl Acad Sci U S A ; 114(35): 9427-9432, 2017 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-28808000

RESUMO

The type 6 secretion system (T6SS) is used by many Gram-negative bacterial species to deliver toxic effector proteins into nearby bacteria prey cells to kill or inhibit their growth. VgrG proteins are core conserved secretion substrates of the T6SS and one subset of T6SS effectors consists of VgrG proteins with C-terminal extension domains carrying various enzymatic activities. In Vibrio cholerae, VgrG3 has a hydrolase extension domain and degrades peptidoglycan in the periplasm of target bacteria. In this study, we replaced this domain with a nuclease domain from Salmonella enterica subsp. arizonae This modified V. cholerae strain was able to kill its parent using its T6SS. This result also demonstrated that V. cholerae T6SS is capable of delivering effectors that could attack substrates found either in the periplasm or cytosol of target bacteria. Additionally, we found that effectors VgrG3 and TseL, despite lacking a classical Sec or TAT secretion signal, were able to reach the periplasm when expressed in the bacterial cytosol. This effector trafficking likely represents an evolutionary strategy for T6SS effectors to reach their intended substrates regardless of which subcellular compartment in the target cell they happen to be delivered to by the T6SS apparatus.


Assuntos
Proteínas de Bactérias/metabolismo , Transporte Proteico/fisiologia , Sistemas de Secreção Tipo VI/fisiologia , Vibrio cholerae/fisiologia , Proteínas de Bactérias/genética , Regulação Bacteriana da Expressão Gênica
14.
J Bacteriol ; 202(1)2019 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-31636107

RESUMO

Antimicrobial treatment can induce many bacterial pathogens to enter a cell wall-deficient state that contributes to persistent infections. The effect of this physiological state on the assembly of transenvelope-anchored organelles is not well understood. The type VI secretion system (T6SS) is a widespread molecular weapon for interspecies interactions and virulence, comprising a long double tubular structure and a transenvelope/baseplate complex. Here, we report that cell wall-deficient spheroplasts assembled highly flexible and elastic T6SS structures forming U, O, or S shapes. Upon contacting the inner membrane, the T6SS tubes did not contract but rather continued to grow along the membrane. Such deformation likely results from continual addition of sheath/tube subunits at the distal end. Induction of TagA repressed curved sheath formation. Curved sheaths could also contract and deliver T6SS substrates and were readily disassembled by the ClpV ATPase after contraction. Our data highlight the dramatic effect of cell wall deficiency on the shape of the T6SS structures and reveal the elastic nature of this double tubular contractile injection nanomachine.IMPORTANCE The cell wall is a physical scaffold that all transenvelope complexes have to cross for assembly. However, the cell wall-deficient state has been described as a common condition found in both Gram-negative and Gram-positive pathogens during persistent infections. Loss of cell wall is known to have pleiotropic physiological effects, but how membrane-anchored large cellular organelles adapt to this unique state is less completely understood. Our study examined the assembly of the T6SS in cell wall-deficient spheroplast cells. We report the elastic nature of contractile T6SS tubules under such conditions, providing key insights for understanding how large intracellular structures such as the T6SS accommodate the multifaceted changes in cell wall-deficient cells.


Assuntos
Sistemas de Secreção Tipo VI/fisiologia , Proteínas de Bactérias/fisiologia , Parede Celular/química , Parede Celular/fisiologia , Elasticidade , Lipoproteínas/fisiologia , Esferoplastos/fisiologia , Sistemas de Secreção Tipo VI/química
15.
Mol Microbiol ; 108(1): 6-15, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29405518

RESUMO

Contractile tail bacteriophages, or myobacteriophages, use a sophisticated biomolecular structure to inject their genome into the bacterial host cell. This structure consists of a contractile sheath enveloping a rigid tube that is sharpened by a spike-shaped protein complex at its tip. The spike complex forms the centerpiece of a baseplate complex that terminates the sheath and the tube. The baseplate anchors the tail to the target cell membrane with the help of fibrous proteins emanating from it and triggers contraction of the sheath. The contracting sheath drives the tube with its spiky tip through the target cell membrane. Subsequently, the bacteriophage genome is injected through the tube. The structural transformation of the bacteriophage T4 baseplate upon binding to the host cell has been recently described in near-atomic detail. In this review we discuss structural elements and features of this mechanism that are likely to be conserved in all contractile injection systems (systems evolutionary and structurally related to contractile bacteriophage tails). These include the type VI secretion system (T6SS), which is used by bacteria to transfer effectors into other bacteria and into eukaryotic cells, and tailocins, a large family of contractile bacteriophage tail-like compounds that includes the P. aeruginosa R-type pyocins.


Assuntos
Bacteriófago T4/química , Bacteriófago T4/fisiologia , Proteínas da Cauda Viral/química , Proteínas da Cauda Viral/fisiologia , Bacteriófago T4/genética , Evolução Biológica , Membrana Celular/química , Membrana Celular/metabolismo , Genoma Viral , Bactérias Gram-Negativas/química , Bactérias Gram-Negativas/genética , Bactérias Gram-Negativas/fisiologia , Piocinas/química , Piocinas/metabolismo , Sistemas de Secreção Tipo VI/química , Sistemas de Secreção Tipo VI/genética , Sistemas de Secreção Tipo VI/fisiologia , Proteínas da Cauda Viral/genética , Difração de Raios X
16.
Fish Shellfish Immunol ; 92: 851-860, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31129187

RESUMO

Innate immunity is regulated by phagocytic cells and is critical for host control of bacterial infection. In many bacteria, the type VI secretion system (T6SS) can affect bacterial virulence in certain environments, but little is known about the mechanisms underlying T6SS regulation of innate immune responses during infection in vivo. Here, we developed an infection model by microinjecting bacteria into the tail vein muscle of 3-day-post-fertilized zebrafish larvae, and found that both macrophages and neutrophils are essential for bacterial clearance. Further study revealed that EvpP plays a critical role in promoting the pathogenesis of Edwardsiella piscicida (E. piscicida) via inhibiting the phosphorylation of Jnk signaling to reduce the expression of chemokine (CXC motif) ligand 8 (cxcl8a), matrix metallopeptidase 13 (mmp13) and interleukin-1ß (IL-1ß) in vivo. Subsequently, by utilizing Tg (mpo:eGFP+/+) zebrafish larvae for E. piscicida infection, we found that the EvpP-inhibited Jnk-caspy (caspase-1 homolog) inflammasome signaling axis significantly suppressed the recruitment of neutrophils to infection sites, and the caspy- or IL-1ß-morpholino (MO) knockdown larvae were more susceptible to infection and failed to restrict bacterial colonization in vivo. taken together, this interaction improves our understanding about the complex and contextual role of a bacterial T6SS effector in modulating the action of neutrophils during infection, and offers new insights into the warfare between bacterial weapons and host immunological surveillance.


Assuntos
Proteínas de Bactérias/fisiologia , Imunidade Inata , Inflamassomos/imunologia , Macrófagos/metabolismo , Neutrófilos/metabolismo , Peixe-Zebra/imunologia , Animais , Edwardsiella/fisiologia , Infecções por Enterobacteriaceae/imunologia , Infecções por Enterobacteriaceae/veterinária , Doenças dos Peixes/imunologia , Sistemas de Secreção Tipo VI/fisiologia
17.
Proc Natl Acad Sci U S A ; 113(13): 3627-32, 2016 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-26951680

RESUMO

Type VI secretion systems (T6SSs) are multiprotein complexes best studied in Gram-negative pathogens where they have been shown to inhibit or kill prokaryotic or eukaryotic cells and are often important for virulence. We recently showed that T6SS loci are also widespread in symbiotic human gut bacteria of the order Bacteroidales, and that these T6SS loci segregate into three distinct genetic architectures (GA). GA1 and GA2 loci are present on conserved integrative conjugative elements (ICE) and are transferred and shared among diverse human gut Bacteroidales species. GA3 loci are not contained on conserved ICE and are confined to Bacteroides fragilis Unlike GA1 and GA2 T6SS loci, most GA3 loci do not encode identifiable effector and immunity proteins. Here, we studied GA3 T6SSs and show that they antagonize most human gut Bacteroidales strains analyzed, except for B. fragilis strains with the same T6SS locus. A combination of mutation analyses,trans-protection analyses, and in vitro competition assays, allowed us to identify novel effector and immunity proteins of GA3 loci. These proteins are not orthologous to known proteins, do not contain identified motifs, and most have numerous predicted transmembrane domains. Because the genes encoding effector and immunity proteins are contained in two variable regions of GA3 loci, GA3 T6SSs of the species B. fragilis are likely the source of numerous novel effector and immunity proteins. Importantly, we show that the GA3 T6SS of strain 638R is functional in the mammalian gut and provides a competitive advantage to this organism.


Assuntos
Bacteroides fragilis/fisiologia , Bacteroidetes/fisiologia , Microbioma Gastrointestinal/fisiologia , Sistemas de Secreção Tipo VI/fisiologia , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/fisiologia , Bacteroides fragilis/genética , Bacteroides fragilis/imunologia , Bacteroidetes/genética , Bacteroidetes/imunologia , Microbioma Gastrointestinal/imunologia , Genes Bacterianos , Humanos , Camundongos , Mutação , Sistemas de Secreção Tipo VI/genética , Sistemas de Secreção Tipo VI/imunologia
18.
Proc Natl Acad Sci U S A ; 113(13): 3639-44, 2016 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-26957597

RESUMO

The human gut microbiome is a dynamic and densely populated microbial community that can provide important benefits to its host. Cooperation and competition for nutrients among its constituents only partially explain community composition and interpersonal variation. Notably, certain human-associated Bacteroidetes--one of two major phyla in the gut--also encode machinery for contact-dependent interbacterial antagonism, but its impact within gut microbial communities remains unknown. Here we report that prominent human gut symbionts persist in the gut through continuous attack on their immediate neighbors. Our analysis of just one of the hundreds of species in these communities reveals 12 candidate antibacterial effector loci that can exist in 32 combinations. Through the use of secretome studies, in vitro bacterial interaction assays and multiple mouse models, we uncover strain-specific effector/immunity repertoires that can predict interbacterial interactions in vitro and in vivo, and find that some of these strains avoid contact-dependent killing by accumulating immunity genes to effectors that they do not encode. Effector transmission rates in live animals can exceed 1 billion events per minute per gram of colonic contents, and multiphylum communities of human gut commensals can partially protect sensitive strains from these attacks. Together, these results suggest that gut microbes can determine their interactions through direct contact. An understanding of the strategies human gut symbionts have evolved to target other members of this community may provide new approaches for microbiome manipulation.


Assuntos
Microbioma Gastrointestinal/fisiologia , Animais , Bacteroides fragilis/genética , Bacteroides fragilis/imunologia , Bacteroides fragilis/fisiologia , Feminino , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/imunologia , Genoma Bacteriano , Vida Livre de Germes , Humanos , Masculino , Camundongos , Modelos Animais , Filogenia , Simbiose/genética , Simbiose/imunologia , Simbiose/fisiologia , Sistemas de Secreção Tipo VI/genética , Sistemas de Secreção Tipo VI/imunologia , Sistemas de Secreção Tipo VI/fisiologia
19.
J Bacteriol ; 200(14)2018 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-29555703

RESUMO

The bacterium Proteus mirabilis can communicate identity through the secretion of the self-identity protein IdsD via the type VI secretion (T6S) system. IdsD secretion is essential for self-versus-nonself recognition behaviors in these populations. Here we provide an answer to the unresolved question of how the activity of a T6S substrate, such as IdsD, is regulated before secretion. We demonstrate that IdsD is found in clusters that form independently of the T6S machinery and activity. We show that the IdsC protein, which is a member of the proposed DUF4123 chaperone family, is essential for the maintenance of these clusters and of the IdsD protein itself. We provide evidence that amino acid disruptions in IdsC are sufficient to disrupt IdsD secretion but not IdsD localization into subcellular clusters, strongly supporting the notion that IdsC functions in at least two different ways: maintaining IdsD levels and secreting IdsD. We propose that IdsC, and likely other DUF4123-containing proteins, functions to regulate T6S substrates in the donor cell both by maintaining protein levels and by mediating secretion at the T6S machinery.IMPORTANCE Understanding the subcellular dynamics of self-identity proteins is crucial for developing models of self-versus-nonself recognition. We directly addressed how a bacterium restricts self-identity information before cell-cell exchange. We resolved two conflicting models for type VI secretion (T6S) substrate regulation by focusing on the self-identity protein IdsD. One model is that a cognate immunity protein binds the substrate, inhibiting activity before transport. Another model proposes that DUF4123 proteins act as chaperones in the donor cell, but no detailed molecular mechanism was previously known. We resolve this discrepancy and propose a model wherein a chaperone couples IdsD sequestration with its localization. Such a molecular mechanism restricts the communication of identity, and possibly other T6S substrates, in producing cells.


Assuntos
Proteínas de Bactérias/metabolismo , Escherichia coli/metabolismo , Proteus mirabilis/metabolismo , Percepção de Quorum/fisiologia , Sistemas de Secreção Tipo VI/fisiologia , Proteínas de Bactérias/genética , Regulação Bacteriana da Expressão Gênica/fisiologia , Chaperonas Moleculares
20.
Environ Microbiol ; 20(1): 1-15, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29027348

RESUMO

The type VI secretion system (T6SS) is a bacterial nanomachine used to inject effectors into prokaryotic or eukaryotic cells and is thus involved in both host manipulation and interbacterial competition. The T6SS is widespread among Gram-negative bacteria, mostly within the Proteobacterium Phylum. This secretion system is commonly found in commensal and pathogenic plant-associated bacteria. Phylogenetic analysis of phytobacterial T6SS clusters shows that they are distributed in the five main clades previously described (group 1-5). The even distribution of the system among commensal and pathogenic phytobacteria suggests that the T6SS provides fitness and colonization advantages in planta and that the role of the T6SS is not restricted to virulence. This manuscript reviews the phylogeny and biological roles of the T6SS in plant-associated bacteria, highlighting a remarkable diversity both in terms of mechanism and function.


Assuntos
Plantas/microbiologia , Proteobactérias/metabolismo , Proteobactérias/patogenicidade , Sistemas de Secreção Tipo VI/fisiologia , Proteínas de Bactérias/genética , Filogenia , Doenças das Plantas/microbiologia , Proteobactérias/genética , Sistemas de Secreção Tipo VI/genética , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA